Molecular Biology Depiction of the P22 bacteriophage virion as solved by electron cryomicroscopy. This structure reveals the major components of the infection machinery, as well as the manner in which DNA is spooled coaxially within the icosahedral capsid. The contour level is set so that the cross section of individual dsDNA strands can be seen with only the outer, most ordered shell of DNA visible. A dodecameric structure at the center of the infection machinery functions as a pressure sensor, which during the packaging of DNA during virus maturation, sends a termination signal to the packaging machinery once the capsid has been fully packaged. Reconstruction and graphics by graduate student Gabriel Lander. Work done in the laboratory of John E. Johnson, Ph.D., in collaboration with Bridget Carragher and Clint Potter, Department of Cell Biology, National Research Resource for Automated Microscopy. Kurt Wüthrich, Ph.D. Cecil H. and Ida M. Green Professor of Structural Biology MOLECULAR BIOLOGY 2006 MOLECULAR BIOLOGY H. Jane Dyson, Ph.D. Professor S TA F F John H. Elder, Ph.D.*** Professor DEPAR TMENT OF Peter E. Wright, Ph.D.* Professor and Chairman Cecil H. and Ida M. Green Investigator in Medical Research Ruben Abagyan, Ph.D. Professor Martha J. Fedor, Ph.D.* Associate Professor James Arthur Fee, Ph.D. Professor of Research Elizabeth D. Getzoff, Ph.D.**** Professor Carlos F. Barbas III, Ph.D.***** Professor Janet and W. Keith Kellogg II Chair David B. Goodin, Ph.D. Associate Professor Rajesh Belani, Ph.D. Adjunct Assistant Professor Joel M. Gottesfeld, Ph.D. Professor Ola Blixt, Ph.D. Assistant Professor of Molecular Biology Michael N. Boddy, Ph.D. Assistant Professor Charles L. Brooks III, Ph.D. Professor David A. Case, Ph.D. Professor Geoffrey Chang, Ph.D.* Associate Professor Jerold Chun, M.D., Ph.D.*** Professor Luis De Lecea, Ph.D.** Associate Professor Stanford University Palo Alto, California Aymeric Pierre De Parseval, Ph.D. Assistant Professor of Molecular Biology Lluis Ribas De Pouplana, Ph.D. Adjunct Assistant Professor Barcelona Science Park Barcelona, Spain Ashok Deniz, Ph.D. Assistant Professor David S. Goodsell Jr., Ph.D. Associate Professor Jennifer Harris, Ph.D. Assistant Professor of Biochemistry Christian A. Hassig, Ph.D. Adjunct Assistant Professor Kalypsis, Inc. San Diego, California Peter B. Hedlund, M.D., Ph.D. Assistant Professor of Molecular Biology Mirko Hennig, Ph.D.** Assistant Professor Medical University of South Carolina Charleston, South Carolina John E. Johnson, Ph.D. Professor Gerald F. Joyce, M.D., Ph.D.***** Professor Dean, Faculty Ehud Keinan, Ph.D. Adjunct Professor Richard A. Lerner, M.D., Ph.D.***** President, Scripps Research Lita Annenberg Hazen Professor of Immunochemistry Cecil H. and Ida M. Green Chair in Chemistry THE SCRIPPS RESEARCH INSTITUTE 153 Scott Lesley, Ph.D. Assistant Professor of Biochemistry Vaughn V. Smider, Ph.D. Assistant Professor of Molecular Biology Tianwei Lin, Ph.D. Associate Professor Robyn L. Stanfield, Ph.D. Assistant Professor Clare McGowan, Ph.D. † Associate Professor James Steven, Ph.D. Assistant Professor Duncan E. McRee, Ph.D. Adjunct Associate Professor ActiveSight San Diego, California Raymond C. Stevens, Ph.D. ††† Professor David P. Millar, Ph.D. Associate Professor Louis Noodleman, Ph.D. Associate Professor Arthur J. Olson, Ph.D. Professor James C. Paulson, Professor Ph.D. †† Vijay Reddy, Ph.D. Assistant Professor Steven I. Reed, Professor Ph.D. † Paul Russell, Ph.D. Professor Michel Sanner, Ph.D. Associate Professor Harold Scheraga, Ph.D. Adjunct Professor George W. and Grace L. Todd Professor of Chemistry, Emeritus Cornell University Ithaca, New York Paul R. Schimmel, Ph.D.***** Ernest and Jean Hahn Professor of Molecular Biology and Chemistry Anette Schneemann, Ph.D. Associate Professor Charles D. Stout, Ph.D. Associate Professor Peiqing Sun, Ph.D. Associate Professor J. Gregor Sutcliffe, Ph.D. Professor John A. Tainer, Ph.D.* Professor Fujie Tanaka, Ph.D. Assistant Professor Elizabeth Anne Thomas, Ph.D. Assistant Professor James R. Williamson, Ph.D.***** Professor Associate Dean, Kellogg School of Science and Technology Ian A. Wilson, D.Phil.* Professor Curt Wittenberg, Ph.D. † Professor Kurt Wüthrich, Ph.D. Cecil H. and Ida M. Green Professor of Structural Biology Xiang-Lei Yang, Ph.D. Assistant Professor of Molecular Biology Subhash C. Sinha, Ph.D.* Associate Professor Todd O. Yeates, Ph.D. Adjunct Professor University of California Los Angeles, California Gary Siuzdak, Ph.D. Adjunct Associate Professor Qinghai Zhang, Ph.D. Assistant Professor 154 MOLECULAR BIOLOGY 2006 SERVICE FACILITIES Ryan Burnett, Ph.D. John Chung, Ph.D. Manager, Nuclear Magnetic Resonance Facilities Zhongguo Chen, Ph.D. Gerard Kroon Assistant Manager, Nuclear Magnetic Resonance Facilities Michael E. Pique Director, Computer Graphics Development Nahid Razi, Ph.D. Assistant Core Manager, Consortium for Functional Glycomics Brian Collins, Ph.D.** Momenta Pharmaceuticals Boston, Massachusetts Adrienne Elizabeth Dubin, Ph.D. Maria Alejandra GamezAbascal, Ph.D. Reto Horst, Ph.D. Julio Kovacs, Ph.D. THE SCRIPPS RESEARCH INSTITUTE Beatriz Gonzalez Alonso, Ph.D. Ronald M. Brudler, Ph.D.†††† Lintao Bu, Ph.D. David Alvarez-Carbonell, Ph.D.** Memorial Sloan-Kettering Cancer Center New York, New York Tommy Bui, Ph.D. Rosa Maria Cardoso, Ph.D. Justin E. Carlson, Ph.D. Juliano Alves, Ph.D. Andrew Barry Carmel, Ph.D. Yu An, Ph.D. Qing Chai, M.D., Ph.D. Brigitte Anliker, Ph.D.** Institut für Biochemie und Molekularbiologie II Heinrich-Heine-Universitaet Düsseldorf, Germany Eli Chapman, Ph.D. Amarnath Chatterjee, Ph.D. Andrew James Annalora, Ph.D. Anju Chatterji, Ph.D.** Bayer Healthcare, Diagnostics Division Berkeley, California Roger Armen, Ph.D. Susana Chaves, Ph.D. Richard R. Rivera, Ph.D. Joseph W. Arndt, Ph.D. Kenji Sugase, Ph.D. Karunesh Arora, Ph.D. Mabelle Ashe, Ph.D. S TA F F S C I E N T I S T S Koji Tamura, Ph.D.** Tokyo University of Science Chiba, Japan Anton Vladislavovich Cheltsov, Ph.D.** Burnham Institute La Jolla, California Svitlana Berezhna, Ph.D. Xiaoqin Ye, M.D., Ph.D. Jamie Mitchell Bacher, Ph.D. Brian M. Lee, Ph.D.** Southern Illinois University Carbondale, Illinois Dirk M. Zajonc, Ph.D.** La Jolla Institute for Allergy & Immunology San Diego, California Sung-Hun Bae, Ph.D. Peter Sobieszcsuk, Ph.D. Core Manager, Consortium for Functional Glycomics S E N I O R S TA F F S C I E N T I S T Wayne A. Fenton, Ph.D. Ying Chuan Lin, Ph.D. Mikhail Popkov, Ph.D.** Amunix, Inc. San Jose, California Maria Martinez-Yamout, Ph.D. Garrett M. Morris, Ph.D. Chiaki Nishimura, Ph.D. Jeffrey Speir, Ph.D. Manal Swairjo, Ph.D.** Western University of Health Sciences Pomona, California Mutsuo Yamaguchi, Ph.D. Xueyong Zhu, Ph.D. R E S E A R C H A S S O C I AT E S Sunny Abraham, Ph.D.** Ambit Biosciences San Diego, California Melanie Ann Adams, Ph.D. Fabio Agnelli, Ph.D. Moballigh Ahmad, Ph.D.** University of Illinois Urbana-Chapaign, Illinois Alexander Ivanov Alexandrov, Ph.D. SENIOR RESEARCH A S S O C I AT E S David Barondeau, Ph.D.** Texas A&M Unversity College Station, Texas Kirk Beebe, Ph.D. Published by TSRI Press®. © Copyright 2006, The Scripps Research Institute. All rights reserved. Stephen G. Aller, Ph.D. Marcius Da Silva Almeida, Ph.D.** Universidade Federal do Rio de Janeiro Rio De Janeiro, Brazil Wojciech Augustyniak, Ph.D. Manidipa Banerjee, Ph.D. Christopher Baskerville, Ph.D.** Veterans Administration Medical Center San Diego, California Jianhan Chen, Ph.D. Yen-Ju Chen, Ph.D. Zhiyong Chen, Ph.D. Srinivas Chittaboina, Ph.D. Jungwoo Choe, Ph.D. Chung Jen Chou, Ph.D. Li-Chiou Chuang, Ph.D. Lipika Basummalick, Ph.D.** current employer unknown relocated to Bay Area Jean-Pierre Clamme, Ph.D.** Université Louis Pasteur Strasbourg Strasbourg, France Konstantinos Beis, Ph.D. Linda Maria Columbus, Ph.D. Per Bengston, Ph.D.** Lund University Lund, Sweden Stephen Connelly, Ph.D. Christine, Beuck, Ph.D. William Henry Bisson, Ph.D.** Burnham Institute La Jolla, California Adam Corper, Ph.D. Qizhi Cui, Ph.D. †††† la P. Da Costa, Ph.D. Sanjib Das, Ph.D.** Takeda San Diego, California David Boehr, Ph.D. Surya Kanta De, Ph.D. David Bostick, Ph.D. Robert De Bruin, Ph.D. MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 155 Sohela De Rozieres, Ph.D.** Biomatrica San Diego, California Bong Kwan Han, Ph.D.** University of California San Diego, Calfornia Dae Hee Kim, Ph.D. Derrick Meinhold, Ph.D. Eda Koculi, Ph.D. Elena Menichelli, Ph.D. Qingdong Deng, Ph.D.** Klypsy, Inc. San Diego, California Byung Woo Han, Ph.D. Bethany Koehntop, Ph.D. Jonathan Mikolosko, Ph.D. Shoufa Han, Ph.D. Milka Kostic, Ph.D. Peter J. Mikulecky, Ph.D. Paula Desplats, Ph.D. Wenge Han, Ph.D. Irina Kufareva, Ph.D. Mauro Mileni, Ph.D. Claire Louise Dovey, Ph.D. Shantanu Kumar, Ph.D. Zhanna Druzina, Ph.D. Jason W. Harger, Ph.D.** Illumina, Inc. San Diego, California Susumu Mitsumori, Ph.D.** Shionogi Research Laboratories Osaka, Japan Li-Lin Du, Ph.D. Rodney Harris, Ph.D. Michelle Duquette-Huber, Ph.D. David M. Herman, Ph.D. Scott Eberhardy, Ph.D.** Schering-Plough Union, New Jersey Stephen Edgcomb, Ph.D. Deron Herr, Ph.D. Jason Lanman, Ph.D. Wen-Xu Hong, Ph.D. Kwan Hoon Hyun, Ph.D. Daniel Felitsky, Ph.D. Wonpil Im, Ph.D.** University of Kansas Lawrence, Kansas Jinhyuk Lee, Ph.D.** University of Kansas Lawrence, Kansas Tetsuji Mutoh, Ph.D. June Hyung Lee, Ph.D. Hung Nguyen, Ph.D. Kelly Lee, Ph.D. George Nicola, Ph.D. Edward Lemke, Ph.D. Tadateru Nishikawa, Ph.D. Masanori Imai, Ph.D. Chenglong Li, Ph.D.** Ohio State University Columbus, Ohio Kyoko Noguchi, Ph.D. Liao Liang, Ph.D. Brian V. Norledge, Ph.D. Severn School Severna Park, Maryland Veli-Pekka Jaakola, Ph.D. Kai Jenssen, Ph.D. Vasco Liberal, Ph.D. William M. Lindstrom, Ph.D. Yann Gambin, Ph.D. Hui Gao, Ph.D.** Arena Pharmaceuticals San Diego, California Eric C. Johnson, Ph.D.** Bruker BioSpin Corporation Fremont, California Kunheng Luo, Ph.D. Hui-Yue Christine Lo, Ph.D. †††† Ann MacLaren, Ph.D. Hope Johnson, Ph.D. Darly Joseph Manayani, Ph.D. Shannon E. Gardell, Ph.D. Margaret Alice Johnson, Ph.D. Joshua Gill, Ph.D. Susanna Juraja, Ph.D. Edith Caroline Glazer, Ph.D. Christian Kannemeier, Ph.D. Bettina Groschel, Ph.D. Mili Kapoor, Ph.D. Fang Guo, Ph.D.** Department of Immonology Scripps Research Seongho Moon, Ph.D.** Samsung Electronic Seoul, South Korea Samrat Mukhopadhyay, Ph.D. Glenn C. Johns, Ph.D. Ionian Technologies Upland, California Elsa D. Garcin, Ph.D. Marissa Mock, Ph.D. Chul Won Lee, Ph.D. Kenichi Hitomi, Ph.D. Li Fan, Ph.D. Pierre Henri Gaillard, Ph.D.** Centre National de la Recherche Scientifique Marseille, France Emma Langley, Ph.D. Chang-Wook Lee, Ph.D. Yunfeng Hu, Ph.D. Josephine Chu Ferreon, Ph.D. Bianca Lam, Ph.D. Joreg Hinnerwisch, Ph.D. Susanna V. Ekholm-Reed, Ph.D. Allan Chris Merrera Ferreon, Ph.D. Sharon Kwan, Ph.D. Andrey Aleksandrovich Karyakin, Ph.D. Yang Khandogin, Ph.D. Jeff Mandell, Ph.D. Maria Victoria MartinSanchez, Ph.D.** University of Geneva Geneva, Switzerland Santiago Cavero Martinez, Ph.D. Sujatha Narayan, Ph.D. Wataru Nomura, Ph.D. Wendy Fernandez Ochoa, Ph.D.** University of California San Diego, California Amy Odegard, Ph.D. Lisa Renee Olano, Ph.D. Brian L. Olson, Ph.D. Mary O’Reilly, Ph.D. Brian Paegel, Ph.D. Sandeep Patel, Ph.D.** University of Delaware Newark, Delaware Stephanie Pebernard, Ph.D. Min Guo, Ph.D. Ilja V. Khavrutskii, Ph.D. Hanna-Stina Martinsson Ahlzén, Ph.D. Mahender Gurram, Ph.D. Reza Khayat, Ph.D. Tsutomu Matsui, Ph.D. Bill Francesco Pedrini, Ph.D. Robert Pejchal, Ph.D. 156 MOLECULAR BIOLOGY Vladimir Pelmenschikov, Ph.D. 2006 Daniela Andrea Slavin, Ph.D. Shun-ichi Wada, Ph.D. †††† Elisabetta Soragni, Ph.D. Ross Walker, Ph.D. San Diego Supercomputer La Jolla, California Jefferson Perry, Ph.D. Suzanne Peterson, Ph.D. Jessica Petrillo, Ph.D. oran Pljevaljcic, Ph.D. Stephanie Pond, Ph.D. Owen Pornillos, Ph.D.** Celgene Corporation San Diego, California Daniel Joseph Price, Ph.D.** GlaxoSmithKline Research Triangle Park, North Carolina John Prudden, Ph.D. Grazia Daniela Raffa, Ph.D.** Università di Roma La Sapienza Rome, Italy THE SCRIPPS RESEARCH INSTITUTE Holly Heaslet Soutter, Ph.D.** Pfizer Global Research & Development Ann Arbor, Michigan Greg Springsteen, Ph.D. Furman University Greenville, South Carolina S.V. Ramasastry Sripada, Ph.D. Thomas Steinbrecher, Ph.D. Gudrun Stengel, Ph.D.** University of Lund Lund, Sweden Shih-Che Su, Ph.D. Magnus Sundstrom, Ph.D. Blair R. Szymczyna, Ph.D. Jessica Williams, Ph.D. Robert Scott Williams, Ph.D. Wei Zhang, Ph.D.** Southwest Medical Center Houston, Texas Yong Zhao, Ph.D. Peizhi Zhu, Ph.D.** University of Michigan Arbor, Michigan Eric L. Wise, Ph.D. S C I E N T I F I C A S S O C I AT E S Jonathan Wojciak, Ph.D. Enrique Abola, Ph.D. Vance Wong, Ph.D. Andrew S. Arvai, M.S. Timothy I. Wood, Ph.D.** Walter Reed Army Medical Center Washington, D.C Ognian V. Bohorov, Ph.D. Eugene Wu, Ph.D.** Duke University Medical Center Durham, North Carolina Dennis Carlton, B.S. Vadim Cherezov, Ph.D. Ellen Yu-Lin Tsai Chien, Ph.D. Xiaoping Dai, Ph.D. Marc Deller, D.Phil Florence Muriel Tama, Ph.D.** University of Arizona Tucson, Arizona Wei Xie, Ph.D. Atsushi Yamagata, Ph.D. Riturparna Sinha Roy, Ph.D. Nardos Tassew, Ph.D.** University of Toronto Toronto, Canada Stanislav Rudyak, Ph.D. †††† Hiroaki Tateno, Ph.D. Sean Ryder, Ph.D.** University of Massachusetts Medical School Worcester, Massachusetts Rebecca E. Taurog, Ph.D. Ewan Richardson Taylor, Ph.D. Yong Yao, Ph.D.** Burnham Institute La Jolla, California Manami R. Saha, Ph.D.†††† Hua Tian, Ph.D. Yongjun Ye, Ph.D. Sanjay Adrian Saldanha, Ph.D. Mauricio Carrillo Tripp, Ph.D. Kye Sook Yi, Ph.D. Gabriela Perez-Alvarado, Ph.D.** Southern Illinois University Carbondale, Illinois Ulrich Ignaz Tschulena, Ph.D. Yong Yin, Ph.D. Nicholas Preece, Ph.D. †††† Andre Schiefner, Ph.D. Julie L. Tubbs, Ph.D. Kenji Yoshimoto, Ph.D. Lauren J. Schwimmer, Ph.D. Lin Wang, Ph.D. Naoto Utsumi, Ph.D. Jennifer S. Scorah, Ph.D. Frank van Drogen, Ph.D.** Institute für Biochemie Zürich, Switzerland Christopher L. Reyes, Ph.D. Biogen Idec Research San Diego, California Alim Seit-Nebi, Ph.D. Pedro Serrano-Navarro, Ph.D. Craig McLean Shepherd, Ph.D. David S. Shin, Ph.D. Develeena Shivakumar, Ph.D. David A. Shore, Ph.D. Published by TSRI Press®. © Copyright 2006, The Scripps Research Institute. All rights reserved. Ajay Vashisht, Ph.D. Philip Arno Venter, Ph.D. Petra Verdino, Ph.D. William Frederick Waas, Ph.D. Gye Won Han, Ph.D. Lan Xu, Ph.D. Michael Allen Hanson, Ph.D. Yoshiki Yamada, Ph.D. Marcy A. Kingsbury, Ph.D. Diane Marie Kubitz, B.A. Qi Yan, Ph.D.** Miramar College San Diego, California Padmaja Natarajan, Ph.D. Marianne Patch, Ph.D.** Qualcomm San Diego, California Naoto Yoshizuka, Ph.D. V I S I T I N G I N V E S T I G AT O R S Veronica Yu, Ph.D.†††† Yuan Yuan, Ph.D. Markus Zeeb, Ph.D. Ying Zeng, Ph.D. Haile Zhang, Ph.D. Qing Zhang, Ph.D. Stephen J. Benkovic, Ph.D. Pennsylvania State University University Park, Pennsylvania Astrid Graslund, Ph.D. Stockholm University Stockholm, Sweden MOLECULAR BIOLOGY Arne Holmgren, M.D., Ph.D. Karolinska Institutet Stockholm, Sweden 2006 * Joint appointment in The Skaggs Institute for Chemical Biology ** Appointment completed; new location shown Barry Honig, Ph.D. Columbia University New York, New York *** Joint appointment in the Molecular and Integrative Neurosciences Department Arthur Horwich, M.D. Yale University New Haven, Connecticut **** Joint appointments in the Shie-Liang Hsieh, Ph.D. National Yang-Ming University Taipei, Taiwan ***** Joint appointments in the Department of Immunology and The Skaggs Institute for Chemical Biology Department of Chemistry and The Skaggs Institute for Chemical Biology † Joint appointment in the Tai-Huang Huang, Ph.D. Academica Sinica Taipei, Taiwan Department of Cell Biology †† Joint appointment in the Department of Molecular and Experimental Medicine Sunghoon Kim, Ph.D. Seoul National University Seoul, Korea Ayori Mitsutake, Ph.D. Keio University Yokohama, Japan Joseph David Ng, Ph.D. University of Alabama, Huntsville Huntsville, Alabama Victoria A. Roberts, Ph.D. University of California San Diego, California Robert D. Rosenstein, Ph.D. Lawrence Berkeley National Laboratory Berkeley, California Lincoln Scott, Ph.D. Cassia, LLC San Diego, California Deborah Tahmassebi, Ph.D. University of San Diego San Diego, California ††† Joint appointment in the Department of Chemistry †††† Appointment completed THE SCRIPPS RESEARCH INSTITUTE 157 158 MOLECULAR BIOLOGY 2006 Peter E. Wright, Ph.D. Chairman’s Overview esearch in the Department of Molecular Biology encompasses a broad range of disciplines, extending from structural and computational biology at one extreme to molecular genetics at the other. During the past year, our scientists have continued to make rapid progress toward understanding the fundamental molecular events underlying the processes of life. Major advances have been made in elucidating the structural biology of signal transduction and viral assembly, in understanding mechanisms of viral infectivity, in determining the structures of membrane proteins and multidrug transporters, in understanding the molecular basis of nucleic acid recognition and DNA repair, and in determining the mechanisms of protein folding and ribosome assembly. Progress has been made in elucidating the molecular events involved in regulation of the cell cycle, in tumor development, in induction of sleep, in the molecular origins of neuronal development and of CNS disorders, in the regulation of transcription, and in the decoding of genetic information in translation. Finally, new advances have been made in the design of novel low molecular weight compounds that can specifically regulate genes, and in the area of biomolecular engineering, building novel functions into viruses, antibodies, and zinc finger proteins, RNA, and DNA. Progress in these and other areas is described in detail on the following pages, and R THE SCRIPPS RESEARCH INSTITUTE only a few highlights are mentioned below. The Department of Molecular Biology is also home to two major National Institutes of Health initiatives, the Joint Center for Structural Genomics and the Consortium for Functional Glycomics. One of the outstanding achievements of the past year was the determination of the structure of the intact and infectious P22 virion by electron cryomicroscopy. Research led by Jack Johnson has provided a remarkably detailed view of the virion structure at an unprecedented 17-Å resolution. The structure revealed the DNA tightly spooled around the portal in the interior of the capsid and suggested that the virus uses a pressure-sensing mechanism to control DNA packaging. The structure also provides insights into the mechanisms of virion assembly and injection of DNA into target cells. Structural biology continues to be a major focus in the department, and many new x-ray and nuclear magnetic resonance structures of major biomedical significance were completed during the past year. Geoffrey Chang and colleagues reported new structural studies of the Escherichia coli multidrug transporter EmrD, obtaining new insights into the mechanisms by which a diverse range of drugs are transported through the cell membrane. Such understanding is of major importance, given the rapidly growing problem of drug resistance in bacteria. John Tainer and his coworkers used a combination of electron cryomicroscopy and x-ray crystallography to determine the structure of the Type IV pilus filament of Neisseria gonorrhoeae. These studies provide new insights into assembly and disassembly mechanisms and are of importance because of the role played by Type IV pili in allowing antibiotic resistant strains to escape the immune system and cause persistent infections. Dr. Tainer and colleagues have also determined new structures of DNA repair enzymes; these include the xeroderma pigmentosum group B helicase, an enzyme that plays an essential role in nucleotide-excision repair by removing DNA lesions caused through exposure to ultraviolet light, and the exonuclease domain of WRN, a protein that protects against premature aging and cancer. Defects in the gene for WRN result in Werner’s syndrome, an inherited disease that causes premature aging. Research in the laboratories of Jane Dyson and Peter Wright has provided new insights into the role of protein conformational fluctuations in enzyme catalysis. Protein dynamics have long been thought to play an important role in catalysis. This new work shows how the dynamic MOLECULAR BIOLOGY 2006 energy landscape of the enzyme dihydrofolate reductase channels the protein through the reaction cycle. Conformational transitions between the various conformational substates of the enzyme occur at a rate that is directly relevant to catalysis. Several research groups are working in areas directly related to drug discovery and protein therapeutics. Joel Gottesfeld and his colleagues have developed smallmolecule histone deacetylase inhibitors that reactivate frataxin, the gene responsible for the neurodegenerative disease Friedreich’s ataxia, a disease that is associated with the expansion of triplet repeats in DNA. These compounds hold great promise as potential therapeutics for Freidreich’s ataxia. Subhash Sinha, Carlos Barbas, and Richard Lerner have developed a unique self-assembly strategy to direct antibodies against specific cellular targets. Their novel approach has led to new compounds targeted against metastatic breast cancer. Many of the research groups in this department are applying the tools of molecular and structural biology to understand the molecular basis of human disease. In research led by James Paulson and Ian Wilson, glycan microarray technology is being used to identify mutations that could allow avian influenza viruses to adapt to the human population. The glycan array is a powerful surveillance tool for mapping the pathways by which new human pathogenic viruses can emerge. This research has revealed a potential mutational pathway that could switch the specificity of the highly pathogenic H5N1 avian influenza virus and allow it to adapt to humans. Strikingly, the 3-dimensional structure of the H5N1 hemagglutinin, the protein responsible for binding the virus to host cell receptors, bears a closer resemblance to the hemagglutinin from the virus that caused the 1918 influenza pandemic than to that associated with more recent influenza outbreaks. Finally, research during the past year has greatly advanced our understanding of the complex mechanisms of cell-cycle regulation. Curt Wittenberg and his colleagues have identified a yeast protein that plays a central role in repressing transcription during the cell cycle. The protein functions in a parallel manner to the important metazoan transcriptional regulator E2F. Work in Steven Reed’s laboratory has provided new insights into the mechanisms of multiubiquitinylation and degradation of cyclin E, a process that is essential for the normal regulation of the cell cycle. Misregulation of either of these processes, transcriptional repression or cyclin turnover, is associated with cancer. THE SCRIPPS RESEARCH INSTITUTE 159 Molecular biology remains a field of enormous opportunity and excitement. The scientists in this department are taking full advantage of powerful new technologies to advance our understanding of fundamental biological processes at the molecular level. Their discoveries will ultimately be translated into new advances in biotechnology and in medicine. 160 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE INVESTIGATORS’ R EPORTS Structural Biology of Immune Recognition, Molecular Assemblies, and Anticancer Targets I.A. Wilson, R.L. Stanfield, J. Stevens, X. Zhu, M.A. Adams, Y. An, K. Beis, D.A. Calarese, R.M.F. Cardoso, J.E. Carlson, P.J. Carney, J.-W. Choe, S. Connelly, A.L. Corper, T.H. Cross, F i g . 1 . A, Structure of the H5 A/Vietnam/1203/2004 (Viet04) X. Dai, E.W. Debler, W.L. Densley, M.-A. Elsliger, S. Ferguson, B.W. Han, G.W. Han, M.J. Jimenez-Dalmaroni, J.G. Luz, J.R. Mikolosko, A. Schiefner, D.A. Shore, R.S. Stefanko, J.A. Vanhnasy, P. Verdino, E. Wise, L. Xu, X. Xu, D.M. Zajonc hemagglutinin trimer, represented as a ribbon diagram. The receptor binding domain, cleavage, and basic patch sites are highlighted on one monomer. Only 2 of the 9 glycosylation sites per monomer (positions 34 and 169 in the HA1 chain) had interpretable carbohydrates in the electron density maps. B, Glycan microarray analyses of wild-type human Viet04 hemagglutinin and mutations at positions 226 and 228, known to be important for adaptation of H3 viruses from avian α2-3 specificity to human α2-6 receptor specificity. Binding to the different avian and human α2-3 and α26 sialosides on the array are highlighted. e are working toward a better understanding of the structure and function of a variety of immune-related receptors and of other medically relevant proteins. We use x-ray crystallography to determine structures for these molecules in complex with their ligands and coreceptors. This research is instrumental for the design of future drugs and vaccines to target these proteins. W INFLUENZA VIRUS Influenza virus is a highly contagious and deadly agent that causes acute respiratory illness. The current H5N1 avian influenza virus has reached epizootic levels in domestic and wild birds, with worldwide debate whether the next influenza pandemic could arise from one of these avian strains. Hemagglutinin is the principal viral surface antigen and is responsible for binding to host receptors through interaction with sialylated glycans. The structure of the hemagglutinin from a highly pathogenic H5N1 influenza virus (A/Vietnam/1203/2004; Fig. 1A) is more closely related to the human 1918 H1 hemagglutinin than to the other human, avian, and swine hemagglutinins. We are also examining crystal structures of (1) various influenza neuraminidases to determine the specificity of the enzymes and their involvement in interaction/escape of the virus from current drugs and (2) influenza viral proteins that interact with components of the apoptosis signaling pathway. In collaboration with O. Blixt and J. Paulson of the Consortium for Functional Glycomics, La Jolla, California, we used their recently described glycan microarray technology to assess the propensity of the avian receptor H5N1 A/Vietnam/1203/2004 hemagglutinin to change from its avian receptor binding (α2-3-linked sialic acids) to adapt to human receptors (α2-6-linked sialic acids; Fig. 1B) and have elucidated a possible route by which H5 viruses could gain a foothold in the human population. IL-2 RECEPTOR IL-2 is a cytokine that functions as a T-cell growth factor and a central immune system regulator. Its importance is underlined by its broad use as a therapeutic agent against cancers of the immune system, and IL-2 antagonists are used to prevent rejection of transplanted organs. We have determined the structure of the heterotrimeric IL-2 receptor ectodomains (IL-2Rαβγc) in complex with IL-2 at 3.0-Å resolution (Fig. 2). Surprisingly, IL-2Rα makes no contacts with IL-2Rβ or IL-2Rγc, and only minor changes occur in IL-2 in response to receptor binding. Thus, our findings support the notion that IL-2Rα delivers IL-2 to the signaling complex and acts as a regulator of signal transduction. This research was performed in collaboration with K.A. Smith, Cornell University Weill Medical College, New York, New York. T H E I N N AT E I M M U N E S Y S T E M Toll-like receptors (TLRs) play key roles in activating immune responses during infection. The 2.1-Å structure of the human TLR3 ectodomain revealed a large horseshoe-shaped solenoid structure assembled from 23 leucine-rich repeats. Seven conserved hydro- MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 161 Department of Immunology, we are studying the membrane-bound part of the NADPH complex to correlate how mutations in NADPH oxidase can cause chronic granulomatous disease. The nucleotide oligomerization binding domain 2 is an important intracellular receptor that recognizes bacterial peptidoglycans. Mutations in this receptor are associated with the inflammatory Crohn’s disease. Structural studies are under way on the domains and on full-length protein, in collaboration with R. Ulevitch, Department of Immunology. C ATA LY T I C A N T I B O D I E S F i g . 2 . Architecture of the trimeric human IL-2 receptor (desig- nated IL2R in the figure) signaling complex. View of the quaternary IL-2 signaling assembly composed of α, β, and γc chains of the IL2R and IL-2, with the C terminus of the β and γ chains close to the membrane. IL-2 binds to the elbow regions of IL-2Rβ and IL-2Rγc, as in other cytokine receptors such as human growth hormone receptor and erythropoietin receptor. The novel IL-2Rα chain docks on top of this assembly but does not form any contacts with the other 2 receptor subunits. Six N-linked carbohydrates (S1–S6) are displayed as ball-and-stick models. S1 is wedged between D1 and D2 of IL-2Rβ and thus contributes to the stabilization of a specific D1/D2 interdomain angle. IL-2Rβ and IL-2Rγ form a 3-way junction with IL-2 at the heart of the quaternary high-affinity IL-2 signaling complex and provide a structural basis for the cooperativity in assembly of the complete IL-2 signaling complex. phobic residues in the leucine-rich repeat motif form a tight hydrophobic core, and conserved asparagines contribute extensive hydrogen-bonding networks for solenoid stabilization. TLR3 is largely masked by carbohydrate, but the only glycosylation-free face may provide potential ligand-binding sites and an oligomerization interface. We are doing biochemical analysis of the interaction between the TLR3 ectodomain and various doublestranded RNA oligomers and structural investigations of TLR1, TLR2, TLR6, and the TLR2 coreceptor CD36. These projects are a collaboration with B. Beutler and R. Ulevitch, Department of Immunology. Neutrophils and other phagocytes play an important role in innate immunity by serving as a first line of defense against invading pathogens. Generation of superoxide by the phagocyte NADPH oxidase complex initiates this process by catalyzing the transfer of metabolic electrons across the plasma membrane for reduction of molecular oxygen. Individuals deficient in this enzymatic activity have chronic granulomatous disease, characterized by recurrent, life-threatening bacterial and fungal infections. In collaboration with G. Bokoch, Abuse of cocaine is a major public health problem; however, no treatments approved by the Food and Drug Administration are available for cocaine abuse, addiction, or overdose. Development of effective treatments for cocaine abuse has been frustrated by the complex neurochemistry of cocaine addiction. Nevertheless, within the past decade, immunotherapy for cocaine abuse has been evaluated in preclinical and clinical trials. In collaboration with K.D. Janda, Department of Chemistry, we determined high-resolution structures for the cocaine catalytic antibody 7A1 for all major steps along the catalytic reaction pathway, through cocrystallization with substrate, products, and transition-state analogs (Fig. 3). On the basis of this comprehensive series of F i g . 3 . Crystal structure of the antibody 7A1 Fab′ fragment in complex with cocaine. The secondary structure of the Fab′ and the substrate cocaine are shown. Cocaine is trapped in the active site and is hydrolyzed to nontoxic metabolites. crystal structures, a catalytic mechanism has been proposed, as well as possible mutations to improve catalytic proficiency. 162 MOLECULAR BIOLOGY 2006 C O FA C T O R - C O N TA I N I N G A N T I B O D I E S Although antibodies are generally thought to function without use of cofactors, they are major carrier proteins in human circulation for the biologically important cofactor riboflavin. A riboflavin-containing bright-yellow antibody, IgG GAR, was purified from a patient with multiple myeloma 30 years ago and is the only available material for studies of the structure and function of natural cofactor-containing antibodies. Our recent 3.0-Å crystal structure of GAR reveals the location in the antibody-combining site for the riboflavin potential cofactor (Fig. 4). This research was carried out THE SCRIPPS RESEARCH INSTITUTE biochemical studies with our collaborators, R.A. Lerner, K.D. Janda, P.G. Schultz, and F.E. Romesberg, Department of Chemistry. EVOLUTION OF LIGAND RECOGNITION AND SPECIFICITY To enhance our understanding of how recognition and specificity for different ligands can be accomplished by different antibodies that have high levels of sequence homology, we are studying the evolution of ligand-binding properties by site-directed mutagenesis. The most active catalytic Diels-Alder antibody known to date, 1E9, and the steroid-binding antibody DB3 are derived from the same germ line and have 85% sequence identity. Through sequential amino acid exchanges, the specificity of 1E9 was changed to that of DB3. Thus, only a few binding site residues are responsible for achieving either efficient catalysis of the Diels-Alder reaction or, when mutated, a strong steroid binder. In collaboration with D. Hilvert, ETH, Zürich, Switzerland, we are structurally characterizing these 1E9 mutants to show how relatively minor changes can be rationally used to modify antibody specificity and function. HIV TYPE 1 NEUTRALIZING ANTIBODIES F i g . 4 . The antigen-binding site of the original yellow antibody IgG GAR. The riboflavin cofactor is inserted into the combining site with its isoalloxazine ring stacked between aromatic residues TyrH33, PheH58, and TyrH100A. Together with hydrogen bonds between the N5 atom of the ring to AsnH50 and the ribityl side chain to ArgH52 and GluH56, these interactions reveal the structural basis for high-affinity riboflavin binding. in collaboration with R.A. Lerner and P. Wentworth, Jr., Department of Chemistry. BLUE AND PURPLE FLUORESCENT ANTIBODIES Catalytic antibodies are designed to accelerate chemical reactions by acting on the electronic ground state. However, antibodies have been generated that can interact with and direct the photochemical behavior of the electronically excited state of stilbene, a model compound for studies in photochemistry and photophysics. We are exploring the structural basis of the diverse fluorescent properties of these complexes by using x-ray crystallography in combination with biophysical and The search for an effective HIV type 1 vaccine has prompted the study of the few known broadly neutralizing antibodies to HIV type 1 in complex with their antigens, in order to structurally characterize important viral epitopes. The potent and broadly neutralizing antibodies include 4E10 and Z13, which bind to conserved and overlapping epitopes on the membrane-proximal region of gp41, and 2G12, which binds to a carbohydrate cluster rich in mannose on gp120. These crystal structures are then used as the basis for rational design of immunogens for a candidate vaccine against HIV type 1. This research is done in collaboration with D. Burton, Department of Immunology; P. Dawson, Department of Cell Biology; C.-H. Wong, Department of Chemistry; S. Danishefsky, Sloan-Kettering Institute, New York, New York; J.K. Scott, Simon Fraser University, Burnaby, British Columbia; J. Moore, Cornell University, Ithaca, New York; H. Katinger, R. Kunert, and G. Stiegler, University für Bodenkultur, Vienna, Austria; R. Wyatt and P. Kwong, Vaccine Research Center, National Institutes of Health, Bethesda, Maryland; and the Neutralizing Antibody Consortium of the International AIDS Vaccine Initiative, New York, New York. CLASSICAL AND NONCLASSICAL MHC AND T-CELL RECEPTOR SIGNALING An inflammatory joint disease with many similarities to human rheumatoid arthritis develops spontaneously in MOLECULAR BIOLOGY 2006 KRN T-cell receptor (TCR) transgenic mice (F1 K/B x N mice). Class II MHC I-Ag7 presentation to KRN of selfpeptide derived from glucose-6-phosphate isomerase is a critical step in the initiation of the disease. In collaboration with L. Teyton, Department of Immunology, we determined the crystal structures of I-Ag7–glucose-6phosphate isomerase peptide and of the TCR KRN. We are attempting to crystallize the KRN–I-A g7 complex to enhance our understanding of how this autoimmune disease is mediated at the molecular level. The CD3 TCR coreceptor comprises several distinct cell-surface glycoproteins that associate with TCR to enable intracellular signal transduction upon the formation of complexes consisting of TCR and MHC-peptides. Structural investigation into the interaction between the TCR and CD3 subunits can aid in elucidation of the events that lead to T-cell activation. The CD8 glycoprotein is essential for the class I MHC-restricted T-cell response to peptide antigen, analogous to the CD4 coreceptor of class II–restricted T cells. CD8 is expressed at the cell surface as CD8αα and CD8αβ. We have determined structures for both CD8αα and CD8αβ in complex with antibody Fab fragments. Comparison of both forms of the CD8 coreceptor have provided insight into how the α and β forms contribute to the functionality of CD8. These studies are a collaboration with S. Davis, University of Oxford, Oxford, England, and L. Teyton, Department of Immunology. The CD1 family is structurally related to MHC molecules, but members of the family present lipid antigens rather than peptides to CD1-restricted TCRs. We have determined several structures of mouse CD1d in complex with α-galacturonosyl ceramide, cis-tetracosenoyl sulfatide, or mycobacterial phosphatidylinositol dimannoside. For each CD1d-ligand, the lipid tails are embedded in the CD1 hydrophobic binding groove, and a restricted set of CD1d residues orient and stabilize the various different antigenic headgroups for TCR recognition (Fig. 5). Collaborators in research on CD1 and TCRs include D.B. Moody and M.B. Brenner, Harvard Medical School, Boston, Massachusetts; C.-H. Wong, Department of Chemistry; L. Teyton, Department of Immunology; M. Kronenberg, La Jolla Institute for Allergy and Immunology, San Diego, California; V. Kumar, Torrey Pines Institute for Molecular Studies, San Diego, California; and W. Severn and G. Painter, Industrial Research Ltd., Upper Hut, New Zealand. PROTEIN TRAFFICKING Molecular tethers play a critical role in the organization of the membrane architecture of the exocytic and THE SCRIPPS RESEARCH INSTITUTE 163 F i g . 5 . Structure of mouse CD1d with inositol-dimannoside. Close-up view of the binding site shows the hydrogen-bonding network between the glycolipid and CD1d. Both alkyl chains of the ligand are deeply buried inside the binding groove (not shown), whereas the complex inositol-dimannoside headgroup is optimally positioned above the binding groove to directly interact with the TCR. endocytic pathways of eukaryotic cells. In collaboration with W. Balch, Department of Cell Biology, we have determined the 2.0-Å structure of the Rab1 GTPaseregulated N-terminal domain of the p115 tether involved in transport and structural organization of the Golgi complex. The structure reveals a dimeric handshakelike assembly consisting of 2 α-solenoid chains, each with 12 novel armadillo-like, tetherin trihelical repeat elements that form a superhelical elliptical cylinder. This structure supports a model for binding of Rab1 on opposing membranes to promote membrane tether assembly for membrane docking and fusion and for understanding the large family of molecular tethers. JOINT CENTER FOR STRUCTURAL GENOMICS The Joint Center for Structural Genomics is a large consortium of scientists from Scripps Research; the Stanford Synchrotron Radiation Laboratory; the University of California, San Diego; the Burnham Institute; and the Genomics Institute of the Novartis Research Foundation. The center is funded by the Protein Structure Initiative of the National Institute of General Medical Sciences. Its purpose is the high-throughput structure determination of large protein families with no structural representatives, a biologically important group of targets that are conserved in the central machinery of 164 MOLECULAR BIOLOGY 2006 life; the complete proteome from Thermotoga maritima; and targets suggested by the community. To date, members of the consortium have pioneered many novel highthroughput methods, constructed a high-throughput pipeline, and determined more than 270 nonredundant structures. PUBLICATIONS Almeida, M.S., Herrmann, T., Peti, W., Wilson, I.A., Wüthrich, K. NMR structure of the conserved hypothetical protein TM0487 from Thermotoga maritima: implications for 216 homologous DUF59 proteins. Protein Sci. 14:2880, 2005. Brunel, F.M., Zwick, M.B., Cardoso, R.M., Nelson, J.D., Wilson, I.A., Burton, D.R., Dawson, P.E. Structure-function analysis of the epitope for 4E10, a broadly neutralizing human immunodeficiency virus type 1 antibody. J. Virol. 80:1680, 2006. Burton, D.R., Stanfield, R.L., Wilson, I.A. Antibody vs HIV in a clash of evolutionary titans. Proc. Natl. Acad. Sci. U. S. A. 102:14943, 2005. Calarese, D.A., Lee, H.K., Huang, C.Y., Best, M.D., Astronomo, R.D., Stanfield, R.L., Katinger, H., Burton, D.R., Wong, C.-H., Wilson, I.A. Dissection of the carbohydrate specificity of the broadly neutralizing anti-HIV-1 antibody 2G12. Proc. Natl. Acad. Sci. U. S. A. 102:13372, 2005. Cheng, H., Chong, Y., Hwang, I., Tavassoli, A., Zhang, Y., Wilson, I.A., Benkovic, S.J., Boger, D.L. Design, synthesis, and biological evaluation of 10-methanesulfonyl-DDACTHF, 10-methanesulfonyl-5-DACTHF, and 10-methylthio-DDACTHF as potent inhibitors of GAR Tfase and the de novo purine biosynthetic pathway. Bioorg. Med. Chem. 13:3577, 2005. Cheng, H., Hwang, I., Chong, Y., Tavassoli, A., Webb, M.E., Zhang, Y., Wilson, I.A., Benkovic, S.J., Boger, D.L. Synthesis and biological evaluation of N-[4-[5(2,4-diamino-6-oxo-1,6-dihydropyrimidin-5-yl)-2-(2,2,2-trifluoroacetyl)pentyl]benzoyl]-L-glutamic acid as a potential inhibitor of GAR Tfase and the de novo purine biosynthetic pathway. Bioorg. Med. Chem. 13:3593, 2005. Choe, J., Kelker, M.S., Wilson, I.A. Crystal structure of human Toll-like receptor 3 (TLR3) ectodomain. Science 309:581, 2005. Chong, Y., Hwang, I., Tavassoli, A., Zhang, Y., Wilson, I.A., Benkovic, S.J., Boger, D.L. Synthesis and biological evaluation of α- and γ-carboxamide derivatives of 10CF3CO-DDACTHF. Bioorg. Med. Chem. 13:3587, 2005. DiDonato, M., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of a single-stranded DNA-binding protein (TM0604) from Thermotoga maritima at 2.60 Å resolution. Proteins 63:256, 2006. Giabbai, B., Sidobre, S., Crispin, M.D., Sanchez-Ruiz, Y., Bachi, A., Kronenberg, M., Wilson, I.A., Degano, M. Crystal structure of mouse CD1d bound to the self ligand phosphatidylcholine: a molecular basis for NKT cell activation. J. Immunol. 175:977, 2005. Glaser, L., Stevens, J., Zamarin, D., Wilson, I.A., Garcia-Sastre, A., Tumpey, T.M., Basler, C.F., Taubenberger, J.K., Palese, P. A single amino acid substitution in 1918 influenza virus hemagglutinin changes receptor binding specificity. J. Virol. 79:11533, 2005. Han, G.W., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of an apo mRNA decapping enzyme (DcpS) from mouse at 1.83 Å resolution. Proteins 60:797, 2005. THE SCRIPPS RESEARCH INSTITUTE Johnson, M.A., Peti, W., Herrmann, T., Wilson, I.A., Wüthrich, K. Solution structure of Asl1650, an acyl carrier protein from Anabaena sp PCC 7120 with a variant phosphopantetheinylation-site sequence. Protein Sci. 15:1030, 2006. Klock, H.E., Schwarzenbacher, R., Xu, Q., et al. Crystal structure of a conserved hypothetical protein (gi: 13879369) from mouse at 1.90 Å resolution reveals a new fold. Proteins 61:1132, 2005. Luz, J.G., Yu, M., Su, Y., Wu, Z., Zhou, Z., Sun, R., Wilson, I.A. Crystal structure of viral macrophage inflammatory protein I encoded by Kaposi’s sarcoma-associated herpesvirus at 1.7 Å. J. Mol. Biol. 352:1019, 2005. Mathews, I.I., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of phosphoribosylformylglycinamidine synthase II (smPurL) from Thermotoga maritima at 2.15 Å resolution. Proteins 63:1106, 2006. Moody, D.B., Zajonc, D.M., Wilson, I.A. Anatomy of CD1-lipid antigen complexes. Nat. Rev. Immunol. 5:387, 2005. Peti, W., Page, R., Moy, K., O’Neil-Johnson, M., Wilson, I.A., Stevens, R.C., Wüthrich, K. Towards miniaturization of a structural genomics pipeline using micro-expression and microcoil NMR. J. Struct. Funct. Genomics 6:259, 2005. Rife, C., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of the global regulatory protein CsrA from Pseudomonas putida at 2.05 Å resolution reveals a new fold. Proteins 61:449, 2005. Rife, C., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of a putative modulator of DNA gyrase (pmbA) from Thermotoga maritima at 1.95 Å resolution reveals a new fold. Proteins 61:444, 2005. Shore, D.A., Teyton, L., Dwek, R.A., Rudd, P.M., Wilson, I.A. Crystal structure of the TCR co-receptor CD8αα in complex with monoclonal antibody YTS 105.18 Fab fragment at 2.88 Å resolution. J. Mol. Biol. 358:347, 2006. Stanfield, R.L., Gorny, M.K., Zolla-Pazner, S., Wilson, I.A. Crystal structures of human immunodeficiency virus type 1 (HIV-1) neutralizing antibody 2219 in complex with three different V3 peptides reveal a new binding mode for HIV-1 crossreactivity. J. Virol. 80:6093, 2006. Stanfield, R.L., Zemla, A., Wilson, I.A., Rupp, B. Antibody elbow angles are influenced by their light chain class. J. Mol. Biol. 357:1566, 2006. Stauber, D.J., Debler, E.W., Horton, P.A., Smith, K.A., Wilson, I.A. Crystal structure of the IL-2 signaling complex: paradigm for a heterotrimeric cytokine receptor. Proc. Natl. Acad. Sci. U. S. A. 10:2788 2006. Stevens, J., Blixt, O., Glaser, L., Taubenberger, J.K., Palese, P., Paulson, J.C., Wilson, I.A. Glycan microarray analysis of the hemagglutinins from modern and pandemic influenza viruses reveals different receptor specificities. J. Mol. Biol. 355:1143, 2006. Stevens, J., Blixt, O., Tumpey, T.M., Taubenberger, J.K., Paulson, J.C., Wilson, I.A. Structure and receptor specificity of the hemagglutinin from an H5N1 influenza virus. Science 312:404, 2006. Van Rhijn, I., Zajonc, D.M., Wilson, I.A., Moody, D.B. T-cell activation by lipopeptide antigens. Curr. Opin. Immunol. 17:222, 2005. Wilson, I.A., Stanfield, R.L. MHC restriction: slip-sliding away. Nat. Immunol. 6:434, 2005. Wiseman, R.L., Johnson, S.M., Kelker, M.S., Foss, T., Wilson, I.A., Kelly, J.W. Kinetic stabilization of an oligomeric protein by a single ligand binding event. J. Am. Chem. Soc. 127:5540, 2005. Huang, C.C., Tang, M., Zhang, M.Y., Majeed, S., Montabana, E., Stanfield, R.L., Dimitrov, D.S., Korber, B., Sodroski, J., Wilson, I.A., Wyatt, R., Kwong, P.D. Structure of a V3-containing HIV-1 gp120 core. Science 310:1025, 2005. Wu, D., Zajonc, D.M., Fujio, M., Sullivan, B.A., Kinjo, Y., Kronenberg, M., Wilson, I.A., Wong, C.-H. Design of natural killer T cell activators: structure and function of a microbial glycosphingolipid bound to mouse CD1d. Proc. Natl. Acad. Sci. U. S. A. 103:3972, 2006. Jaroszewski, L., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of Hsp33 chaperone (TM1394) from Thermotoga maritima at 2.20 Å resolution. Proteins 61:669, 2005. Xu, Q., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of virulence factor CJ0248 from Campylobacter jejuni at 2.25 Å resolution reveals a new fold. Proteins 62:292, 2006. Jin, K.K., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of TM1367 from Thermotoga maritima at 1.90 Å resolution reveals an atypical member of the cyclophilin (peptidylprolyl isomerase) fold. Proteins 63:1112, 2006. Zajonc, D.M., Cantu, C. III, Mattner, J., Zhou, D., Savage, P.B., Bendelac, A., Wilson, I.A., Teyton, L. Structure and function of a potent agonist for the semiinvariant natural killer T cell receptor. Nat. Immunol. 6:810, 2005. MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 165 Zajonc, D.M., Maricic, I., Wu, D., Halder, R., Roy, K., Wong, C.-H., Kumar, V., Wilson, I.A. Structural basis for CD1d presentation of a sulfatide derived from myelin and its implications for autoimmunity. J. Exp. Med. 202:1517, 2005. Zhang, Y., Wang, L., Schultz, P.G., Wilson, I.A. Crystal structures of apo wild-type M jannaschii tyrosyl-tRNA synthetase (TyrRS) and an engineered TyrRS specific for O-methyl-L-tyrosine. Protein Sci. 14:1340, 2005. Zhu, X., Dickerson, T.J., Rogers, C.J., Kaufmann, G.F., Mee, J.M., McKenzie, K.M., Janda, K.D., Wilson, I.A. Complete reaction cycle of a cocaine catalytic antibody at atomic resolution. Structure 14:205, 2006. Zhu, X., Wentworth, P., Jr., Kyle, R.A., Lerner, R.A., Wilson, I.A. Cofactor-containing antibodies: crystal structure of the original yellow antibody. Proc. Natl. Acad. Sci. U. S. A. 103:3581, 2006. Structure and Function of Proteins as Molecular Machines E.D. Getzoff, M. Aoyagi, A.S. Arvai, D.P. Barondeau, F i g . 1 . The NADPH-binding site in the crystallographic structure of R.M. Brudler, T.H. Cross, E.D. Garcin-Hosfield, C. Hitomi, the neuronal NOS reductase module. A triad of amino acid residues conserved in NOS reductases and homologous flavoproteins (Tyr1322, Ser1313, and Arg1314) stabilize the 2′ phosphate group (2′P) that distinguishes NADPH from NADH. In contrast, Arg1400 is specific to the calcium-regulated neuronal and endothelial NOS enzymes, in which it performs a isozyme-specific regulatory function. In the absence of calcium-bound calmodulin, Arg1400 helps stabilize the regulatory C-terminal tail, inhibiting nonproductive electron transfer. K. Hitomi, C.J. Kassmann, M.E. Pique, M.E. Stroupe, J.L. Tubbs, T.I. Wood ur goals are to understand how proteins function as molecular machines. We use structural, molecular, and computational biology to study proteins of biological and biomedical interest, especially proteins that work synergistically with coupled chromophores, metal ions, or other cofactors. O NITRIC OXIDE SYNTHASES To synthesize nitric oxide, a cellular signal and defensive cytotoxin, nitric oxide synthases (NOSs) require calmodulin-orchestrated interactions between their catalytic, heme-containing oxygenase module and their electron-supplying reductase module. Crystallographic structures of wild-type and mutant NOS oxygenase dimers with substrate, intermediate, inhibitors, cofactors, and cofactor analogs, determined in collaboration with J. Tainer, Department of Molecular Biology, and D. Stuehr, the Cleveland Clinic, Cleveland, Ohio, provided insights into the catalytic mechanism and dimer stability. Our structure-based drug design projects are aimed at selectively inhibiting inducible NOS, to prevent inflammatory disorders, or neuronal NOS, to prevent migraines, while maintaining blood pressure regulation by endothelial NOS. Our structure of the neuronal NOS reductase has provided news insights into the complex regulatory mechanisms of this enzyme family. We have designed and assayed site-directed mutant enzymes that support our mechanistic hypotheses for isozyme-specific inhibition and regulation (Fig. 1). We integrated biochemical data with our structures of NOS oxygenase, NOS reductase, and calmodulin in complex with peptides derived from NOS to propose a model for the assembled holoenzyme that provides a movingdomain mechanism for electron flow from NOS reductase to the NOS oxygenase heme. Preliminary small-angle x-ray scattering measurements in solution provide molecular envelopes for NOS proteins that support our model. Our model also explains how regulatory site-specific phosphorylation and dephosphorylation activate and inactivate nitric oxide synthesis in vivo. PHOTOACTIVE PROTEINS AND CIRCADIAN CLOCKS To understand in atomic detail how proteins translate sunlight into defined conformational changes for biological functions, we are exploring the reaction mechanisms of the blue-light receptors photoactive yellow protein (PYP), photolyase, and cryptochrome. PYP is the prototype for the Per-Arnt-Sim domain proteins of circadian clocks, whereas proteins of the photolyase and cryptochrome family catalyze DNA repair or act in circadian clocks. To understand the protein photocycle, we combined ultra-high-resolution and time-resolved crystallographic structures of the dark state and 2 photocycle intermediates of PYP with sitedirected mutagenesis; ultraviolet-visible spectroscopy; 166 MOLECULAR BIOLOGY 2006 time-resolved Fourier transform infrared spectroscopy; deuterium-hydrogen exchange mass spectrometry, in collaboration with V. Woods, University of California, San Diego; and quantum mechanical and electrostatic computational methods, in collaboration with L. Noodleman, Department of Molecular Biology. Cryptochrome flavoproteins are homologs of lightdependent DNA repair photolyases that function as bluelight receptors in plants and as components of circadian clocks in animals. We determined the first crystallographic structure of a cryptochrome, which revealed commonalities with photolyases in DNA binding and redox-dependent function but showed differences in active-site and interaction-surface features. Recently, we showed that this cryptochrome binds the same antenna cofactor found in a photolyase homolog but uses different residues for the cofactor-binding site. New structures of photolyases from 2 other branches of the photolyase/cryptochrome family that repair cyclobutane pyrimidine dimers and photoproducts help us decipher the cryptic structure, function, and evolutionary relationships of these fascinating redox-active proteins. We are also studying clock proteins with PYP-like and Per-Arnt-Sim domains that bind to mammalian cryptochromes. Our goal is to determine the detailed chemistry and atomic structure of these proteins, define their mechanisms of action and interaction, and use our results to understand their biological function and regulation. THE SCRIPPS RESEARCH INSTITUTE targeting to specific cellular locations. By completing the metalloprotein design cycle from prediction to highly accurate structures, we can rigorously evaluate and improve algorithms for the design of metal sites. In related research, we discovered that the architecture of GFP and RFP promotes a remarkable range of posttranslational modification chemistry. High-resolution crystallographic structures of GFP and RFP intermediates in fluorophore cyclization and oxidation lead to a novel mechanism for the spontaneous synthesis of this tripeptide fluorophore within the protein scaffold. Remarkably, the same protein architectural features that favor peptide cyclization can drive peptide hydrolysis (Fig. 2) and red shift the spectral properties of the P R O T E I N D E S I G N A N D P O S T R A N S L AT I O N A L M O D I F I C AT I O N C H E M I S T R Y An ultimate goal for protein engineers is to design and construct new protein variants with desirable catalytic or physical properties. As members of the Scripps Research Metalloprotein Structure and Design Group, we are testing our understanding of affinity, selectivity, and activity of metal ions by transplanting metal sites from structurally characterized metalloproteins into new protein scaffolds. To aid our design efforts, we have organized quantitative information and interactive viewing of protein metal sites at the Metalloprotein Database and Browser (available at http://metallo.scripps.edu). For green fluorescent protein (GFP) and the homologous red fluorescent protein (RFP), we designed, constructed, and characterized metal-ion biosensors, in which binding of metal ions is signaled by changes in spectroscopic properties of the naturally occurring fluorophores. Use of GFP allows optimization with random mutagenesis, noninvasive expression in living cells, and F i g . 2 . Spontaneous peptide hydrolysis and decarboxylation reactions promoted by the protein architecture of GFP. A, Crystallographic structure of a designed GFP variant reveals peptide-bond cleavage and decarboxylation chemistry at the site of GFP fluorophore synthesis. S65G and Y66S mutations converted the fluorophore tripeptide SYG sequence to GSG. The simulated annealing omit electron density map (mesh) clearly shows the resultant break in the polypeptide chain at this site. B, Corresponding reaction and posttranslational products for this self-cleaving GFP variant. MOLECULAR BIOLOGY 2006 chromophore. Decarboxylation reactions in designed variants of GFP (Fig. 2) support a role for the GFP environment in facilitating formation of radicals and 1-electron chemistry. Together, our results provide the groundwork for the design of proteins with novel catalytic or reporter properties. PUBLICATIONS Barondeau, D.P., Kassmann, C.J., Tainer, J.A., Getzoff, E.D. Understanding GFP posttranslational chemistry: structures of designed variants that achieve backbone fragmentation, hydrolysis, and decarboxylation. J. Am. Chem. Soc. 128:4685, 2006. Barondeau, D.P., Tainer, J.A., Getzoff, E.D. Structural evidence for an enolate intermediate in GFP fluorophore biosynthesis. J. Am. Chem. Soc. 128:3166, 2006. Brudler, R., Gessner, C.R., Li, S., Tyndall, S., Getzoff, E.D., Woods, V.L., Jr. PAS domain allostery and light-induced conformational changes in photoactive yellow protein upon I2 intermediate formation, probed with enhanced hydrogen/deuterium exchange mass spectrometry. J. Mol. Biol. 363:148, 2006. Panda, K., Haque, M.M., Garcin-Hosfield, E.D., Durra, D., Getzoff, E.D., Stuehr, D.J. Surface charge interactions of the FMN module governs catalysis by nitricoxide synthase. J. Biol. Chem., in press. Stroupe, M.E., Getzoff, E.D. The role of siroheme in sulfite and nitrite reductases. In: Tetrapyrroles. Warren, M.J., Smith, A. (Eds.). Landes Bioscience, Georgetown, TX, in press. Tiso, M., Konas, D.W., Panda, K., Garcin, E.D., Sharma, M., Getzoff, E.D., Stuehr, D.J. C-terminal tail residue Arg1400 enables NADPH to regulate electron transfer in neuronal nitric-oxide synthase. J. Biol. Chem. 280:39208, 2005. Wood, T.I., Barondeau, D.P., Hitomi, C., Kassmann, C.J., Tainer, J.A., Getzoff, E.D. Defining the role of arginine 96 in green fluorescent protein fluorophore biosynthesis. Biochemistry 44:16211, 2005. Structural Biology of Molecular Interactions and Design J.A. Tainer, A.S. Arvai, D.P. Barondeau, M. Bjoras, B.R. Chapados, L. Craig, T.H. Cross, L. Fan, C. Hitomi, K. Hitomi, J.L. Huffman, C.J. Kassmann, I. Li, G. Moncalian, M.E. Pique, D.S. Shin, O. Sundheim, R.S. Williams, T.I. Wood, A. Yamagata ur studies reveal overall themes and common relationships for fundamental principles and processes of protein regulators and effectors of DNA damage responses, reactive oxygen species, and pathogenesis. We combine x-ray crystallography and solution small-angle x-ray scattering methods, often at our advanced synchrotron facility SIBLYS, to gain a clear view of the structural chemistry that drives biology. Further fusing these techniques with electron microscopy, we bridge the size gaps between high-resolution macromolecular structures and lower resolution multiprotein machine complexes. We then investigate the associated dynamic reversible interactions within cells, O THE SCRIPPS RESEARCH INSTITUTE 167 potential for structure-based design of inhibitors relevant to the development of novel therapeutic agents and chemical tools, and structural implications by biochemistry and mutagenesis. For DNA repair, we collaborate with P. Russell and N. Boddy, Department of Molecular Biology, to couple our structures with genetics and phenotypes. For protein design, we collaborate with E. Getzoff, Department of Molecular Biology, to understand and control the formation of self-synthesizing chromophores in green fluorescent protein and its homologs. P R O T E I N M O D I F I C AT I O N S A N D F U N C T I O N The finding that the number of protein-coding genes in the human genome is more than 10-fold lower than the number of proteins found in human cells by the Human Genome Project is surprising. This huge increase in protein diversity must primarily be due to alternative splicing and posttranslational modification of proteins. A particularly important and intriguing posttranslational modification is the spontaneous peptide backbone cyclization and oxidation chemistry required to convert 3 amino acids into a fluorophore for the family of green fluorescent proteins. REACTIVE OXYGEN AND XENOBIOTIC CONTROL ENZYMES Superoxide dismutases and nitric oxide synthases are master regulators for reactive oxygen species involved in injury, pathogenesis, aging, and degenerative diseases. We are characterizing the hydrogen-bonding networks that underlie the activity of mitochondrial manganese superoxide dismutases. For human copper, zinc superoxide dismutase, we are probing how single-site mutations cause the neurodegeneration in Lou Gehrig disease or familial amyotrophic lateral sclerosis. For nitric oxide synthases, we are examining the structure and chemistry that control levels of nitric oxide, which acts as an important signal and cytotoxin with implications for inflammatory and neurodegenerative diseases. DNA REPAIR AND GENETIC EVOLUTION All the information for heredity is encoded in DNA molecules that are constantly under attack from sunlight, ionizing radiation, and other environmental carcinogens. Surprisingly, however, most DNA damage is due to chemical reactions and free radicals that arise from normal cellular metabolism that is necessary for life. Thus, paradoxically, life is impossible even in the absence of environmental toxins unless coupled to DNA repair. Mutations that cause defects in DNA repair systems may cause cancer and degenerative diseases associated with aging, but fortunately the mutations can also be exploited for cancer therapy. 168 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE AGING AND THE WRN STRUCTURE Mutation of the DNA repair protein WRN can give rise to Werner syndrome, which is characterized by rapid aging and cancer disorders. We have characterized the structure of the WRN nuclease component (Fig. 1). F i g . 2 . Conserved XPB helicase core and DNA-induced open-to- closed conformational changes. XPB contains 4 conserved functional domains: the damage recognition domain (DRD), 2 helicase domains (HD1 and HD2), and a thumb insert (ThM). The interaction of the helicase with DNA may induce a rotation of about 170° of domain HD2 and ThM to form the closed conformation as observed in the crystal structure of hepatitis C virus (HCV) NS3 helicase bound to a single-stranded DNA. F i g . 1 . Hexameric ring model for the WRN nuclease (WRN exo) component. A, WRN x-ray crystal structures aligned as a ring by homology comparisons. B, DNA processing is altered in the WRN Trp145A mutant. C, Electron density map (3σ, 5σ) of dGMP bound to WRN exo. D, The similar internal and external dimensions of Ku70/80 (left) and the WRN exo hexamer model (right). This component is an editing nuclease resembling those found in DNA polymerases. Furthermore, the editing of DNA ends by the WRN exonuclease is stimulated for broken DNA end joining by the Ku DNA end-binding complex. Our findings suggest how the editing of DNA ends during DNA damage responses can critically affect aging and carcinogenesis. NUCLEOTIDE EXCISION REPAIR Nucleotide excision repair, a critical defense mechanism that removes DNA lesions caused by the mutational effects of sunlight (ultraviolet radiation) and toxic chemicals, is also central to the success of anticancer drugs such as cisplatin. We have focused on understanding the mechanisms of nuclear excision repair for potential improvements in cancer treatment. We determined the crystal structure of an enzyme called xeroderma pigmentosum group B (XPB) helicase (Fig. 2). We found several unexpected functions of XPB helicase in nuclear excision repair. These findings helped us address important questions about the enzyme’s role in DNA transcription and repair. XPB helicase recognizes DNA damage that causes blockages in reading the DNA code and aids initiation of efficient repair. and Neisseria meningitidis. Pili play key roles in surface motility, adhesion, formation of microcolonies and biofilms, natural transformation, and signaling. We are characterizing structures of type IV pilin subunits: the assembled pilus fiber, the pilus membrane protein partners, and the assembly ATPase. Pili induce a calcium influx in host cells that plays a role in pathogenesis by altering endocytic trafficking and lysosome homeostasis in infected cells. Because calcium is a central second messenger that regulates several signal cascades, pilus-induced calcium bursts most likely influence bacterial infectivity in key ways. For infections caused by N meningitidis, these calcium bursts are expected to activate neuronal nitric oxide synthases, resulting in toxic levels of nitric oxide that may in part explain the fatal effects of N meningitidis infections of the brain. PUBLICATIONS Ayala, I., Perry, J.P., Szczepanski, J., Tainer, J.A., Vala, M.T., Nick, H.S., Silverman, D.N. Hydrogen bonding in human manganese superoxide dismutase containing 3-fluorotyrosine. Biophys. J. 89:4171, 2005. Ayala, P., Wilbur, J.S., Wetzler, L.M., Tainer, J.A., Snyder, A., So, M. The pilus and porin of Neisseria gonorrhoeae cooperatively induce Ca2+ transients in infected epithelial cells. Cell. Microbiol. 7:1736, 2005. Barondeau, D.P., Kassmann, C.J., Tainer, J.A., Getzoff, E.D. Understanding GFP posttranslational chemistry: structures of designed variants that achieve backbone fragmentation, hydrolysis, and decarboxylation. J. Am. Chem. Soc. 128:4685, 2006. Barondeau, D.P., Tainer, J.A., Getzoff, E.D. Structural evidence for an enolate intermediate in GFP fluorophore biosynthesis. J. Am. Chem. Soc. 128:3166, 2006. Craig, L., Volkmann, N., Arvai, A.S., Pique, M.E., Yeager, M., Egelman, E.H., Tainer, J.A. Type IV pilus structure by cryo-electron microscopy and crystallography: implications for pilus assembly and functions. Mol Cell. 23:651, 2006. BACTERIAL PILI AND INFECTIOUS DISEASES Type IV pili are essential virulence factors for many gram-negative bacteria, such as Neisseria gonorrhoeae Doi, Y., Katafuchi, A., Fujiwara, Y., Hitomi, K., Tainer, J.A., Ide, H., Iwai, S. Synthesis and characterization of oligonucleotides containing 2′-fluorinated thymidine glycol as inhibitors of the endonuclease III reaction. Nucleic Acids Res. 34:1540, 2006. MOLECULAR BIOLOGY 2006 Fan, L., Arvai, A., Cooper, P.K., Iwai, S., Hanaoka, F., Tainer, J.A. Conserved XPB core structure and motifs for DNA unwinding: implications for pathway selection of transcription or excision repair. Mol. Cell 22:27, 2006. Fan, L., Kim, S., Farr, C.L., Schaefer, K.T., Randolph, K.M., Tainer, J.A., Kaguni, L.S. A novel processive mechanism for DNA synthesis revealed by structure, modeling and mutagenesis of the accessory subunit of human mitochondrial DNA polymerase. J. Mol. Biol. 358:1229, 2006. Fan, L., Perry, J.J.P., Tainer, J.A. Reactive oxygen control and DNA repair structural biology: implications for aging and neuropathology. Neuroscience, in press. Hitomi, K., Iwaia, S., Tainer, J.A. The intricate structural chemistry of base excision repair machinery: implications for DNA damage recognition, removal, and repair. DNA Repair (Amst.), in press. Ivanov, I., Chapados, B.R., McCammon, J.A., Tainer, J.A. Proliferating cell nuclear antigen loaded onto double-stranded DNA: dynamics, minor groove interactions and functional implications. Nucleic Acids Res. in press. Pascal, J.M., Tsodikov, O.V., Hura, G.L., Song, W., Cotner, E.A., Classen, S., Tomkinson, A.E., Tainer, J.A., Ellenberger, T. A flexible interface between DNA ligase and PCNA supports conformational switching and efficient ligation of DNA. Mol. Cell. 24:279-91, 2006. Perry, J.J.P., Yannone, S.M., Holden, L.G., Hitomi, C., Asaithamby, A., Han, S., Cooper, P.K., Chen, D.J., Tainer, J.A. WRN exonuclease structure and molecular mechanism imply an editing role in DNA end processing. Nat. Struct. Mol. Biol. 13:414, 2006. Putnam, C.D., Hura, G.L., Tainer, J.A. Combining x-ray solution and crystal diffraction and scanning force microscopies to characterize reversible macromolecular interactions and conformational states. Q. Rev. Biophys., in press. Putnam, C.D., Tainer, J.A. Protein mimicry of DNA and pathway regulation. DNA Repair (Amst.) 4:1410, 2005. Sundheim, O., Vågbø, C.B., Bjørås, M., de Sousa, M.M.L., Talstad, V., Aas, P.A., Drabløs, F., Krokan, H.E., Tainer, J.A., Slupphaug, G. Human ABH3 structure and key residues for oxidative demethylation to reverse DNA/RNA damage. EMBO J. 25:3389, 2006. Tsutakawa, S., Tainer, J.A. Combined methods of SAXS and crystallography to characterize dynamic protein conformations at atomic resolution. J. Struct. Biol., in press. Wood, T.I., Barondeau, D.P., Hitomi, C., Kassmann, C.J., Tainer, J.A., Getzoff, E.D. Defining the role of arginine 96 in green fluorescent protein fluorophore biosynthesis. Biochemistry 44:16211, 2005. Structural Biology of Integral Membrane Proteins G. Chang, S. Aller, A. Chen, Y. Chen, X. He, A. Karyakin, C.R. Reyes, P. Szewczyk, A. Ward, S. Wada, J. Yu, Y. Yin he structural biology of integral membrane proteins is an exciting frontier. We are interested in 5 areas: (1) the molecular structural basis for lipid and drug transport across the cell membrane by multidrugresistance (MDR) transporters, (2) the high-resolution structure of yeast and mammalian MDR transporters, (3) signal transduction by receptors, (4) the discovery and design of potent MDR reversal agents, and (5) the development of an in vitro cell-free system capable of T THE SCRIPPS RESEARCH INSTITUTE 169 overproducing integral membrane proteins suitable for biophysical study. We use several experimental methods, including detergent/lipid protein biochemistry, 3-dimensional crystallization of integral membrane proteins, protein x-ray crystallography, and functional analysis of transporters. We are addressing the molecular basis of MDR in the treatment of infectious disease and cancer. A major cause of MDR is drug efflux pumps imbedded in the cell membrane. Through our structural studies on MDR transporters, we are gaining insights into the molecular mechanics of translocating amphipathic substrates across the cell membrane and the rational design of powerful inhibitors. We are combining chemistry and biology with structure for the discovery and design of potent MDR reversal agents for cancer chemotherapy in collaboration with M.G. Finn, Department of Chemistry; I. Urbatsch, Texas Tech University Health Sciences Center, Lubbock, Texas; and S. Reutz, Novartis International AG, Basel, Switzerland. In collaboration with M. Saier, University of California, San Diego, and Q. Zhang, Department of Molecular Biology, we are probing the structures and function of bacterial MDR transporters. In a collaboration with R.A. Milligan, Department of Cell Biology, we are using electron cryomicroscopy to visualize the low-resolution structures of our transporters. Recently, we determined the x-ray structure of an MDR transporter called EmrD. EmrD is from the Major Facilitator Superfamily, and it expels amphipathic compounds across the inner membrane of E coli. The structure reveals an interior that is composed mostly of hydrophobic residues, a finding consistent with the role of EmrD in transporting amphipathic molecules. Two long loops extend into the inner leaflet side of the cell membrane. This region can recognize and bind substrate directly from the lipid bilayer. We propose that multisubstrate specificity, binding, and transport are facilitated by these loop regions and the internal cavity. PUBLICATIONS Yin, Y., He, X., Szewczyk, P., Nguyen, T., Chang, G. Structure of the multidrug transporter EmrD from Escherichia coli. Science 312:741, 2006. 170 MOLECULAR BIOLOGY 2006 Structure and Function of Membrane-Bound Enzymes C.D. Stout, H. Heaslet, M. Yamaguchi, V.M.M. Luna, A. Annalora, J. Chartron, V. Sundaresan e focus on the structure and function of membrane-bound enzymes and the development of methods for crystallizing membrane proteins. We study the mechanism of transhydrogenase, a mitochondrial respiratory enzyme complex that couples proton translocation with hydride transfer. We use x-ray crystallography, biochemical and spectroscopic methods, electron microscopy studies in collaboration with M. Yeager, Department of Cell Biology, and nuclear magnetic resonance studies in collaboration with J. Dyson, Department of Molecular Biology. Crystal structures of transhydrogenase soluble domains, alone and in complex, have been determined (Fig. 1). Currently, our pri- W F i g . 1 . Superposition of 3 heterotrimers of transhydrogenase soluble domains observed in cocrystals. The presence of additional copies of the smaller soluble domain (dIII, lower right) in the crystal lattice provides a possible model for the intact enzyme in the membrane. mary effort is to determine the structure of the intact 200-kD enzyme in its membrane-bound configuration. We are developing applications of nanodiscs for biophysical studies of integral membrane proteins in collaboration with P. Dawson, Department of Cell Biology, and S.G. Sligar, University of Illinois, Urbana-Champaign, Illinois. Nanodiscs are composed of phospholipid-binding peptides that self-assemble into discrete, water-soluble, bilayer-containing particles. Integral membrane proteins incorporated into these particles retain their enzymatic activity, are amenable to biochemical assays, and may have superior properties for crystallization in the absence of detergents. Both transhydrogenase and cytochrome ba 3 oxidase have been incorporated into nanodiscs. THE SCRIPPS RESEARCH INSTITUTE In collaboration with J.A. Fee, Department of Molecular Biology, we are studying the mechanism of action of cytochrome ba 3 oxidase, the terminal enzyme of respiration. The high-resolution structure of the enzyme from Thermus thermophilus, crystallized in the presence of a detergent, has been determined. Crystallographic experiments, in concert with mutagenesis and spectroscopy, can be used to visualize intermediates in the reduction of oxygen to water and to define the paths of oxygen molecules and protons into the active site. In collaboration with E.F. Johnson, Department of Molecular Biology; J.R. Halpert, University of Texas Medical Branch, Galveston, Texas; and others, we are characterizing structures of mammalian cytochrome P450s. These membrane-associated enzymes are involved in the biosynthesis of lipophilic hormones and specifically metabolize a remarkable diversity of exogenous compounds and drugs. More than 60 genes for P450 occur in the human genome. High-resolution structures, including substrate and inhibitor complexes, have been determined for the P450s 1A2, 2C5, 2C8, 2C9, 2A6, 2A13, 3A4, and 2B4. For 2B4, 3 structures of the enzyme in markedly different conformations provide insight to substrate binding and membrane insertion. A major effort to determine the basis of HIV resistance to antiviral drugs is ongoing in collaboration with A.J. Olson and J.H. Elder, Department of Molecular Biology; B.E. Torbett, Department of Molecular and Experimental Medicine; and D.E. McRee, ActiveSight, San Diego, California. One aspect of this project entails determining the crystal structure of HIV protease-resistant mutants in complex with a wide range of inhibitors. Additional research projects involve crystallographic collaborations on iron-sulfur enzymes, with K.S. Carroll, University of Michigan, Ann Arbor, Michigan; electron transfer proteins, with J.A. Fee, Department of Molecular Biology; and synthetic self-assembling peptides, with M.R. Ghadiri, Department of Chemistry. PUBLICATIONS Chartron, J., Carroll, K.S., Shiau, C., Gao, H., Leary, J.A., Bertozzi, C.R., Stout, C.D. Substrate recognition, protein dynamics, and novel iron-sulfur cluster in Pseudomonas aeruginosa adenosine 5′-phosphosulfate reductase. J. Mol. Biol. 364:152, 2006. Heaslet, H., Kutilek, V., Morris, G.M., Lin, Y.-C., Elder, J.H., Torbett, B.E., Stout, C.D. Structural insights into the mechanisms of drug resistance in HIV-1 protease NL4-3. J. Mol. Biol. 356:967, 2006. Hillier, B.J., Sundaresan, V., Stout, C.D., Vacquier, V.D. Expression, purification, crystallization and preliminary x-ray analysis of the olfactomedin domain from the sea urchin cell-adhesion protein amassin. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 62(Pt. 1):16, 2006. Johnson, E.F., Stout, C.D. Structural diversity of human xenobiotic-metabolizing cytochrome P450 monooxygenases. Biochem. Biophys. Res. Commun. 338:331, 2005. MOLECULAR BIOLOGY 2006 Yadav, M.K., Leman, L.J., Price, D.J., Brooks, C.L. III, Stout, C.D., Ghadiri, M.R. Coiled coils at the edge of configurational heterogeneity: structural analyses of parallel and antiparallel homotetrameric coiled coils reveal configurational sensitivity to a single solvent-exposed amino acid substitution. Biochemistry 45:4463, 2006. Zhao, Y., White, M.A., Muralidhara, B.K., Sun, L., Halpert, J.R., Stout, C.D. Structure of microsomal cytochrome P450 2B4 complexed with the antifungal drug bifonazole: insight into P450 conformational plasticity and membrane interaction. J. Biol. Chem. 281:5973, 2006. Cytochrome ba3 From Thermus thermophilus: New Windows on the Mechanisms of Energy Transduction by Cytochrome c Oxidases J.A. Fee, Y. Chen relatively small integral-membrane protein containing 2 iron and 3 copper atoms distributed in 3 redox active sites generates approximately one third of a human’s metabolic energy. That enzyme is cytochrome c oxidase, and its mechanism of action remains a mystery. The enzyme was first recognized by Charles MacMunn as “histohaematin” in the 1880s and was studied intensely by Otto Warburg as “atmungsferment” during the 1920s and 1930s and by David Keilin as “cytochrome” into the late 1950s. Today, cytochrome c oxidase is still the subject of an international effort. Cytochrome c oxidase catalyzes the following deceptively simple reaction: A 4 cytochrome c2+ + O2 + 8 H+in → 4 cytochrome c3+ + 2 H 2O + 4 H +out, where the subscripts in and out refer, respectively, to matrix and the intermembrane space of the mitochondrion or, in bacteria, to the cytoplasm and the periplasmic space. The free energy of dioxygen reduction is thus captured as a proton gradient; the out side is positive and the in side is negative. During the past 5 decades, enormous progress has been realized in understanding the chemical properties of the 3 redox centers, and the enzyme from several different sources has been crystalized and its structure determined at resolutions ranging from about 3 to 1.8 Å. Moreover, much has been learned about the pathways of electron transfer within the enzyme and the THE SCRIPPS RESEARCH INSTITUTE 171 detailed mechanisms whereby the oxygen molecule is reduced to 2 water molecules. The outstanding questions pertain to the flow of protons into the enzyme from its in side, across the hydrophobic core of the membrane, to exit on its out side. The mystery lies in how all this scalar chemistry comes together to “pump” 4 protons across the membrane. Although the enzyme has been examined by using virtually every available spectroscopic technique, no one has addressed directly the pathways of those protons becoming either water or part of the proton gradient. Much of the past work was done with enzymes in a single clade typified by the mitochondrial enzyme (derived from an ancient bacterium) and with enzymes isolated from common bacteria, notably Rhodobacter sphaeroides, Paraccocus denitrificans, and Escherichia coli (the quinol oxidase). The enzymes from these sources are highly similar in amino acid sequence, 3-dimensional structure, electron-transfer paths, mechanism of oxygen reduction, and, most likely, mechanisms of proton pumping. Our research is based on a 1988 description of a highly sequence-divergent form of the enzyme from Thermus thermophilus, cytochrome ba3, that represents a distinct clade of enzymes widely distributed among archaebacteria. Respectively, these clades represent A- and B-type oxidases. We recently developed a homologous expression system for cytochrome ba 3. This system allows easy purification of the enzyme in amounts of 2–3 mg/L of culture medium by using an N-terminal heptahistidine tag on subunit I. The recombinant enzyme is equally active with native, wild-type protein, and the results of a 2.3-Å x-ray structure determination, done in collaboration with C.D. Stout, Department of Molecular Biology, revealed the expected details at full occupancy and at least one notable surprise. All the A-type oxidases have a glutamic acid residue within the hydrophobic interior of the enzyme that is thought to be at the “end” of the D pathway of proton transport and close to the Fe a3 -Cu B site of dioxygen reduction (Fig. 1A). Mutation of this residue to, for example, glutamine blocks all but a small percentage of the enzyme’s electron-transfer activity, and infrared studies indicate that this residue “senses” changes in the chemical structure of the dioxygen reduction site. Indeed, scientists think that glutamate 286 actually donates the pumped proton to the exit part of the molecule, becoming deprotonated with a pK a of about 9.4. However, no direct evidence exists for this notion, and because 172 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE Hunsicker-Wang, L.M., Pacoma, R.L., Chen, Y., Fee, J.A., Stout, C.D. A novel cryoprotection scheme for enhancing the diffraction of crystals of recombinant cytochrome ba3 oxidase from Thermus thermophilus. Acta Crystallogr. D Biol. Crystallogr. 61(Pt. 3):340, 2005. Developing Reagents for Stabilization of Membrane Proteins Q. Zhang, M.G. Finn,* X. Ma, R.S. Roy * Department of Chemistry, Scripps Research ntegral membrane proteins are extremely unstable outside the hydrophobic membrane bilayer, a situation that makes their in vitro biophysical and structural characterization difficult. An artificial environment is therefore needed to stabilize the proteins in their native state. We focus on developing new membranesimulating reagents for the stabilization of membrane proteins for structural and functional studies. Detergents, structurally similar to cell lipids, selfassemble into micellar structures and are indispensable in dissolving integral membrane proteins into single particles to facilitate protein crystallization. We intend to incorporate more hydrophobicity in the interior of detergent micelles to improve their stability and consequently their ability to stabilize integral membrane proteins. This change is accomplished by appending branches along the alkyl chains of detergents and, most interestingly, by adding a short branch at the interface between the hydrophobic tail and the hydrophilic head. These branches may behave in 2 distinct ways like small amphiphile additives successfully used in crystallization of integral membrane proteins, thereby decreasing the micellar radius and extruding water from the hydrophobic core of the micelles. We are also working on a unique class of molecules with facial amphiphilicity. The facial amphiphiles are structurally distinct from the classical detergents that have end polarity. Although not clear, the binding mode with integral membrane proteins by the facial amphiphiles should differ from that of classical detergents. A smaller protein–facial amphiphile complex may be formed because of the amphiphile’s small aggregation number, which is expected to be beneficial for obtaining well-ordered protein crystals. We have shown that our newly designed facial amphiphiles can maintain the full catalytic function of an ATP-binding cassette transporter protein. I F i g . 1 . Cross-eyed stereo view of the Fe a3-Cu B binuclear activesite structures (leftmost Fe and Cu) of the cytochrome aa3 from R sphaeroides (A) and the cytochrome ba 3 from T thermophilus (B), emphasizing the overlapping position of the glutamate 286 (Glu238) in R sphaeroides and the isoleucine 235 (Ile-235) in T thermophilus. Under the influence of oxygen reduction, protons most likely enter the protein from the upper left of the figure and exit at the lower right. glutamate 286 resides in a highly hydrophobic region of the structure, its pKa most likely is much higher. The surprise in the structure of cytochrome ba 3 is that an isoleucine residue is isopositional with glutamate 286 as isoleucine 235, as shown in Fig. 1B. We mutated this residue to both a glutamine and a glutamate residue in the cytochrome with no apparent loss of electron-transfer activity. To determine if the substituted glutamate residue can be used to monitor changes in the Fea3-Cu B pair, as it does in the A-type oxidases, we have initiated an infrared study of these mutant proteins in collaboration with R. Gennis, University of Illinois, Urbana-Champaign, Illinois, and J. Heberle, Jülich Research Center, Jülich, Germany. How these studies will advance our understanding of proton-pumping mechanisms remains unclear. What is clear is that cytochrome ba 3 provides new openings to explore the mechanism of the cytochrome oxidases. PUBLICATIONS Chen, Y., Hunsicker-Wang, L.M., Pacoma, R.L., Luna, E., Fee, J.A. A homologous expression system for obtaining engineered cytochrome ba3 from Thermus thermophilus HB8. Protein Expr. Purif. 40:299, 2005. Farver, O., Chem, Y., Fee, J.A., Pecht, I. Electron transfer among the CuA-, heme b- and a3-centers of Thermus thermophilus cytochrome ba3. FEBS Lett. 580:3417, 2006. MOLECULAR BIOLOGY 2006 The structural determination of integral membrane proteins with our synthesized amphiphiles is being investigated in collaboration with members of the Center for Innovative Membrane Protein Technologies of the Joint Center for Structural Genomics at Scripps Research. PUBLICATIONS Bosco, D.A., Fowler, D.M., Zhang, Q., Nieva, J., Powers, E.T., Wentworth, P., Jr., Lerner, R.A., Kelly, J.W. Elevated levels of oxidized cholesterol metabolites in Lewy body disease brains accelerate α-synuclein fibrillization [published correction appears in Nat. Chem. Biol. 2:346, 2006]. Nat. Chem. Biol. 2:249, 2006. Structural Neurobiology and Development of Protein Therapeutic Agents R.C. Stevens, E.E. Abola, A.I. Alexandrov, H.M. Archer, J.W. Arndt, G.A. Asmar-Rovira, R.R. Benoit, M.H. Bracey, A. Brooun, Q. Chai, V.G. Cherezov, E. Chien, A. Gámez, M.T. Griffith, C. Grittini, M.A. Hanson, V.-P. Jaakola, J. Joseph, K. Masuda, M. Mileni, K. Moy, M. Nelson, C. Roth, K. Saikatendu, V. Subramanian, J. Velasquez, L. Wang, M.K. Yadav HIGH-THROUGHPUT STRUCTURAL BIOLOGY ut of frustration with the rate at which information on structural biology became known in the past, we focused on developing new tools to change the field of structural biology by accelerating the rate of determination of protein structures. This endeavor included pioneering microliter expression/purification for structural studies, nanovolume crystallization, automated collection of images, and synchrotron beam line automation. These technologies were initially tested by staff at the Joint Center for Structural Genomics (htpp://www.jcsg.org), where the power of the new tools was demonstrated. Although the Joint Center for Structural Genomics 2 has continued as a successful second-phase structural genomics production center, in collaboration with P. Kuhn, Department of Cell Biology, we have created 2 new technology-focused centers funded by the National Institutes of Health. The first center is the Joint Center for Innovative Membrane Protein Technologies (http://jcimpt.scripps.edu). Here, in collaboration with K. Wüthrich, Q. Zhang, and G. Chang, Department of Molecular Biology; M.G. Finn, Department of Chemistry; and P. Kuhn and M. Yeager, Department of Cell Biology, we do research exclusively on eukaryotic and prokaryotic membrane proteins. The O THE SCRIPPS RESEARCH INSTITUTE 173 second center is the Accelerated Technologies Center for Gene to 3D Structure (http://www.atcg3d.org). Here we are doing collaborative work with Dr. Kuhn and with researchers from deCODE biostructures, Bainbridge Island, Washington; Lyncean Technologies, Palo Alto, California; and the University of Chicago, Chicago, Illinois. In 2005, scientists at the centers showed that high-resolution electron density maps and refined models can be obtained from in situ diffraction of crystals grown in microcapillaries. In 2007, the first laboratory-sized synchrotron will be installed at Scripps Research. The synchrotron has performance characteristics comparable to those of a synchrotron beam line in terms of intensity and tunability and will enable us to use direct diffraction analysis of ongoing in situ crystallization experiments to accelerate the determination of macromolecular structures. STRUCTURAL NEUROBIOLOGY Although we have developed high-throughput methods to accelerate the determination of protein structures, our primary interest is using these tools to study the chemistry and biology of neurotransmission and of diseases that affect neurons, particularly childhood neurologic disorders. Our goals are to understand how neuronal cells function on a molecular level and, on the basis of that understanding, create new molecules and materials that mimic neuronal signal transduction and recognition. BIOSYNTHESIS OF NEUROTRANSMITTERS For neuronal signal transduction, the presynaptic cell synthesizes neurotransmitters that then traverse the synaptic cleft. We are using the high-throughput methods to determine the inclusive structures of complete biochemical pathways. Specifically, we are interested in determining the structures of all the enzymes in the biosynthesis pathways of neurotransmitters in order to understand the mechanistic details of each individual enzymatic reaction at the atomic level. This approach also allows us to determine the best path of drug discovery for the biosynthesis of neurotransmitters. T H E R A P E U T I C A G E N T S F O R T R E AT M E N T O F PHENYLKETONURIA In addition to the basic hydroxylase enzymology questions under investigation, recent clinical studies suggest that some patients with the metabolic disease phenylketonuria are responsive to (6R)-L-erythro-5,6,7,8tetrahydrobiopterin, the natural cofactor of phenylalanine hydroxylase. We are doing studies in collaboration with scientists at BioMarin Pharmaceutical Inc., Novato, 174 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE California, to correlate how structure can be used to predict which patients with phenylketonuria most likely will respond to treatment with this cofactor. Phase 3 clinical trials for the treatment of mild phenylketonuria with the proprietary form of the cofactor, Phenoptin, have been completed. For classical phenylketonuria, we are developing an enzyme replacement therapeutic agent that is being tested in animal models. The therapy is based on administration of a modified form of phenylalanine ammonia lyase discovered in our structural studies (Fig. 1). Last, F i g . 1 . A, Crystal structure of phenylalanine ammonia lyase (PAL) determined at 1.6-Å resolution. This protein structure was engineered and chemically modified as a once-a-week injectable therapeutic agent for treatment of phenylketonuria. B, ENU2 mice are used as a model for phenylketonuria in preclinical studies. C and D, A reduction in phenylalanine and immune response levels occurs in ENU2 mice after the injection of PAL that has been chemically modified (pegylated). These PEG-PAL formulations show promise as therapeutic agents for treatment of phenylketonuria. we are determining the structural basis of diseases caused by several other enzymes involved in the biosynthesis of neurotransmitters. NEUROTOXINS The clostridial neurotoxins include tetanus toxin and the 7 serotypes of botulinum toxin. We are determining the molecular events involved in the binding, pore formation, translocation, and catalysis of botulinum neurotoxin. Although botulinum toxin is most known for its deadly effects, it is now being used therapeutically to treat involuntary muscle disorders such as cerebral palsy and neuromuscular dystonias. Previously, we determined the structure of the 150-kD holotoxin form of the toxin, the holotoxin bound to antibodies, the catalytic domains of several serotypes (A, B, D, F, G), and the catalytic domain bound to substrates and inhibitors (Fig. 2). These structures are being used to under- F i g . 2 . Serotype structures of botulinum neurotoxin (BoNT), its light chain (LC), the closely related tetanus neurotoxin (TeNT), and the crystal structure of 150-kD botulinum neurotoxin A bound to a fragment of a neutralizing monoclonal antibody. stand and redesign the toxin’s mechanism of action and to determine additional therapeutic applications of the toxin. CANNABINOID SIGNALING In collaboration with B.F. Cravatt, Department of Cell Biology, we solved the structure of fatty acid amide hydrolase, a degradative integral membrane enzyme responsible for setting intracellular levels of endocannabinoids, to 2.8 Å. Fatty acid amide hydrolase is intimately associated with CNS signaling processes such as retrograde synaptic transmission, a process that is also modulated by the illicit substance δ-9-tetrahydrocannabinol. With our knowledge of the 3-dimensional structure, we are trying to understand how the enzyme works at a basic level and how it might be the basis for potential drug discovery. PUBLICATIONS Arndt, J.W., Chai, Q., Christian, T., Stevens, R.C. Structure of botulinum neurotoxin type D light chain at 1.65 Å resolution: repercussions for VAMP-2 substrate specificity. Biochemistry 45:3255, 2006. MOLECULAR BIOLOGY 2006 Arndt, J.W., Jacobson, M.J., Abola, E.E., Tepp, W.H., Johnson, E.A., Stevens, R.C. A structural perspective of the sequence variability within botulinum neurotoxin subtypes A1-A4. J. Mol. Biol. 362:733, 2006. Blau, N., Koch, R., Matalon, R., Stevens, R.C. Five years of synergistic scientific effort on phenylketonuria therapeutic development and molecular understanding. Mol. Genet. Metab. 86(Suppl. 1):S1, 2005. Collins, B., Stevens, R.C., Page, R. Crystallization optimum solubility screening: using crystallization results to identify the optimal buffer for protein crystal formation. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 61(Pt. 12):1035, 2005. DiDonato, M., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of a single-stranded DNA-binding protein (TM0604) from Thermotoga maritima at 2.60 Å resolution. Proteins 63:256, 2006. Han, G.W., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of an apo mRNA decapping enzyme (DcpS) from mouse at 1.83 Å resolution. Proteins 60:797, 2005. THE SCRIPPS RESEARCH INSTITUTE 175 Scriver, C.R., Hurtubise, M., Prevost, L., Phommarinh, M., Konecki, D., Erlandsen, H., Stevens, R.C., Waters, P.J., Ryan, S., McDonald, D., Sarkissan, C. A PAH gene knowledge base: content, informatics, utilization. In: PKU and BH4: Advances in Phenylketonuria and Tetrahydrobiopterin Research. Blau, N. (Ed.). SPS Publications, Heilbrun, Germany, 2006, p. 434. Swaminathan, S., Stevens, R.C. Three-dimensional protein structures of botulinum neurotoxin light chains serotypes A, B, and E. In: Treatments from Toxins: The Therapeutic Potential of Clostridial Neurotoxins. Foster, K.A., Hambleton, P., Shone, C.C. (Eds.). CRC Press: Boca Raton, FL, in press. Xu, Q., Schwarzenbacher, R., Krishna, S.S., et al. Crystal structure of acireductone dioxygenase (ARD) from Mus musculus at 2.06 Å resolution. Proteins 64:808, 2006. Xu, Q., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of virulence factor CJ0248 from Campylobacter jejuni at 2.25 Å resolution reveals a new fold. Proteins 62:292, 2006. Yadav, M.K., Gerdts, C.J., Sanishvili, R., Smith, W.W., Roach, L.S., Ismagilov, R.F., Kuhn, P., Stevens, R.C. In situ data collection and structure refinement from microcapillary protein crystallization. J. Appl. Crystallogr. 38:900, 2005. Han, G.W., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of the ApbE protein (TM1553) from Thermotoga maritima at 1.58 Å resolution. Proteins 64:1083, 2006. Jaroszewski, L., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of Hsp33 chaperone (TM1394) from Thermotoga maritima at 2.20 Å resolution. Proteins 61:669, 2005. Jin, K.K., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of TM1367 from Thermotoga maritima at 1.90 Å resolution reveals an atypical member of the cyclophilin (peptidylprolyl isomerase) fold. Proteins 63:1112, 2006. Joseph, J.S., Saikatendu, K.S., Subramanian, V., Neuman, B.W., Brooun, A., Griffith, M., Moy, K., Yadav, M.K., Velazquez, J., Buchmeier, M.J., Stevens, R.C., Kuhn, P. Crystal structure of non-structural protein-10 (nsp10) from the SARS coronavirus reveals a novel fold with two zinc-binding motifs. J. Virol. 80:7894, 2006. Klock, H.E., Schwarzenbacher, R., Xu, Q., et al. Crystal structure of a conserved hypothetical protein (gi: 13879369) from mouse at 1.90 Å resolution reveals a new fold. Proteins 61:1132, 2005. Matalon, R., Michals-Matalon, K., Koch, R., Grady, J., Tyring, S., Stevens, R.C. Response of patients with phenylketonuria in the US to tetrahydrobiopterin. Mol. Genet. Metab. 86(Suppl. 1):S17, 2005. Mathews, I.I., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of phosphoribosylformylglycinamidine synthase II (smPurL) from Thermotoga maritima at 2.15 Å resolution. Proteins 63:1106, 2006. Mathews, I.I., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of phosphoribosylformyl-glycinamidine synthase II, PurS subunit (TM1244) from Thermotoga maritima at 1.90 Å resolution. Proteins 65:249, 2006. Pérez, B., Desviat, L.R., Gómez-Puertas, P., Martínez, A., Stevens, R.C., Ugarte, M. Kinetic and stability analysis of PKU mutations identified in BH4-responsive patients. Mol. Genet. Metab. 86(Suppl. 1):S11, 2005. Peti, W., Page, R., Moy, K., O’Neil-Johnson, M., Wilson, I.A., Stevens, R.C., Wüthrich, K. Towards miniaturization of a structural genomics pipeline using micro-expression and microcoil NMR. J. Struct. Funct. Genomics 6:259, 2005. Ratia, K., Saikatendu, K.S., Santarsiero, B.D., Barretto, N., Baker, S.C., Stevens, R.C., Mesecar, A.D. Severe acute respiratory syndrome coronavirus papain-like protease: structure of a viral deubiquitinating enzyme. Proc. Natl. Acad. Sci. U. S. A. 103:5717, 2006. Saikatendu, K.S., Joseph, J.S., Subramanian, V., Clayton, T., Griffith, M., Moy, K., Velasquez, J., Neuman, B.W., Buchmeier, M.J., Stevens, R.C., Kuhn, P. Structural basis of severe acute respiratory syndrome coronavirus ADP-ribose-1′′-phosphate dephosphorylation by a conserved domain of nsP3. Structure 13:1665, 2005. Schwarzenbacher, R., McMullan, D., Krishna, S.S., et al. Crystal structure of a glycerate kinase (TM1585) from Thermotoga maritima at 2.70 Å resolution reveals a new fold. Proteins 65:243, 2006. High-Throughput Approaches to Protein Structure and Function S.A. Lesley, M. Deller, D. Carlton, H. Johnson, Y. Elias, T. Clayton enomic information from the large number of sequenced species has provided as many questions as it has answered. Evaluating protein structure and function is of primary importance for understanding the basic biology of the cell and is a challenge in the context of the genome. To address this challenge, we have established high-throughput approaches for evaluating structural and functional diversity of proteins. We have developed the capacity to clone, express, purify, and crystallize large numbers of proteins in parallel as part of our structural genomics effort with the Joint Center for Structural Genomics, and we hope to apply these same tools to characterize the molecular basis of the specificity of enzyme substrates. The goals of the Joint Center for Structural Genomics are to develop a high-throughput and cost-effective structure pipeline and to use the pipeline to determine novel protein folds and explore protein structure-function relationships. We have used this approach in the extensive study of the thermophilic bacterium Thermotoga maritima and for targets from mouse and other bacterial genomes. Our technologies have enabled us to perform comprehensive structural studies of these proteomes. To date, these efforts have resulted in more than 300 novel protein structures from the center. Functional studies of selected targets have been performed. For example, in collaboration with A. Kohen, University of Iowa, Iowa City, we explored the mecha- G 176 MOLECULAR BIOLOGY 2006 nism of thymidylate synthase from T maritima. This protein has a novel fold and a unique flavin-dependent biochemical mechanism. The gene for thymidylate synthase is an essential one, and the protein is an important potential antibacterial target because of its structural dissimilarity with the human protein. We have also developed the method of deuterium exchange by mass spectrometry in collaboration with V. Woods, University of California, San Diego, to characterize protein regions with highly flexible regions that interfere with crystallization. Subsequent elimination of these regions dramatically improves crystallization and has resulted in structures for several problematic structures. Expression of membrane proteins continues to be one of the most difficult challenges in studying this important protein class. Our structural genomics efforts in collaboration with S. Eshaghi, Karolinska Institutet, Stockholm, Sweden, have led to the structure of the integral membrane protein CorA, a magnesium transporter from T maritima. In collaboration with P. Schultz, Department of Chemistry, we are exploring the use of unnatural amino acids to enhance the purification and crystallization of integral membrane proteins. THE SCRIPPS RESEARCH INSTITUTE Jin, K.K., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of TM1367 from Thermotoga maritima at 1.90 Å resolution reveals an atypical member of the cyclophilin (peptidylprolyl isomerase) fold. Proteins 63:1112, 2006 Klock, H.E., Schwarzenbacher, R., Xu, Q., et al. Crystal structure of a conserved hypothetical protein (gi: 13879369) from mouse at 1.90 Å resolution reveals a new fold. Proteins 61:1132, 2005. Klock, H.E., White, A., Koesema, E., Lesley, S.A. Methods and results for semiautomated cloning using integrated robotics. J. Struct. Funct. Genomics 6:89, 2005. Kreusch, A., Han, S., Brinker, A., Zhou, V., Choi, H., He, Y., Lesley, S.A., Caldwell, J., Gu, X. Crystal structures of a new class of HSP90 inhibitors, dihydroxyphenylpyrazoles. Bioorg. Med. Chem. Lett. 15:1475, 2005. Kreusch, A., Han, S., Brinker, A., Zhou, V., Choi, H.S., He, Y., Lesley, S.A., Caldwell, J., Gu, X.J. Crystal structures of human HSP90α complexed with dihydroxyphenylpyrazoles. Bioorg. Med. Chem. Lett. 15:1475, 2005. Lesley, S.A., Wilson, I.A. Protein production and crystallization at the Joint Center for Structural Genomics. J. Struct. Funct. Genomics 6:71, 2005. Levin, I., Miller, M.D., Schwarzenbacher, R., et al. Crystal structure of an indigoidine synthase A (IndA)-like protein (TM1464) from Thermotoga maritima at 1.90 Å resolution reveals a new fold. Proteins 59:864, 2005. Mason, A., Agrawal, N., Washington, M.T., Lesley, S.A., Kohen, A. A lag-phase in the reduction of flavin dependent thymidylate synthase (FDTS) revealed a mechanistic missing link. Chem. Commun. (Camb.) 1781, 2006, Issue 16. Mathews, I., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of S-adenosylmethionine:tRNA ribosyltransferase-isomerase (QueA) from Thermotoga maritima at 2.0 Å resolution reveals a new fold. Proteins 59:869,2005. Mathews, I.I., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of phosphoribosylformylglycinamidine synthase II (smPurL) from Thermotoga maritima at 2.15 Å resolution. Proteins 63:1106, 2006. PUBLICATIONS Arndt, J.W., Schwarzenbacher, R., Page, R., et al. Crystal structure of an α/β serine hydrolase (YDR428C) from Saccharomyces cerevisiae at 1.85 Å resolution. Proteins 58:755, 2005. McMullan, D., Canaves, J.M., Quijano, K., Abdubek, P., Nigoghossian, E., Haugen, J., Klock, H.E., Vincent, J., Hale, J., Paulsen, J., Lesley, S.A. High-throughput protein production for x-ray crystallography and use of size-exclusion chromatography to validate computational biological unit predictions. J. Struct. Funct. Genomics 6:135, 2005. Chamberlain, P.P., Sandberg, M.L., Sauer, K., Cooke, M.P., Lesley, S.A., Spraggon, G. Structural insights into enzyme regulation for inositol 1,4,5-trisphosphate 3kinase B. Biochemistry 44:14486, 2005. Page, R., Deacon, A.M., Lesley, S.A., Stevens, R.C. Shotgun crystallization strategy for structural genomics, II: crystallization conditions that produce high resolution structure for T maritima proteins. J. Struct. Funct. Genomics 6:209, 2005. Columbus, L., Lipfert, J., Klock, H., Millett, I., Doniach, S., Lesley, S.A. Expression, purification, and characterization of Thermotoga maritima membrane proteins for structure determination. Protein Sci. 15:961, 2006. Rife, C., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of the global regulatory protein CsrA from Pseudomonas putida at 2.05 Å resolution reveals a new fold. Proteins 61:449, 2005. DiDonato, M., Krishna, S.S., Schwarzenbacher, R., et al. Crystal structure of a single-stranded DNA-binding protein (TM0604) from Thermotoga maritima at 2.60 Å resolution. Proteins 63:256, 2006. Rife, C., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of a putative modulator of DNA gyrase (pmbA) from Thermotoga maritima at 1.95 Å resolution reveals a new fold. Proteins 61:444, 2005. Eshaghi, S., Niegowski, D., Kohl, A., Martinez Molina, D., Lesley, S.A., Nordlund, P. Crystal structure of a divalent metal ion transporter CorA at 2.9 Å resolution [published correction appears in Science 313:1389, 2006]. Science 313:354, 2006. Wang, Y., Klock, H., Yin, H., Wolff, K., Bieza, K., Niswonger, K., Matzen, J., Gunderson, D., Hale, J., Lesley, S., Kuhen, K., Caldwell, J., Brinker, A. Homogeneous high-throughput screening assays for HIV-1 integrase 3β-processing and strand transfer activities. J. Biomol. Screen. 10:456, 2005. Han, G.W., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of an apo mRNA decapping enzyme (DcpS) from mouse at 1.83 Å resolution. Proteins 60:797, 2005. Han, G.W., Schwarzenbacher, R., Page, R., et al. Crystal structure of an alanineglyoxylate aminotransferase from Anabaena sp. at 1.70 Å resolution reveals a noncovalently linked PLP cofactor. Proteins 58:971, 2005. Han, S., Zhou, V., Pan, S., Liu, Y., Hornsby, M., McMullan, D., Klock, H., Lesley, S.A., Gray, N., Caldwell, J., Gu, X.J. Identification of coumarin derivatives as a novel class of allosteric MEK1 inhibitors. Bioorg. Med. Chem. Lett. 15:5467, 2005. Jaroszewski, L., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of Hsp33 chaperone (TM1394) from Thermotoga maritima at 2.20 Å resolution. Proteins 61:669, 2005. Xu, Q., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of a formiminotetrahydrofolate cyclodeaminase (TM1560) from Thermotoga maritima at 2.80 Å resolution reveals a new fold. Proteins 58:976, 2005. Xu, Q., Schwarzenbacher, R., McMullan, D., et al. Crystal structure of virulence factor CJ0248 from Campylobacter jejuni at 2.25 Å resolution reveals a new fold. Proteins 62:292, 2006. MOLECULAR BIOLOGY 2006 Nuclear Magnetic Resonance Spectroscopy in Protein Structural Biology and Structural Genomics M. Almeida, W. Augustyniak, L. Columbus, M. Geralt, R. Horst, M. Johnson, B. Pedrini, W.J. Placzek, P. Serrano, K. Wüthrich ur research program focuses on 2 areas. First, in a collaboration with A. Horwich, Yale University, New Haven, Connecticut, who is a visiting scientist at Scripps Research, we are investigating structural and mechanistic aspects of the function of GroEtype chaperonin systems in Escherichia coli. This research concerns the process of protein folding in healthy and diseased organisms and thus is directly related to the currently extensively discussed protein misfolding diseases. Because of the large size of the GroE-type supramolecular structures, this project depends on continuous improvement of existing solution nuclear magnetic resonance (NMR) techniques and development of new techniques. Second, we develop and apply NMR methods for use in structural genomics. We participate in the Joint Center for Structural Genomics (JCSG), the JCSG Center for Innovative Membrane Protein Technologies, and the Consortium for Functional and Structural Proteomics of SARS-CoV–Related Proteins (FSPS). On the one hand, we explore the use of automated microscale NMR equipment for the screening of recombinant protein preparations for folded globular domains. On the other hand, we use NMR spectroscopy to determine the structures of selected proteins from the proteomes under study. The following sections highlight our research on proteins from the severe acute respiratory syndrome–associated coronavirus (SARS-CoV) proteome, which is pursued under the auspices of the FSPS (http://sars.scripps.edu) and the JCSG (http://www.jcsg.org). O THE SARS-COV PROBLEM In 2003, a major global outbreak of SARS was caused by a newly emerged coronavirus. The coronavirus genome is composed of a single plus-strand RNA of about 30 kb and is the largest genome among known RNA viruses. About two thirds of the genome is devoted to encoding the replicase polyprotein, which is cleaved by viral proteases to release the mature THE SCRIPPS RESEARCH INSTITUTE 177 nonstructural proteins that perform enzymatic functions of the virus within the host cell. These functions include RNA-processing steps and other functions that are currently unknown. The genome also encodes viral structural proteins, which form part of the mature viral particle along with genomic RNA. Several of the replicase proteins of coronaviruses have little or no apparent relationship to other known proteins, and little is known about how they function during viral infection. In addition, the reasons for the severe signs and symptoms caused by SARS-CoV in comparison with other human coronaviruses, which usually cause much less severe infections, are currently unknown. We are using NMR spectroscopy for structural and functional investigations of SARS-CoV proteins to gain information about the viral life cycle and to identify possible new antiviral strategies. NONSTRUCTURAL PROTEIN 1 Nonstructural protein 1 (nsp1) is the leader protein of the SARS-CoV genome and the first to be translated and cleaved by the viral protease to its mature form. It has little apparent relationship to proteins of other coronaviruses and may perform a function unique to SARS-CoV. We used NMR spectroscopy to investigate the solution structure and dynamics of nsp1. The protein adopts a new 3-dimensional fold, with a distorted, 6-stranded β-barrel covered by an α-helix. This stable, folded globular domain carries a long, flexibly disordered polypeptide “tail” at the C terminus. We used bioinformatics techniques to search for local structural features that might provide insight into the functional properties of this protein. We detected a possible protease active site on one end of the β-barrel, indicating that nsp1 may be a previously unrecognized viral protease. Follow-up studies indicated that formation of a functional active site may require the presence of the long C-terminal tail, or of other protein cofactors. NONSTRUCTURAL PROTEIN 3 The viral element nsp3 is a large protein of about 2000 amino acid residues that most likely includes multiple functional domains. We designed smaller constructs of this protein encompassing predicted individual domains, and used 1-dimensional 1H NMR screening to identify those domains that were independently folded. We then determined the solution structure of the N-terminal domain, nsp3a. Unexpectedly, we found that its structure is similar to that of the α/β roll fold of ubiquitin. This structural motif is most commonly found in proteins of eukaryotes that are involved in cellular sig- 178 MOLECULAR BIOLOGY 2006 naling pathways. Therefore, the nsp3a domain may be used by the virus to interact with signaling proteins of the host cell and to interfere with cellular pathways in order to increase virulence. We are also investigating a possible second function of this protein in RNA processing. Using NMR spectroscopy and mass spectrometry, we identified RNA molecules that bind to nsp3a. We are studying these interactions to determine possible enzymatic or scaffolding functions. THE SCRIPPS RESEARCH INSTITUTE PUBLICATIONS Almeida, M.S., Herrmann, T., Peti, W., Wilson, I.A., Wüthrich, K. NMR structure of the conserved hypothetical protein TM0487 from Thermotoga maritima: implications for 216 homologous DUF59 proteins. Protein Sci. 14:2880, 2005. Columbus, L., Peti, W., Etezady-Esfarjani, T., Herrmann, T., Wüthrich, K. NMR structure determination of the conserved hypothetical protein TM1816 from Thermotoga maritima. Proteins 60:552, 2005. Horst, R., Bertelsen, E.B., Fiaux, J., Wider, G., Horwich, A.L., Wüthrich, K. Direct NMR observation of a substrate protein bound to the chaperonin GroEL. Proc. Natl. Acad. Sci. U. S. A. 102:12748, 2005. Peti, W., Herrmann, T., Zagnitko, O., Grzechnik, S.K., Wüthrich, K. NMR structure of the conserved hypothetical protein TM0979 from Thermotoga maritima. Proteins 59:387, 2005. NONSTRUCTURAL PROTEIN 7 The viral component nsp7 is highly conserved between the different coronaviruses and probably performs an essential core function in this virus family. Interestingly, the solution structure of nsp7 also shows a new fold that was not previously observed in any known protein structure. The structure consists of 4 helices, and although many 4-helix bundle proteins are known, nsp7 does not form a bundle. Rather 3 helices are assembled into a flat sheet, with the helices antiparallel, and the fourth helix is stacked across one side of this sheet (Fig. 1). The other surface of the flat sheet Peti, W., Johnson, M.A., Herrmann, T., Neuman, B.W., Buchmeier, M.J., Nelson, M., Joseph, J., Page, R., Stevens, R.C., Kuhn, P., Wüthrich, K. Structural genomics of the severe acute respiratory syndrome coronavirus: nuclear magnetic resonance structure of the protein nsp7. J. Virol. 79:12905, 2005. Peti, W., Page, R., Moy, K., O’Neil-Johnson, M., Wilson, I.A., Stevens, R.C., Wüthrich, K. Towards miniaturization of a structural genomics pipeline using micro-expression and microcoil NMR. J. Struct. Funct. Genomics 6:259, 2005. Nuclear Magnetic Resonance of 3-Dimensional Structure and Dynamics of Proteins in Solution P.E. Wright, H.J. Dyson, R. Burge, J. Ferreon, N. Greenman, T.-H. Huang, B.B. Koehntop, M. Kostic, B. Lee, C.W. Lee, M. Landes, M. Martinez-Yamout, T. Nishikawa, K. Sugase, J. Wojciak, M. Zeeb, E. Manlapaz, L.L. Tennant, J. Chung, D.A. Case, J. Gottesfeld, R. Evans,* M. Montminy* * Salk Institute, La Jolla, California F i g . 1 . Ensemble of 20 conformers representing the polypeptide backbone in the solution structure of SARS-CoV nsp7. The 3 helices α2, α3, and α4 assemble into a flat sheet, with the α1 helix stacked diagonally across one surface of this sheet. Reprinted with permission from Peti, W., et al. J. Virol. 79:12905, 2005. e use multidimensional nuclear magnetic resonance (NMR) spectroscopy to investigate the structures, dynamics, and interactions of proteins in solution. Such studies are essential for understanding the mechanisms of action of these proteins and for elucidating structure-function relationships. The focus of our current research is protein-protein and protein–nucleic acid interactions involved in the regulation of gene expression. Copyright 2005 American Society for Microbiology. T R A N S C R I P T I O N FA C T O R – N U C L E I C A C I D C O M P L E X E S contains hydrophobic and negatively charged patches, which most likely are sites for protein-protein interactions. Currently, we are using NMR spectroscopy to identify interactions with other SARS-CoV proteins. Such interactions could be targeted for the design of inhibitory molecules with antiviral activity. NMR methods are being used to determine the 3-dimensional structures and intramolecular dynamics of zinc finger motifs from several eukaryotic transcriptional regulatory proteins, both free and complexed with target nucleic acid. Zinc fingers are among the most abundant domains in eukaryotic genomes. They play a central role in the regulation of gene expression at both W MOLECULAR BIOLOGY 2006 the transcriptional and the posttranscriptional level, mediated through their interactions with DNA, RNA, or protein components of the transcriptional machinery. The C2H2 zinc finger, first identified in transcription factor IIIA (TFIIIA), is used by numerous transcription factors to achieve sequence-specific recognition of DNA. There is growing evidence, however, that some C2H2 zinc finger proteins control gene expression both through their interactions with DNA regulatory elements and, at the posttranscriptional level, by binding to RNA. The best-characterized example of a C2H 2 zinc finger protein that binds specifically to both DNA and to RNA is TFIIIA, which contains 9 zinc fingers. We showed previously that different subsets of zinc fingers are responsible for high-affinity binding of TFIIIA to DNA (fingers 1–3) and to 5S RNA (fingers 4–6). To obtain insights into the mechanism by which the TFIIIA zinc fingers recognize both DNA and RNA, we have used NMR methods to determine the structures of the complex formed by zf1-3 (a protein containing fingers 1–3) with DNA and by zf4-6 (a protein consisting of fingers 4–6) with a fragment of 5S RNA. Three-dimensional structures were determined previously for the complex of zf1-3 with the cognate 15-bp oligonucleotide duplex. The structures contain several novel features and reveal that prevailing models of DNA recognition, which assume that zinc fingers are independent modules that contact bases through a limited set of amino acids, are outmoded. In addition to its role in binding to and regulating the 5S RNA gene, TFIIIA also forms a complex with the 5S RNA transcript. NMR structures of the complex formed by zinc fingers 4–6 with a truncated form of 5S RNA have been completed and give important insights into the structural basis for 5S RNA recognition. Finger 4 of the protein recognizes both the structure of the RNA backbone and the specific bases in the loop E motif of the RNA, in a classic lock-and-key interaction. Fingers 5 and 6, with a single residue between them, undergo mutual induced-fit folding with the loop A region of the RNA, which is highly flexible in the absence of the protein. NMR studies of 2 alternate splice variants of the Wilms tumor zinc finger protein are in progress. These proteins differ only through insertion of 3 additional amino acids (the tripeptide lysine-threonine-serine) in the linker between fingers 3 and 4, yet have marked differences in their DNA-binding properties and subcellular localization. 15N relaxation measurements indi- THE SCRIPPS RESEARCH INSTITUTE 179 cate that the insertion increases the flexibility of the linker between fingers 3 and 4 and abrogates binding of the fourth zinc finger to its cognate site in the DNA major groove, thereby modulating DNA-binding activity. The x-ray structure of the DNA complex has been determined, providing insights into the mechanism by which disease-causing mutations interfere with DNA binding. NMR studies of the RNA complex are in progress. We have also determined the structure of a novel zinc finger protein that binds to double-stranded RNA and have begun experiments to define the mechanism of RNA recognition. Several novel zinc binding motifs have recently been identified that mediate gene expression at the posttranscriptional level by regulating mRNA processing and metabolism. Regulatory proteins of the TIS11 family bind specifically, through a pair of novel CCCH zinc fingers, to the adenosine-uridine–rich element in the 3′ untranslated region of short-lived cytokine, growth factor, and proto-oncogene mRNAs and control expression by promoting rapid degradation of the message. We recently determined the NMR structure of the complex formed between the tandem zinc finger domain of TIS11d and its binding site on the adenosine-uridine–rich element. This structure showed sequence-specific recognition of single-stranded RNA through formation of a network of hydrogen bonds between the polypeptide backbone and the Watson-Crick edges of the bases. PROTEIN-PROTEIN INTERACTIONS IN T R A N S C R I P T I O N A L R E G U L AT I O N Transcriptional regulation in eukaryotes relies on protein-protein interactions between DNA-bound factors and coactivators that, in turn, interact with the basal transcription machinery. The transcriptional coactivator CREB-binding protein (CBP) and its homolog p300 play an essential role in cell growth, differentiation, and development. Understanding the molecular mechanisms by which CBP and p300 recognize their various target proteins is of fundamental biomedical importance. CBP and p300 have been implicated in diseases such as leukemia, cancer, and mental retardation and are novel targets for therapeutic intervention. We previously determined the structure of the kinaseinducible activation domain of the transcription factor CREB bound to its target domain (the KIX domain) in CBP. Ongoing work is directed toward mapping the interactions between KIX and the transcriptional activation domains of the proto-oncogene c-Myb and of the mixedlineage leukemia protein. The solution structure of the 180 MOLECULAR BIOLOGY 2006 ternary complex between KIX, c-Myb and the mixedlineage leukemia protein has been completed and provides insights into the structural basis for the ability of the KIX domain to interact simultaneously and allosterically with 2 different effectors. Our work has also provided new understanding of the thermodynamics of the coupled folding and binding processes involved in interaction of KIX with transcriptional activation domains. We are using R 2 relaxation dispersion experiments to elucidate the mechanism by which folding of the kinaseinducible activation domain of CREB is coupled to binding to its KIX target domain. These experiments reveal formation of a transient and largely unfolded encounter complex, which then folds on the surface of the KIX domain to form the helical structure observed in the fully bound state. Recently, we determined the structure of the complex between the hypoxia-inducible factor Hif-1α and the CH1 domain of CBP. The interaction between Hif-1α and CBP/p300 is of major therapeutic interest because of the central role Hif-1α plays in tumor progression and metastasis; disruption of this interaction leads to attenuation of tumor growth. A protein named CITED2 functions as a negative feedback regulator of the hypoxic response by competing with Hif-1α for binding to the CH1 domain of CBP. We determined the structure of the complex formed between CITED2 and the CH1 domain and were able to show that the CH1 domain is folded into a stable 3-dimensional structure even in the absence of binding partners. The intrinsically unstructured Hif-1α and CITED2 domains use partly overlapping surfaces of the CH1 motif to achieve high-affinity binding and compete effectively with each other for CBP/p300. We are continuing to map the multiplicity of interactions between CBP/p300 domains and their numerous biological targets to understand the complex interplay of interactions that mediate key biological processes in health and disease. PUBLICATIONS De Guzman, R.N., Goto, N.K., Dyson, H.J., Wright, P.E. Structural basis for cooperative transcription factor binding to the CBP coactivator. J. Mol. Biol. 355:1005, 2006. Kostic, M., Matt, T., Martinez-Yamout, M.A., Dyson, H.J., Wright, P.E. Solution structure of the Hdm2 C2H2C4 RING, a domain critical for ubiquitination of p53. J. Mol. Biol. 363:433, 2006. Lee, B.M., Xu, J., Clarkson, B.K., Martinez-Yamout, M.A., Dyson, H.J., Case, D.A., Gottesfeld, J.M., Wright, P.E. Induced fit and “lock and key” recognition of 5S RNA by zinc fingers of transcription factor IIIA. J. Mol. Biol. 357:275, 2006. THE SCRIPPS RESEARCH INSTITUTE Folding of Proteins and Protein Fragments P.E. Wright, H.J. Dyson, C. Nishimura, D. Felitsky, Y. Yao, J. Chung, L.L. Tennant, V. Bychkova,* T. Uzawa,** S. Takahashi** * Institute of Protein Research, Puschino, Russia ** Kyoto University, Kyoto, Japan he molecular mechanism by which proteins fold into their 3-dimensional structures remains one of the most important unsolved problems in structural biology. Nuclear magnetic resonance (NMR) spectroscopy is uniquely suited to provide information on the structure of transient intermediates formed during protein folding. Previously, we used NMR methods to show that many peptide fragments of proteins have a tendency to adopt folded conformations in water solution. The presence of transiently populated folded structures, including reverse turns, helices, nascent helices, and hydrophobic clusters, in water solutions of short peptides has important implications for initiation of protein folding. Formation of elements of secondary structure probably plays an important role in the initiation of protein folding by reducing the number of conformations that must be explored by the polypeptide chain and by directing subsequent folding pathways. T A P O M Y O G L O B I N F O L D I N G PAT H WAY A major program in our laboratory is directed toward a structural and mechanistic description of the apomyoglobin folding pathway. Previously, we used quenched-flow pulse-labeling methods in conjunction with 2-dimensional NMR spectroscopy to map the kinetic folding pathway of the wild-type protein. With these methods, we showed that an intermediate in which the A, G, and H helices and part of the B helix adopt hydrogen-bonded secondary structure is formed within 6 milliseconds of the initiation of refolding. Folding then proceeds by stabilization of additional structure in the B helix and in the C and E helices. We are using carefully selected myoglobin mutants and both optical stopped-flow spectroscopy and NMR methods to further probe the kinetic folding pathway. For some of the mutants studied, the changes in amino acid sequence resulted in changes in the folding pathway of the protein. These experiments are providing novel insights into both the local and the long-range interactions that stabilize the kinetic folding intermediate. Of particular importance, long-range interactions have been observed MOLECULAR BIOLOGY 2006 that indicate nativelike packing of some of the helices in the kinetic molten globule intermediate. Apomyoglobin provides a unique opportunity for detailed characterization of the structure and dynamics of a protein-folding intermediate. Conditions were previously identified under which the apomyoglobin molten globule intermediate is sufficiently stable for acquisition of multidimensional heteronuclear NMR spectra. Analysis of 13C and other chemical shifts and measurements of polypeptide dynamics provided unprecedented insights into the structure of this state. The A, G, and H helices and part of the B helix are folded and form the core of the molten globule. This core is stabilized by relatively nonspecific hydrophobic interactions that restrict the motions of the polypeptide chain. Fluctuating helical structure is formed in regions outside the core, although the population of helix is low and the chain retains considerable flexibility. The F helix acts as a gate for heme binding and only adopts stable structure in the fully folded holoprotein. The acid-denatured (unfolded) state of apomyoglobin is an excellent model for the fluctuating local interactions that lead to the transient formation of unstable elements of secondary structure and local hydrophobic clusters during the earliest stages of folding. NMR data indicated substantial formation of helical secondary structure in the acid-denatured state in regions that form the A and H helices in the folded protein and also revealed nonnative structure in the D and E helix regions. Because the A and H regions adopt stabilized helical structure in the earliest detectable folding intermediate, these results lend strong support to folding models in which spontaneous formation of local elements of secondary structure plays a role in initiating formation of the A-[B]-G-H molten globule folding intermediate. In addition to formation of transient helical structure, formation of local hydrophobic clusters has been detected by using 15N relaxation measurements. Significantly, these clusters are formed in regions where the average surface area buried upon folding is large. In contrast to acid-denatured unfolded apomyoglobin, the urea-denatured state is largely devoid of structure, although residual hydrophobic interactions have been detected by using relaxation measurements. We measured residual dipolar couplings for unfolded states of apomyoglobin by using partial alignment in strained polyacrylamide gels. These data provide novel insights into the structure and dynamics of the unfolded polypeptide chain. We have shown that the residual THE SCRIPPS RESEARCH INSTITUTE 181 dipolar couplings arise from the well-known statistical properties of flexible polypeptide chains. Residual dipolar couplings provide valuable insights into the dynamic and conformational propensities of unfolded and partly folded states of proteins and hold great promise for charting the upper reaches of protein-folding landscapes. To probe long-range interactions in unfolded and partially folded states of apomyoglobin, we introduced spin-label probes at several sites throughout the polypeptide chain. These experiments led to the surprising discovery that structures with nativelike topology exist within the ensemble of conformations formed by the acid-denatured state of apomyoglobin. They also indicated that the packing of helices in the molten globule state is similar to that in the native folded protein. The view of protein folding that results from our work on apomyoglobin is one in which collapse of the polypeptide chain to form increasingly compact states leads to progressive accumulation of secondary structure and increasing restriction of fluctuations in the polypeptide backbone. Chain flexibility is greatest at the earliest stages of folding, in which transient elements of secondary structure and local hydrophobic clusters are formed. As the folding protein becomes increasingly compact, backbone motions become more restricted, the hydrophobic core is formed and extended, and nascent elements of secondary structure are progressively stabilized. The ordered tertiary structure characteristic of the native protein, with well-packed side chains and relatively low-amplitude local dynamics, appears to form rather late in folding. We recently introduced a variation on the classic quench-flow technique, which makes use of the capabilities of modern NMR spectrometers and heteronuclear NMR experiments, to study the proteins labeled along the folding pathway in an unfolded state in an aprotic organic solvent. This method allows detection of many more amide proton probes than in the classic method, which required formation of the fully folded protein and the measurement of its NMR spectrum in water solutions. This method is particularly useful in documenting changes in the folding pathway that result in the destabilization of parts of the protein in the molten globule intermediate. We recently showed that selfcompensating mutations designed to change the amino acid sequence such that the average area buried upon folding is significantly changed while the 3-dimensional structure of the final folded state remains the same. These studies showed that the average area buried 182 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE upon folding is an accurate predictor of those parts of the apomyoglobin molecule that will fold first and participate in the molten globule intermediate. Quench-flow hydrogen exchange experiments performed on a series of hydrophobic core mutants indicated that the overall helix-packing topology of the kinetic folding intermediate is like that of the native protein, despite local nonnative interactions in packing of the G and H helices. Finally, using a rapid mixing device, we have reduced the dead time of the kinetic refolding experiments and have shown that a compact helical intermediate is formed within 400 microseconds after initiation of apomyoglobin refolding. FOLDING-UNFOLDING TRANSITIONS IN CELLULAR M E TA B O L I S M Many species of bacteria sense and respond to their own population density by an intricate autoregulatory mechanism known as quorum sensing; the bacteria release extracellular signal molecules, called autoinducers, for cell-cell communication within and between bacterial species. A number of bacteria appear to use quorum sensing for regulation of gene expression in response to fluctuations in cell population density. Processes regulated in this way include symbiosis, virulence, competence, conjugation, production of antibiotics, motility, sporulation, and formation of biofilms. We determined the 3-dimensional solution structure of a complex composed of the N-terminal 171 residues of the quorum-sensing protein SdiA of Escherichia coli and an autoinducer molecule, N-octanoyl-1-homoserine lactone (HSL) (Fig. 1). The SdiA-HSL system shows the “folding switch” behavior associated with quorumsensing factors produced by other bacterial species. In the presence of HSL, the SdiA protein is stable and folded and can be produced in good yields from an E coli expression system. In the absence of the autoinducer, the protein is expressed into inclusion bodies. Samples of the SdiA-HSL complex can be denatured but cannot be refolded in aqueous buffers. The solution structure of the complex provides a likely explanation for this behavior. The autoinducer molecule is tightly bound in a deep pocket in the hydrophobic core and is bounded by specific hydrogen bonds to the side chains of conserved residues. The autoinducer thus forms an integral part of the hydrophobic core of the folded SdiA. F i g . 1 . Folding of protein and protein fragments. Ribbon dia- gram showing the lowest energy structure of the complex between HSL and E coli SdiA. basis of their interactions, but unfolded proteins are impossible to characterize structurally by x-ray crystallography, and spectroscopic methods of all kinds are limited. It is necessary to explore unfolded proteins under conditions that approximate their physiologic milieu: in solution, at physiologic pHs and salt concentrations, and in the presence of specific cofactors. Structural insights will be obtained not only from the delineation of 3-dimensional structures but also from the description of conformational ensembles and of the motions of polypeptide chains under various conditions. To gain new insights into the structural basis for the ability of unfolded and partly folded proteins to function in living systems, we are studying the interactions of “client” proteins and cochaperones with a well-known eukaryotic chaperone, Hsp90. Some of the protein components are much larger than have traditionally been studied by using solution NMR. However, we have designed a set of experiments that will allow us to draw valid conclusions about the extent and role of disorder in Hsp90 interactions. In particular, we will apply techniques recently developed in our laboratory for the analysis of hydrogen-deuterium exchange from unstable partially folded proteins by trapping the 2H-labeled species in the aprotic solvent dimethyl sulfoxide. This powerful new technique will be used to probe the structure, stability, and interactions of client proteins and cochaperones with Hsp90. CHAPERONE–COCHAPERONE–CLIENT PROTEIN INTERACTIONS Understanding the role of unfolded states in cellular processes will require an understanding of the structural PUBLICATIONS Dyson, H.J., Wright, P.E. According to current textbooks, a well-defined threedimensional structure is a prerequisite for the function of the protein: is this correct? IUBMB Life 58:107, 2006. MOLECULAR BIOLOGY 2006 Dyson, H.J., Wright, P.E., Scheraga, H.A. The role of hydrophobic interactions in initiation and propagation of protein folding. Proc. Natl. Acad. Sci. U. S. A. 103:13057, 2006. Kamikubo, Y., Kroon, G., Curriden, S.A., Dyson, H.J., Loskutoff, D.J. The reduced, denatured somatomedin B domain of vitronectin refolds into a stable, biologically active form. Biochemistry 45:3297, 2006. Martinez-Yamout, M.A., Venkitakrishnan, R.P., Preece, N.E., Kroon, G., Wright, P.E., Dyson, H.J. Localization of sites of interaction between p23 and Hsp90 in solution. J. Biol. Chem. 281:14457, 2006. Nishimura, C., Dyson, H.J., Wright, P.E. Identification of native and non-native structure in kinetic folding intermediates of apomyoglobin. J. Mol. Biol. 355:139, 2006. Papadopoulos, E., Oglecka, K., Mäler, L., Jarvet, J., Wright, P.E., Dyson, H.J., Gräslund, A. NMR solution structure of the peptide fragment 1-30, derived from mouse Doppel protein, in DHPC micelles. Biochemistry 45:159, 2006. Yao, Y., Martinez-Yamout, M.A., Dickerson, T.J., Brogan, A.P., Wright, P.E., Dyson, H.J. Structure of the Escherichia coli quorum sensing protein SdiA: activation of the folding switch by acyl homoserine lactones. J. Mol. Biol. 355:262, 2006. Yao, Y., Martinez-Yamout, M.A., Dyson, H.J. Backbone and side chain 1H, 13C and 15N assignments for Escherichia coli SdiA1-171, the autoinducer-binding domain of a quorum sensing protein [letter]. J. Biomol. NMR 31:373, 2005. Nuclear Magnetic Resonance Studies of the Structure and Dynamics of Enzymes H.J. Dyson, P.E. Wright, S.H. Bae, D. Boehr, G. Kroon, M. Martinez-Yamout, N.E. Preece, S.C. Sue, L.M. Tuttle, Y. Yao, L.L. Tennant, J. Chung, C.L. Brooks, S.J. Benkovic,* A. Holmgren,** E.A. Komives*** * Pennsylvania State University, University Park, Pennsylvania ** Karolinska Institutet, Stockholm, Sweden *** University of California, San Diego, California e use site-specific information on structure and dynamics obtained via nuclear magnetic resonance (NMR) to further the understanding of protein function. We focus on the mechanism of enzymes and the relationship between dynamics and function in a number of medically important systems. W DYNAMICS IN ENZYME ACTION Dynamic processes are implicit in the catalytic function of all enzymes. We use state-of-the-art NMR methods to elucidate the dynamic properties of several enzymes. New methods have been developed for analysis of NMR relaxation data for proteins that tumble anisotropically and for analysis of slow timescale motions. Dihydrofolate reductase plays a central role in folate metabolism and is the target enzyme for a number of antibacterial and anticancer agents. 15N relaxation experiments on dihydrofolate reductase from Escherichia coli revealed a rich diversity of backbone dynamical THE SCRIPPS RESEARCH INSTITUTE 183 features for a broad range of timescales (picoseconds to milliseconds). A major focus is on the characterization of all intermediates in the dihydrofolate reductase reaction cycle. We have identified functionally important motions in loops that control access to the active site of dihydrofolate reductase on timescales similar to those of the hydride transfer chemistry and the rate-determining step of product release. These motions differ in amplitude and timescale depending on the presence of substrate and/or cofactor in the active site, priming the nicotinamide ring of the cofactor and the pterin ring of the substrate for hydride transfer. In addition, measurements of the population distribution of aliphatic sidechain rotamers provided evidence for coupled motion of active-site side chains that could enhance the catalytic process. Most recently, we used relaxation dispersion measurements to obtain direct information on microsecondmillisecond timescale motions in dihydrofolate reductase, allowing us to characterize the structures of excited states involved in some of these catalysis-relevant processes. Fluctuations between these states, which involve motions of the nicotinamide ring of the cofactor into and out of the active site, occur on a timescale that is directly relevant to the structural transitions involved in progression through the catalytic cycle (Fig. 1). Dihydrofolate reductase is also the test system for a series of experiments to address the question, If all of the chemistry goes on at the active site, what is the purpose of the rest of the enzyme? We are using chimeric mutants, synthesized by our collaborator S.J. Benkovic, Pennsylvania State University, by using a library approach. The purpose of these experiments is to test the hypothesis that local variations in amino acid sequence, 3-dimensional structure, and polypeptide chain dynamics strongly influence the local interactions that mediate enzyme catalysis and may constitute the essential circumstance that allows enzymes to achieve high turnover rates as well as exquisite specificity in their reactions. A combination of NMR structure and dynamics measurements, single-molecule fluorescence measurements, and analysis of the catalytic steps in these mutant proteins will provide new insights into the role of the protein in enzyme catalysis. S T R U C T U R E A N D D Y N A M I C S O F P R I O N VA R I A N T S Onset of prion diseases is caused by conversion of the cellular prion protein PrPC into an abnormally folded isoform, PrPSc, that has the same primary structure as 184 MOLECULAR BIOLOGY 2006 F i g 1 . Schematic diagram showing the energy landscape of dihydrofolate reductase catalysis. Ground state (larger) and higher energy (smaller) structures of each intermediate in the cycle, modeled on published x-ray structures are shown. For each intermediate in the catalytic cycle, the higher energy conformations detected in the relaxation dispersion experiments resemble the ‘ground-state’ conformations of adjacent intermediates. Rate constants for the interconversion between the complexes, measured by pre–steady state enzyme kinetics at 298 K, pH6 are indicated with gray arrows, while the rates measured in relaxation dispersion experiments are shown with black arrows. From Boehr et al., Science 313:1638, 2006. Reprinted with permission from AAAS. PrPC but a totally different 3-dimensional conformation. The abnormally folded (“scrapie”) form of the protein is associated with several diseases, including scrapie in sheep, bovine spongiform encephalopathy (mad cow disease), and human Creutzfeldt-Jakob disease and other inherited prion diseases. We are gathering information on the mechanism of PrPSc formation that can be obtained from structural and dynamic studies of mutant prion proteins corresponding to inherited prion diseases. Individuals carrying familial mutations such as P102L (P101L in our study) are more susceptible than those without such mutations to prion disease. On the other hand, sheep or humans carrying Q167R and/or Q218K mutations are resistant to scrapie and CreutzfeldtJakob disease, respectively. We are using the proteaseresistant cores of wild-type and mutant mouse prion proteins to study the structural and dynamic basis of PrP C -to-PrP Sc conversion in inherited prion diseases. The core is sufficient to transmit infectivity. DYNAMICS AND THE FUNCTION OF IΚB α It is becoming increasingly clear that the function of many systems in living cells depends not only on the structures of the components but also on their flexibility. Numerous examples exist in which components of THE SCRIPPS RESEARCH INSTITUTE an important biological interaction are unstructured or partly structured. In addition, even those interacting molecules that can be classified as “folded” have areas of mobility. Often, these areas are located precisely in the active site of an enzyme or in the binding site of an interacting molecule. A central molecular interaction in cellular control is the interaction between the nuclear transcription factor NF-κB and its inhibitor IκBα. IκBα consists of a series of ankyrin repeats, which appear to have differential mobility. Using hydrogen-deuterium exchange and mass spectrometry, our collaborator E.A. Komives, University of California, San Diego, found that the second, third, and fourth ankyrin repeats of IκBα are well folded, whereas the fifth and sixth repeats, apparently with exactly the same structure, are highly dynamic. These observations prompt a number of questions: Are the motions inferred from the hydrogen-deuterium mass spectrometry experiments also reflected in the backbone and side-chain dynamics of the protein, as measured by NMR relaxation? Are the motions still present in the IκBα–NF-κB complex? Are they necessary for complex formation, so that if they are damped out, for example, by site-directed mutagenesis at appropriate positions, is the formation of the complex disfavored? To answer these questions, we are doing a series of NMR experiments on IκBα and its complexes with NF-κB. PUBLICATIONS Boehr, D.D., Dyson, H.J., Wright, P.E. An NMR perspective on enzyme dynamics. Chem. Rev. 106:3055, 2006. Boehr, D.D., McElheny, D., Dyson, H.J., Wright, P.E. The dynamic energy landscape of dihydrofolate reductase catalysis. Science 313:1638, 2006. Ring Assemblies Mediating ATP-Dependent Protein Folding and Unfolding A.L. Horwich, W.A. Fenton, E. Chapman, E. Koculi, J. Hinnerwisch arge ring assemblies function in many cellular contexts as compartments within a compartment, where actions can be carried out on a substrate bound in the central space inside an oligomeric ring by a high local concentration of surrounding active sites. Both protein folding and unfolding are carried out in an ATP-dependent fashion by such assemblies (Fig. 1). L MOLECULAR BIOLOGY 2006 F i g . 1 . Protein folding and unfolding by chaperone ring assemblies. In protein folding mediated by the chaperonin GroEL (left), the energy of binding ATP and the cochaperonin GroES is used to produce rigid body movements of a GroEL ring that eject a bound nonnative substrate polypeptide into a GroES-encapsulated central cavity, switched from hydrophobic (shaded) to hydrophilic wall character, where productive folding proceeds. The free energy provided by a set of hydrogen bonds formed between the γ-phosphate of ATP and the nucleotide pocket is critical to producing a power stroke of apical domain movement that can eject the substrate polypeptide into the folding chamber. In contrast, in ClpA-mediated unfolding (right), this chaperone seems to use ATP hydrolysis by its D2 ATPase domain to drive a forceful distalward movement of a loop facing its central channel, exerting mechanical force on a bound protein that is proposed to exert an unfolding action. We are studying the essential double-ring components known as chaperonins that assist protein folding to the native state. We are focusing on the bacterial chaperonin GroEL. We have also been examining an opposite number, an “unfoldase,” the bacterial heat-shock protein 100 ring assembly known as ClpA. During the past year, we continued to investigate the structural correlates of the ATPase cycle of GroEL and the polypeptide-binding mechanisms of ClpA. GroEL STRUCTURE AND ALLOSTERY GroEL is an allosteric and highly cooperative machine with positive cooperativity of ATP binding among the subunits of a ring and negative cooperativity of binding between the 2 rings. A long-standing question has been how the information on nucleotide binding and ATP-vsADP state is transmitted from one ring to the other to produce these changes in affinity that lie at the heart of the chaperonin protein-folding cycle. In collaboration with N.A. Ranson, University of Leeds, Leeds, England, and H.R. Saibil, University of London, London, England, we completed a electron cryomicroscopy reconstruction of the transient complex composed of GroEL, its cochaperonin GroES, and ATP by taking advantage of a mutant form of GroEL (D398A). The mutant form binds ATP normally but hydrolyzes it at about 2% of the normal rate, permitting the complex to be captured by rapid freezing on electron cryomicroscopy grids. By comparing this structure (7.7-Å resolution) THE SCRIPPS RESEARCH INSTITUTE 185 with a similarly obtained GroEL-GroES-ADP structure (8.7-Å resolution), we discovered that the differences occur mainly in the trans (unliganded) ring. Information on the nucleotide state of the cis ring appears to be transmitted through the positioning of helix D, which extends from the nucleotide-binding site to the inter-ring interface, where it interacts with the corresponding helix in the trans ring. ATP binding changes the relationship of these helices from that of an unliganded ring, resulting in a series of changes in the interface and the position of the trans helix that lead to a closing of the nucleotide pocket and a reduction in exposure of the substrate-binding hydrophobic residues in the trans apical domain. Hydrolysis of ATP to ADP reverses these changes, opening both the trans apical domains and the trans nucleotide pocket to permit ligand binding and the formation of a new folding chamber on this ring. T H E T R A J E C T O R Y O F P R O T E I N F O L D I N G AT G r o E L Another question of interest regarding protein folding by GroEL is whether, and how, GroEL affects the folding pathway taken by a substrate protein. Most likely some action beyond simply preventing aggregation is occurring, because some substrate proteins cannot fold spontaneously without GroEL and GroES function. In collaboration with R. Horst and K. Wüthrich, Department of Molecular Biology, we are studying the folding pathways of human dihydrofolate reductase during both spontaneous and GroEL-assisted folding. Using hydrogen-deuterium exchange during refolding of 15N-labeled human dihydrofolate reductase and nuclear magnetic resonance analysis of the final native form, we are examining pathways taken inside and outside the chaperonin. ROLE OF THE N-DOMAINS OF ClpA We and others have shown that the N-domains of ClpA are not required for action on substrates bearing the ssrA degradation tag, but removal of the N-domains slows the unfolding and degradation of such substrates. We explored this observation further and found that whereas binding and unfolding of ssrA-tagged substrate are not affected by removal of the N-domains, removal of the domains results in diminished stability of a complex composed of ClpA and ClpP, a double-ring protease that cooperates with ClpA to degrade certain proteins. Consequently, proteins unfolded and translocated by ClpA interact with the protease component and hence escape degradation. In contrast, substrate proteins bearing a RepA degradation tag appear to be completely dependent on the N-domains for initial binding to ClpA. 186 MOLECULAR BIOLOGY 2006 LOOPS IN ClpA In collaboration with A. Deniz, Department of Molecular Biology, we are examining the role of the recently identified loops in ClpA that mediate ssrA binding and substrate protein translocation and associated unfolding. We have designed a series of cysteine substitution variants of ClpA and ClpP that can be labeled with fluorescent reporters and used in single-molecule experiments designed to observe the putative motion of these loops during the substrate translocation/unfolding cycle of ClpA. Such experiments may enable us to correlate movement with ATP hydrolysis and to determine whether such movement is coordinated among the 6 subunits of a ClpA ring or is random. PUBLICATIONS Hinnerwisch, J., Reid, B.G., Fenton, W.A., Horwich, A.L. Roles of the N-domains of the ClpA unfoldase in binding substrate proteins and in stable complex formation with the ClpP protease. J. Biol. Chem. 280:40838, 2005. Horst, R., Wider, G., Fiaux, J., Bertelsen, E.B., Horwich, A.L., Wüthrich, K. Proton-proton Overhauser NMR spectroscopy with polypeptide chains in large structures. Proc. Natl. Acad. Sci. U. S. A. 103:15445, 2006. Ranson, N.A., Clare, D.K., Farr, G.W., Houldershaw, D., Horwich, A.L., Saibil, H.R. Allosteric signaling of ATP hydrolysis in GroEL-GroES complexes. Nat. Struct. Mol. Biol. 13:147, 2006. Chemical Regulation of Gene Expression D. Alvarez, R. Burnett, C.J. Chou, D. Herman, K. Jenssen, S. Ku, E. Soragni, J. Puckett,* S. Tsai,* M. Farkas,* P.B. Dervan,* J.M. Gottesfeld * California Institute of Technology, Pasadena, California he ability to control gene expression at will has been a longstanding goal in molecular biology and human medicine. We focus on pyrrole-imidazole polyamides, a class of small molecules that can be programmed by chemical synthesis to recognize a wide range of DNA sequences. The following is a summary of our recent efforts to develop polyamides as therapeutic agents for human disease and to identify another class of small molecules that offer promise in the treatment of neurodegenerative diseases. T B L O C K I N G C A N C E R C E L L P R O L I F E R AT I O N W I T H A P O LYA M I D E - C H L O R A M B U C I L C O N J U G AT E The nitrogen mustard chlorambucil is a common DNA alkylator used to treat a variety of lymphatic cancers. Because chlorambucil alkylates DNA at all potentially available guanine residues in the genome, coupling of chlorambucil to a polyamide will increase the DNA- THE SCRIPPS RESEARCH INSTITUTE sequence specificity and perhaps decrease unwanted side effects while retaining the ability of the compound to kill cancer cells. We recently found that a specific polyamide-chlorambucil conjugate called 1R-Chl alters the morphology and growth characteristics of colon carcinoma cells in culture and causes the cells to arrest in the G2/M stage of the cell cycle, without any apparent cytotoxic effects. Cells treated with 1R-Chl do not grow in soft agar and do not form tumors in nude mice, indicating that polyamide-treated cells are no longer tumorigenic. The compound blocks proliferation of metastatic colon carcinoma cells in immunocompromised mice, and no apparent toxic effects occur at doses required for a therapeutic effect. Importantly, this gene-targeted small molecule requires no delivery vehicle because the molecule is cell permeable and localizes in the nucleus of various cancer cell lines. Using microarray analysis, we found that the gene target of 1R-Chl is the gene for histone H4c, a member of the gene family that encodes a critical component of cellular chromatin and a gene that is highly expressed in a wide range of cancer cells. Reduction in histone H4 protein by polyamide treatment was confirmed in cells treated with 1R-Chl, which caused chromatin decondensation. To confirm that downregulation of histone H4c transcription is the primary event leading to cell-cycle arrest by 1R-Chl, we turned to short interfering RNAs directed toward H4c mRNA. Unlike 1R-Chl, which arrests cells at the G2/M phase of the cell cycle, the H4c short interfering RNA arrests cells at the G 1 /S phase. However, G 2/M arrest by 1R-Chl and downregulation of the H4c gene can be confirmed in other tumorigenic cell lines. We found that 1R-Chl causes extensive DNA damage in colon cancer cells, leading to phosphorylation of histone H2A.X at serine 139 and recruitment of the DNA repair protein Nbs1 to discrete sites in the genome. These events are hallmarks of the cellular DNA damage response pathway. Control polyamide-Chl conjugates that lack binding sites in the H4c gene and have no antiproliferative effects by themselves can cause G2/M cell-cycle arrest when used in combination with short interfering RNAs to histone mRNAs. On the basis of these findings, we propose that 1R-Chl exerts its antiproliferative effect through a novel 2-hit mechanism. The highly transcribed H4c gene in several cancer cell lines is a primary target for DNA alkylation by 1R-Chl, resulting in downregulation of H4c transcription and histone H4 protein. Loss of his- MOLECULAR BIOLOGY 2006 tone protein leads to a transition from condensed to open chromatin, exposing otherwise hidden binding sites for 1R-Chl. These sites are then alkylated by 1R-Chl, causing widespread DNA damage and a cascade of events leading to G2/M arrest and loss of tumorigenicity. Our findings indicate how a single molecule can target cancer cells because of a specific gene expression profile and block cancer cell proliferation. Ongoing studies are aimed at the development of 1R-Chl as a potential human cancer therapeutic agent. P O LYA M I D E S A S A C T I VAT O R S O F G E N E E X P R E S S I O N The neurodegenerative disease Friedreich’s ataxia is caused by gene silencing through expansion of GAATTC triplet repeats in the first intron of a nuclear gene that encodes the essential mitochondrial protein frataxin. Normal frataxin alleles have 6–34 repeats whereas alleles from patients with Friedreich’s ataxia have 66–1700 repeats. Longer repeats cause a more profound frataxin deficiency and are associated with earlier onset and increased severity of the disease. Two models have been proposed to account for gene silencing by expanded GAA-TTC repeats: unusual DNA structures and repressive heterochromatin. Molecules that reverse formation of unusual DNA structures and/or heterochromatin in the gene for frataxin most likely increase transcription through expanded GAA-TTC repeats, thereby relieving the deficiency in frataxin mRNA and protein in cells from patients with Friedreich’s ataxia. We found that polyamides targeting GAA-TTC repeats partially alleviated transcription repression of frataxin in a cell line derived from white blood cells from a patient with Friedreich’s ataxia. These molecules also increased frataxin protein levels in these cells, and microarray studies showed that a limited number of genes in the human genome were affected by polyamides targeting GAA-TTC repeat DNA. We hypothesize that polyamides might act as a thermodynamic “sink” and lock GAA-TTC repeats into doublestranded B DNA. Such an event would disfavor duplex unpairing, which is necessary for formation of the unusual DNA structures associated with expanded triplet repeats. Alternatively, polyamides may relieve heterochromatin-mediated repression by opening the chromatin domain containing frataxin. To explore this last hypothesis, we turned to another class of small molecules. H I S T O N E D E A C E T Y L A S E I N H I B I T O R S T H AT R E V E R S E F R ATA X I N S I L E N C I N G We used antibodies to the various modification states of the core histones and chromatin immunoprecipita- THE SCRIPPS RESEARCH INSTITUTE 187 tion methods to examine the chromatin structure of the gene for frataxin in normal cells and in cell lines derived from patients with Friedreich’s ataxia. We found that gene silencing at expanded frataxin alleles was accompanied by hypoacetylation of histones H3 and H4 and methylation of histone H3 at lysine 9, consistent with a heterochromatin-mediated repression mechanism. These findings suggest that histone deacetylase inhibitors, compounds that reverse heterochromatin, might activate frataxin. We identified a commercial histone deacetylase inhibitor, BML-210, that partially reverses silencing in the Friedreich’s ataxia cell line. On the basis of the structure of this compound, we synthesized and assayed a series of derivatives of BML-210 and identified histone deacetylase inhibitors that reverse frataxin silencing in primary lymphocytes from patients with Friedreich’s ataxia. These molecules act directly on the histones associated with frataxin, increasing acetylation at particular lysine residues on histones H3 and H4. Unlike many triplet-repeat diseases (e.g., the polyglutamine expansion diseases such as Huntington’s disease and the spinocerebellar ataxias), expanded GAA-TTC triplets do not alter the coding potential of frataxin. Thus, gene activation would be of therapeutic benefit. Studies in animals are under way to explore the bioavailability and efficacy of these histone deacetylase inhibitors. PUBLICATIONS Alvarez, D., Chou, C.J., Latella, L., Zeitlin, S.G., Ku, S., Puri, P.L., Dervan, P.B., Gottesfeld, J.M. A two-hit mechanism for pre-mitotic arrest of cancer cell proliferation by a polyamide-alkylator conjugate. Cell Cycle 5:1537, 2006. Burnett, R., Melander, C., Puckett, J.W., Son, L.S., Wells, R.D., Dervan, P.B., Gottesfeld, J.M. DNA sequence-specific polyamides alleviate transcription inhibition associated with long GAA-TTC repeats in Friedreich’s ataxia. Proc. Natl. Acad. Sci. U. S. A. 103:11497, 2006. Herman, D., Jenssen, K., Burnett, R., Soragni, E., Perlman, S.L., Gottesfeld, J.M. Histone deacetylase inhibitors reverse gene silencing in Friedreich’s ataxia. Nat. Chem. Biol. 2:551, 2006. Lee, B.M., Xu, J., Clarkson, B.K., Martinez-Yamout, M.A., Dyson, H.J., Case, D.A., Gottesfeld, J.M., Wright, P.E. Induced fit and “lock and key” recognition of 5S RNA by zinc fingers of transcription factor IIIA. J. Mol. Biol. 357:275, 2006. Trzupek, J.D., Gottesfeld J.M., Boger D.L. Alkylation of duplex DNA in nucleosome core particles by duocarmycin SA and yatakemycin. Nat. Chem. Biol. 2:79, 2006. Nucleic Acid Dynamics D.P. Millar, J. Gill, G. Pljevaljc̆ić, S. Pond, G. Stengel, N. Tassew, E.J.C. Van der Schans T he focus of our research is the assembly and conformational dynamics of nucleic acid–based macromolecular machines and assemblies. We 188 MOLECULAR BIOLOGY 2006 use single-molecule fluorescence methods to investigate a range of systems, including ribozymes, ribonucleoprotein complexes, and DNA polymerases. Our studies reveal the dynamic structural rearrangements that occur during the assembly and function of these macromolecular machines. RIBOZYMES RNA conformation plays a central role in the mechanism of ribozyme catalysis. The hairpin ribozyme is a small nucleolytic ribozyme that serves as a model system for studies of RNA folding and catalysis. The hairpin ribozyme consists of 2 internal loops, 1 of which contains the scissile phosphodiester bond, displayed on 2 arms of a 4-way multihelix junction. To attain catalytic activity, the ribozyme must fold into a compact conformation in which the 2 loops become connected by a network of tertiary hydrogen bonds. We monitor the formation of this docked structure by using fluorescence resonance energy transfer (FRET) and ribozyme constructs labeled with donor and acceptor dyes within the loop-bearing arms. By measuring FRET at the level of single ribozyme molecules, we reveal subpopulations of compact and extended conformers that are not detected in ensemble experiments. Using this approach, we found that the ribozyme populates an intermediate state in which the 2 loops are in proximity but tertiary interactions have yet to form. This quasi-docked state forms rapidly (submillisecond timescale), but the subsequent formation of the tertiary contacts between the 2 loops occurs much more slowly. The hairpin ribozyme is an ideal system for exploring this fundamental mechanism of the formation of RNA tertiary structure. R I B O N U C L E O P R O T E I N A S S E M B LY The Rev protein from HIV type 1 is a key regulatory protein that controls the transition from early to late patterns of viral gene expression. Rev binds to a highly structured region within the viral mRNA, known as the Rev response element (RRE), where it forms an oligomeric ribonucleoprotein complex. The formation of this complex inhibits splicing and facilitates export of the viral RNA from the nucleus to the cytoplasm. Because of its critical role in the viral life cycle, the Rev-RRE complex provides a novel target for the development of therapeutic drugs. To dissect the mechanism of assembly of ribonucleoprotein complexes, we use single-molecule fluorescence imaging methods to monitor the progressive formation of oligomeric complexes of Rev on individual RRE mole- THE SCRIPPS RESEARCH INSTITUTE cules immobilized on a solid surface. We also use single-pair FRET to probe changes in the conformation of the RRE during the assembly process. We are using the results of these mechanistic studies to develop novel fluorescence-based methods for high-throughput screening of libraries of chemical compounds. The new screening tools are being used to identify small molecules that block binding of Rev to the RRE or prevent the subsequent Rev-Rev oligomerization. D N A P O LY M E R A S E S DNA polymerases are remarkable for their ability to synthesize DNA at rates approaching several hundred base pairs per second while maintaining an extremely low frequency of errors. To elucidate the origin of polymerase fidelity, we are using single-molecule fluorescence methods to examine the dynamic interactions that occur between a DNA polymerase and its DNA and nucleotide substrates. The FRET method is being used to observe conformational transitions of the enzyme-DNA complex that occur during selection and incorporation of an incoming nucleotide substrate. Our results reveal that binding of a correct nucleotide substrate induces a slow conformational change within the polymerase, causing the “fingers” subdomain to close over the DNA primer terminus and incoming nucleotide. Our studies are providing new insights into the dynamic structural changes responsible for nucleotide recognition and selection by DNA polymerases. Single-pair FRET methods are also being used to monitor the movement of the DNA primer/template between the separate polymerizing and editing sites of the enzyme. This active-site switching of DNA plays a key role in the proofreading process used to remove misincorporated nucleotides from the newly synthesized DNA. The advantage of single-molecule observations is that they eliminate the need to synchronize a population of molecules, allowing these dynamic processes to be directly observed. PUBLICATIONS Bailey, M.F., Van der Schans, E.J.C., Millar, D.P. Dimerization of the Klenow fragment of Escherichia coli DNA polymerase I is linked to its mode of DNA binding. Biochemistry, in press. Tian, F., Debler, E.W., Millar, D. P., Deniz, A.A., Wilson, I.A., Schultz, P.G. Multicolor fluorescent antibodies. Angew. Chemie, in press. MOLECULAR BIOLOGY 2006 Single-Molecule Biophysics A.A. Deniz, S.Y. Berezhna, J.P. Clamme, A.C.M. Ferreon, Y. Gambin, E. Lemke, S. Mukhopadhyay, P. Zhu e develop and use state-of-the-art singlemolecule fluorescence methods to address key biological questions. Single-molecule and small-ensemble methods offer key advantages over traditional measurements, allowing us to directly observe the behavior of individual subpopulations in mixtures of molecules and to measure kinetics of structural transitions of stochastic processes under equilibrium conditions. We use these methods to study multiple structural states or reaction pathways during the folding and assembly of biomolecules. A major goal is to apply single-molecule methods to studies of protein folding and aggregation. Using relatively simple model systems, we are addressing several fundamental questions about folding mechanisms. Partially folded or misfolded protein structures are also thought to play important cellular roles, and these states also can be studied by using single-molecule methods. In this context, we are examining the interplay between folding and aggregation of Sup35, a yeast prion protein, in collaboration with S.L. Lindquist, Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, and of α-synuclein, a protein implicated in the pathogenesis of Parkinson’s disease and other neurodegenerative diseases. In addition, we have developed a single-molecule fluorescence quenching method that will be useful for measuring distances shorter than 30 Å in proteins and RNA, a scale at which the resolution of single-pair fluorescence resonance energy transfer (FRET) is low. This method is being used to monitor structural properties of Sup35 as a function of the aggregation process. To better study the folding, assembly, and activity of larger and multicomponent biological complexes, we are developing new multicolor single-molecule FRET methods. As part of this continuing goal, we have been improving our recently developed diffusion 3-color single-molecule FRET method for simultaneously measuring more than a single intramolecular or intermolecular distance. In collaboration with J.R. Williamson, Department of Molecular Biology, we are using these novel methods to study the detailed mechanisms of assembly of fragments of the bacterial ribosome. Most recently, we began adding microfluidics capabilities to our exper- W THE SCRIPPS RESEARCH INSTITUTE 189 imental repertoire, to further facilitate studies of molecular structure, folding, and function. Finally, using high-sensitivity fluorescence imaging, we are beginning to study and compare the pathways of nuclear and cytoplasmic RNA interference. In studies done in collaboration with P.G. Schultz, Department of Chemistry, our observations of the localization of small interfering RNA in live cells provide evidence for a yetto-be-determined mechanism that directs the RNA to cellular compartments containing the target RNA. PUBLICATIONS Berezhna, S.Y., Supekova, L., Supek, F., Schultz, P.G., Deniz, A.A. siRNA in human cells selectively localizes to target RNA sites. Proc. Natl. Acad. Sci. U. S. A. 103:7682, 2006. Zhu, P., Clamme, J.-P., Deniz, A.A. Fluorescence quenching by TEMPO: a sub-30 Å single-molecule ruler. Biophys. J. 89:L37, 2005. Computer Modeling of Proteins and Nucleic Acids D.A. Case, M. Crowley, Q. Cui, F. Dupradeau,* S. Moon, D. Nguyen, V. Pelmentschikov, D. Shivakumar, R.C. Walker, W. Zhang, J. Ziegler** * Université Jules Verne, Amiens, France ** Universität Bayreuth, Bayreuth, Germany omputer simulations offer an exciting approach to the study of many aspects of biochemical interactions. We focus primarily on molecular dynamics simulations (in which Newton’s equations of motions are solved numerically) to model the solution behavior of biomacromolecules. Recent applications include detailed analyses of electrostatic interactions in short peptides (folded and unfolded), proteins, and oligonucleotides in solution. In addition, molecular dynamics methods are useful in refining solution structures of proteins by using constraints derived from nuclear magnetic resonance (NMR) spectroscopy, and we continue to explore new methods in this area. Our developments are incorporated into the Amber molecular modeling package, designed for large-scale biomolecular simulations, and into other software, including Nucleic Acid Builder, for developing 3-dimensional models of unusual nucleic acid structures; SHIFTS, for analyzing chemical shifts in proteins and nucleic acids; RNAmotif, for finding structural motifs in genomic sequence databases; and DOCK, for placing inhibitors into enzyme active sites. C 190 MOLECULAR BIOLOGY 2006 NMR AND THE STRUCTURE AND DYNAMICS OF PROTEINS AND NUCLEIC ACIDS Our overall goal is to extract the maximum amount of information about biomolecular structure and dynamics from NMR experiments. To this end, we are studying the use of direct refinement methods for determining biomolecular structures in solution, going beyond distance constraints to generate closer connections between calculated and observed spectra. We are also using quantum chemistry to study chemical shifts and spinspin coupling constants. Other types of data, such as chemical shift anisotropies, direct dipolar couplings in partially oriented samples, and analysis of cross-correlated relaxation, are also being used to guide structure refinement. In recent structural studies, we focused on the binding of zinc finger proteins with RNA and on structural influences on amide proton chemical shifts. NUCLEIC ACID MODELING Another project centers on the development of novel computer methods to construct models of “unusual” nucleic acids that go beyond traditional helical motifs. We are using these methods to study circular DNA, small RNA fragments, and 3- and 4-stranded DNA complexes, including models for recombination sites. We continue to develop efficient computer implementations of continuum solvent methods to allow simplified simulations that do not require a detailed description of the solvent (water) molecules; this approach also provides a useful way to study salt effects. Recent efforts have made second derivatives of these energies available, so that normal mode analyses of nucleic acids with dozens to hundreds of nucleotides can be analyzed and the predictions compared with those of simpler, elastic continuum models. These efforts provide a new avenue for developing and testing low-resolution models that can be used for large molecular assemblies. D Y N A M I C S A N D E N E R G E T I C S O F N AT I V E A N D N O N N AT I V E S TAT E S O F P R O T E I N S Analysis methods similar to those described for nucleic acids are also being used to estimate thermodynamic properties of “molten globules” and unfolded states of proteins. These studies are an extension of our earlier work on the folding of peptide fragments of proteins. A key feature is the development of computational methods that can be used to model pH and salt dependence of complex conformational transitions such as unfolding events. A second aspect of this research is a detailed interpretation of NMR results for protein non- THE SCRIPPS RESEARCH INSTITUTE native states through molecular dynamics simulations and the construction of models for molecular motion and disorder. All of these modeling activities are based on molecular mechanics force fields, which provide estimates of energies as a function of conformation. We continue to work on improvements in force fields; recently, we focused on adding aspects of electronic polarizability, going beyond the usual fixed-charge models, and on methods for handling arbitrary organic molecules that might be considered potential inhibitors in drug discovery efforts. Overall, the new models should provide a better picture of the noncovalent interactions between peptide groups and the groups’ surroundings, leading ultimately to more faithful simulations. V I B R AT I O N A L A N A LY S I S O F I R O N - S U L F U R C L U S T E R S IN PROTEINS A wide variety of proteins contain iron-sulfur clusters at their active sites; these proteins participate in electrontransport chains and in important enzymatic reactions such as the reduction of atmospheric nitrogen to ammonia by nitrogenase. Advances in synchrotron radiation sources now make it possible to probe the vibrational behavior of these clusters by using nuclear resonance vibrational spectroscopy (NRVS). This technique senses the coupling of a nuclear (Mossbauer) excitation to molecular vibrations. The result is a set of vibrational frequencies and intensities that indicate what sorts of deformations can take place. When the molecular structure is known, this information can contribute to the understanding of oxidation-reduction behavior and electron transfer kinetics. In situations in which the cluster structure is not known, NRVS data might useful as a “fingerprint” to help identify the structure. We have been using quantum chemistry calculations to help understand NRVS spectra. Figure 1 shows a early example, comparing calculated and experimental spectra for a simple iron-sulfur “cubane” structure, a cluster type found in hundreds of known proteins. The calculations (shown as a dashed line) are in excellent agreement with experimental data (solid line), both in terms of frequencies and in terms of intensities. We are extending these calculations to models for the active site of nitrogenase, where the structure of the complex is still uncertain. If calculations like these can be used to closely track the experimental results, NRVS will be an important new tool for characterizing the active sites of metalloenzymes. MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 191 Rizzo, R.C., Aynechi, T., Case, D.A., Kuntz, I.D. Estimation of absolute free energies of hydration using continuum methods: accuracy of partial charge models and optimization of nonpolar contributions. J. Chem. Theory Comput. 2:128, 2006. Steinbrecher, T., Case, D.A., Labahn, A. A multistep approach to structure-based drug design: studying ligand binding at the human neutrophil elastase. J. Med. Chem. 49:1837, 2006. Wang, J., Wang, W., Kollman, P.A., Case, D.A. Automatic atom type and bond type perception in molecular mechanical calculations. J. Mol. Graphics Model. 25:247, 2006. Xiao, Y., Fisher, K., Smith, M.C., Newton, W.E., Case, D.A., George, S.J., Wang, H., Sturhahn, W., Alp, E.E., Zhao, J., Yoda, Y., Cramer, S.P. How nitrogenase shakes: initial information about P-cluster and FeMo-cofactor normal modes from nuclear resonance vibrational spectroscopy (NRVS). J. Am. Chem. Soc. 128:7608, 2006. Xiao, Y., Koutmos, M., Case, D.A., Coucouvanis, D., Wang H., Cramer, S.P. Dynamics of an [Fe4S4(SPh)4]2– cluster via IR, Raman, and nuclear resonance vibrational spectroscopy (NRVS): analysis using 36S substitution, DFT calculations, and empirical force fields. Dalton Trans. 2192, 2006, Issue 18. Quantum Chemistry of RedoxActive Metalloenzymes L. Noodleman, D.A. Case, W.-G. Han, V. Pelmenschikov, J.A. Fee, L. Hunsicker-Wang,* T. Lovell,** T. Liu*** * Trinity University, San Antonio, Texas ** AstraZeneca R&D, Mölndal, Sweden *** University of Maryland, College Park, Maryland F i g . 1 . Calculated and experimental NRVS spectra for an ironsulfur cluster. PUBLICATIONS Baker, N.A., Bashford, D., Case, D.A. Implicit solvent electrostatics in biomolecular simulation. In: New Algorithms for Macromolecular Simulation. Leimkuhler, B., et al. (Eds.). Springer, New York, 2006, p. 263. Lecture Notes in Computational Science and Engineering, Vol. 49. Brown, R.A., Case, D.A. Second derivatives in generalized Born theory. J. Comput. Chem. 27:1662, 2006. Case, D.A., Cheatham, T.E. III, Darden, T., Gohlke, H., Luo, R., Merz, K.M., Jr., Onufriev, A., Simmerling, C., Wang, B., Woods, R. The Amber biomolecular simulation programs. J. Comput. Chem. 26:1668, 2005. Dixit, S.B., Beveridge, D.L., Case, D.A., Cheatham, T.E. III, Giudice, E., Lankas, R., Lavery, R., Maddocks, J.H., Osman, R., Sklenar, H., Thayer, K.M., Varnai, P. Molecular dynamics simulations of the 136 unique tetranucleotide sequences of DNA oligonucleotides, II: sequence context effects on the dynamical structures of the 10 unique dinucleotide steps. Biophys. J. 89:3721, 2005. Dupradeau, F.-Y., Case, D.A., Yu, C., Jimenez, R., Romesberg, F.E. Differential solvation and tautomer stability of a model base pair within the minor and major grooves of DNA. J. Am. Chem. Soc. 127:15612, 2005. Lee, B.M., Xu, J., Clarkson, B.K., Martinez-Yamout, M.A., Dyson, H.J., Case, D.A., Gottesfeld, J.M., Wright, P.E. Induced fit and “lock and key” recognition of 5S RNA by zinc fingers of transcription factor IIIA. J. Mol. Biol. 357:275, 2006. Mathews, D.H., Case, D.A. Nudged elastic band calculation of minimal energy pathways for the conformational change of a GG noncanonical pair. J. Mol. Biol. 357:1683, 2006. Moon, S., Case, D.A. A comparison of quantum chemical models for calculating NMR shielding parameters in peptides: mixed basis set and ONIOM methods combined with a complete basis set extrapolation. J. Comput. Chem. 27:825, 2006. e use a combination of modern quantum chemistry (density functional theory, DFT) and classical electrostatics to describe the energetics, reaction pathways, and spectroscopic properties of metalloenzymes. Critical biosynthetic and regulatory processes may involve catalytic transformations of fairly small molecules or groups by transition-metal centers. The ironmolybdenum cofactor center of nitrogenase catalyzes the multielectron reduction of molecular nitrogen to 2 molecules of ammonia plus molecular hydrogen. We are continuing our work on the catalytic cycle of this enzyme, following up on our earlier work on the structure and oxidation state of the cofactor complex in the “resting enzyme” before multielectron reduction and nitrogen binding. On the basis of DFT calculated vs experimental physical properties, including redox potentials, cluster geometries, and Mössbauer isomer shifts, the core cluster has a MoFe 7S 9X prismane active site, where the central X most likely is nitride and the “resting cluster oxidation state” is Mo(IV)Fe(II) 4Fe(III) 3. If the central ligand is nitride, as we have proposed, this ligand is not a substrate or a reaction product of the catalytic cycle. Instead, nitride is inserted into a cen- W 192 MOLECULAR BIOLOGY 2006 tral vacancy site of a more open iron-molybdenum cofactor precursor, probably in a noncatalytic deamination process that occurs before insertion of the cluster into the iron-molybdenum protein. Class I ribonucleotide reductases are aerobic enzymes that catalyze the reduction of ribonucleotides to deoxyribonucleotides, providing the required building blocks for DNA replication and repair. These ribonucleotideto-deoxyribonucleotide reactions occur via a long-range radical (or proton-coupled electron transfer) propagation mechanism initiated by a fairly stable tyrosine radical, “the pilot light.” When this pilot light goes out, the tyrosine radical is regenerated by a high-oxidation-state Fe(III)-Fe(IV)-oxo enzyme called intermediate X. We are using DFT and electrostatics calculations in combination with analysis of Mössbauer, electron nuclear double resonance, and magnetic circular dichroism spectroscopies to search for a proper structural and electronic model for intermediate X. We have also examined the mechanism of formation of intermediate X, starting from an earlier Fe(III)2-µperoxo intermediate (Fig. 1). Spectroscopic and quantum chemical DFT evidence indicates that the formation of intermediate X is proton catalyzed. On the basis of calculations of spectroscopic parameters and energies, we propose that intermediate X contains a dioxo bridging the Fe(III)-Fe(IV) in an asymmetric diamond structure. The Fe(IV) site is farther from and the Fe(III) site is closer to the redox-active tyrosine 122. Figure 2 shows the molecular orbitals corresponding to the lowest energy Fe(IV) d→d optical excitation. The 3 Fe(IV) d→d bands that we predict on the basis of DFT vertical self-consistent reaction field methods are in excellent agreement with the bands observed by using magnetic circular dichroism spectroscopy. Further exploration of the tyrosine radical activation and subsequent catalytic cycle are planned. THE SCRIPPS RESEARCH INSTITUTE Noodleman, L., Han, W.-G. Structure, redox, pKa, spin: a golden tetrad for understanding metalloenzyme energetics and reaction pathways. J. Biol. Inorg. Chem. 11:674, 2006. PUBLICATIONS Han, W.-G., Liu, T., Lovell, T., Noodleman, L. Active site structure of class I ribonucleotide reductase intermediate X: a density functional theory analysis of structure, energetics, and spectroscopy. J. Am. Chem. Soc. 127:15778, 2005. Han, W.-G., Liu, T., Lovell, T., Noodleman, L. Density functional theory study of Fe(IV) d-d optical transitions in active-site models of class I ribonucleotide reductase intermediate X with vertical self-consistent reaction field methods. Inorg. Chem. 45:8533, 2006. Han, W.-G., Liu, T., Lovell, T., Noodleman, L. DFT calculations of 57Fe Mössbauer isomer shifts and quadrupole splittings for iron complexes in polar dielectric media: applications to methane monooxygenase and ribonucleotide reductase. J. Comput. Chem. 27:1292, 2006. Han, W.-G., Liu, T., Lovell, T., Noodleman, L. Seven clues to the origin and structure of class-I ribonucleotide reductase intermediate X. J. Inorg. Biochem. 100:771, 2006. F i g . 1 . A feasible path showing how ribonucleotide reductase intermediate X is formed by the reaction of oxygen with the reduced ribonucleotide reductase–R2 di-iron center. Reproduced with permission from J. Am. Chem. Soc. 127:15778, 2005. Copyright 2005 American Chemical Society. MOLECULAR BIOLOGY 2006 F i g . 2 . Our proposed model for the active site of class I ribonucleotide reductase intermediate X. Molecular orbital plots show the lowest energy Fe(IV) d→d optical excitation. Theoretical and Computational Molecular Biophysics C.L. Brooks III, C. An, R. Armen, I. Borelli, D. Bostick, D. Braun, L. Bu, J. Chen, M.F. Crowley, O. Guvench, R. Hills, W. Im,* J. Khandogin, I. Khavrutskii, J. Lee, J. Magee,** R. Manige, M. Michino, A. Mitsutake,*** H.D. Nguyen, S. Patel,**** D.J. Price, V. Reddy, H.A. Scheraga,***** C. Shepard, F. Tama,† I.F. Thorpe, M.C. Tripp, R. Wheeler,†† C. Wildman, K. Yoshimoto * Kansas University, Lawrence, Kansas ** University of Manchester, Manchester, England *** Kelo University, Tokyo, Japan **** University of Delaware, Newark, Delaware ***** Cornell University, Ithaca, New York † University of Arizona, Tucson, Arizona †† University of Oklahoma, Norman, Oklahoma nderstanding the forces that determine the structure of proteins, peptides, nucleic acids, and complexes containing these molecules and the processes by which these structures are adopted is essential to complete our knowledge of the molecular nature of structure and function. To address such questions, we use statistical mechanics, molecular simulation, statistical modeling, and quantum chemistry. Creating atomic-level models to simulate biophysical processes (e.g., protein folding or binding of a ligand to a biological receptor) requires (1) the development of new potential energy functions that accurately represent the atomic interactions and (2) the use of quan- U THE SCRIPPS RESEARCH INSTITUTE 193 tum chemistry to aid in determining the parameters for the models. Calculation of thermodynamic properties requires the development and implementation of new theoretical and computational approaches that connect averages over atomistic descriptions to experimentally measurable thermodynamic and kinetic properties. Interpreting experimental results at more microscopic levels is fueled by the development and investigation of theoretical models for the processes of interest. Massive computational resources are needed to realize these objectives, and this motivates our efforts aimed at the efficient use of new computer architectures, including large supercomputers, Linux Beowulf clusters, computational grids, and Internet-based volunteer supercomputers. Each of the objectives and techniques mentioned represents an ongoing development area within our research program in computational biophysics. The following are highlights of a few specific projects. FOLDING, STRUCTURE, AND FUNCTION OF MEMBRANE-BOUND PROTEINS Folding, insertion, assembly, and stability of membrane proteins are directly governed by the unique hydrophilic and hydrophobic environment provided by biological membranes. Modeling this heterogeneous environment is both an obstacle and an essential requisite to experimental and computational studies of the structure and function of membrane proteins. Because of the biological importance and marked presence of membrane proteins in known genomes (i.e., about 30% of all proteins), one aim of modern molecular biophysics should be the development of methods that can be used in experimental studies to understand the structure and function of these systems. We recently developed theoretical methods that enable the exploration of protein insertion and folding in membranes. These methods combine the sampling methods of replica-exchange molecular dynamics with novel generalized Born implicit solvent/ implicit membrane continuum electrostatic theories. A key question these methods allow us to address is the association of integral membrane proteins to form oligomeric structures. Many important functional complexes of membrane proteins exist as oligomers, such as the signal-transducing G protein–coupled receptors and membrane-bound ion channels and transporters. Our recent approach provides a way to predict the structures of these key oligomeric states. Figure 1 shows the predicted oligomeric structures of glycophorin A (functionally a dimer), the tetrameric M2 transmembrane peptide proton channel, and the phospholamban pentameric 194 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE One recent advance came in exploring the structure of the ribosome in complex with the SecY protein-conducting channel (PCC). The translocation of secreted and membrane proteins across or into cell membranes occurs through PCCs. Using an electron cryomicroscopy reconstruction of the Escherichia coli PCC, which consisted of SecY complexed with the ribosome and a nascent chain containing a signal anchor, we observed the components of protein synthesis and translocation, including mRNA, 3 tRNAs, the nascent chain, and features of both a translocating PCC and a second, nontranslocating PCC bound to mRNA hairpins (Fig. 2). F i g . 1 . The predicted structure of dimeric glycophorin A, a dominant structural component of red blood cells, indicates the “classic” GVXXGV helical interface. For the M2 proton channel involved in replication of the influenza virus, the structure of the functional tetrameric proton-conducting channel is shown. In phospholamban, which is localized in the membrane of the cardiac sarcoplasmic reticulum and involved in phosphorylation-controlled regulation of the cardiac calcium pump, the predicted pentameric structure selectively conducts calcium. oligomer. Our calculations provide detailed predictions of the protein-protein interfaces for these systems and may be useful in elucidating the primary oligomerization states. The predicted models shown in the figure are in excellent agreement with existing structural models (from experiments and other model building). LARGE-SCALE FUNCTIONAL DYNAMICS IN MOLECULAR ASSEMBLIES Many naturally occurring machines, such as ribosomes, myosin, and viruses, require large-scale dynamical motions as a component of their normal functioning. These motions involve the “mechanical” reorganization of major parts of the structure of the machine in response to binding of effectors or the addition of energy in the form of thermal fluctuations or provided by chemical catalysis. Exploring and understanding the character and nature of such large-scale reorganization of biological machines are ongoing goals in our laboratory. Using theoretical approaches derived from the treatment of mechanoelastic materials, we developed new structure refinement methods to model large-scale macromolecular assemblies. The methods are based on atomic-level structures of the component macromolecules (e.g., RNAs, DNAs, and proteins) or on single-particle or tomographic images from electron microscopy. Using these new methods, which we call normal mode flexible fitting, we have collaborated with several colleagues in elucidating new structural models for functionally important molecular assemblies. F i g . 2 . Electron cryomicroscopy image of the ribosome with 2 bound PCCs obtained during the modeling of structural components of the SecY dimer into the electron density for the nontranslocating and translocating PCCs. The figure on the lower right illustrates the structure of the SecY dimer fit into the experimental electron density map by using normal mode flexible fitting. NNMF indicates normal mode flexible fitting. Normal mode flexible fitting of the SecYEb structure into the PCC electron microscopy densities favors a front-to-front arrangement of 2 SecYEG complexes in the PCC and supports channel formation by the opening of 2 linked SecY halves during polypeptide translocation. From the models elucidated by the combination of electron cryomicroscopy and modeling based on normal mode flexible fitting, we were able to develop a model for cotranslational protein translocation. PUBLICATIONS Chen, J., Im, W., Brooks, C.L. III. Application of torsion angle molecular dynamics for efficient sampling of protein conformations. J. Comput. Chem. 26:1565, 2005. Chen, J., Im, W., Brooks, C.L. III. Balancing solvation and intramolecular interactions: toward a consistent generalized Born force field. J. Am. Chem. Soc. 128:3728, 2006. Im, W., Chen, J., Brooks, C.L. III. Peptide and protein folding and conformational equilibria: theoretical treatment of electrostatics and hydrogen bonding with implicit solvent models. Adv. Protein Chem. 72:173, 1005. Khandogin, J., Brooks, C.L. III. Constant pH molecular dynamics with proton tautomerism. Biophys. J. 89:141, 2005. Khavrutskii, I.V., Byrd, R.H., Brooks, C.L. III. A line integral reaction path approximation for large systems via nonlinear constrained optimization: application to alanine dipeptide and the β-hairpin of protein G. J. Chem. Phys. 124:194903, 2006. MOLECULAR BIOLOGY 2006 Konecny, R., Trylska, J., Tama, F., Zhang, D., Baker, N.A., Brooks, C.L. III, McCammon, J.A. Electrostatic properties of cowpea chlorotic mottle virus and cucumber mosaic virus capsids. Biopolymers 82:106, 2005. Mitra, K., Schaffitzel, C., Shaikh, T., Tama, F., Jenni, S., Brooks, C.L. III, Ban, N., Frank, J. Structure of the E. coli protein-conducting channel bound to a translating ribosome. Nature 438:318, 2005. Natarajan, P., Lander, G.C., Shepherd, C.M., Reddy, V.S., Brooks, C.L. III, Johnson, J.E. Exploring icosahedral virus structures with VIPER. Nat. Rev. Microbiol. 3:809, 2005. Patel, S., Brooks, C.L. III. Fluctuating charge force fields: recent developments and applications from small molecules to macromolecular biological systems. Mol. Simul. 32:231, 2006. Patel, S., Brooks, C.L. III. Revisiting the hexane-water interface via molecular dynamics simulations using nonadditive alkane-water potentials. J. Chem. Phys. 124:204706, 2006. Patel, S., Brooks, C.L. III. Structure, thermodynamics, and liquid-vapor equilibrium of ethanol from molecular-dynamics simulations using nonadditive interactions. J. Chem. Phys. 123:164502, 2005. Price, D.J., Brooks, C.L. III. Detailed considerations for a balanced and broadly applicable force field: a study of substituted benzenes modeled with OPLS-AA. J. Comput. Chem. 26:1529, 2005. Tama, F., Brooks, C.L. III. Symmetry, form, and shape: guiding principles for robustness in macromolecular machines. Annu. Rev. Biophys. Biomol. Struct. 35:115, 2006. Tama, F., Brooks, C.L. III. Unveiling molecular mechanisms of biological functions in large macromolecular assemblies using elastic network normal mode analysis. In: Normal Mode Analysis: Theory and Applications to Biological and Chemical Systems. Cui, Q., Bahar, I. (Eds.). Chapman & Hall/CRC Press, Boca Raton, FL, 2006, p. 111. Mathematical and Computational Biology Series. Taufer, M., An, C., Kerstens, A., Brooks, C.L. III. Predictor@Home: a “protein structure prediction supercomputer” based on global computing. IEEE Trans. Parallel Distributed Syst. 7:786, 2006. Thorpe, I.F., Brooks, C.L. III. Conformational substates modulate hydride transfer in dihydrofolate reductase. J. Am. Chem. Soc. 127:12997, 2005. Trylska, J., McCammon, J.A., Brooks, C.L. III. Exploring assembly energetics of the 30S ribosomal subunit using an implicit solvent approach. J. Am. Chem. Soc. 127:11125, 2005. Yadav, M.K., Leman, L.J., Price, D.J., Brooks, C.L. III, Stout, C.D., Ghadiri, M.R. Coiled coils at the edge of configurational heterogeneity: structural analyses of parallel and antiparallel homotetrameric coiled coils reveal configurational sensitivity to a single solvent-exposed amino acid substitution. Biochemistry 45:4463, 2006. Computation and Visualization in Structural Biology A.J. Olson, D.S. Goodsell, M.F. Sanner, S. Dallakyan, A. Gillet, R. Harris, Y. Hu, R. Huey, J. Huntoon, S. Karnati, W. Lindstrom, G.M. Morris, A. Omelchenko, M. Pique, B. Norledge, R. Rosenstein, M. Utsintong, G. Vareille, Q. Zhang, Y. Zhao n the Molecular Graphics Laboratory, we develop novel computational methods to analyze, understand, and communicate the structure and interactions of complex biomolecular systems. This past year, we I THE SCRIPPS RESEARCH INSTITUTE 195 showed the effectiveness of 3-dimensional molecular models as a tangible human-computer interface in educational and research settings. Within our component-based visualization environment, we continue to develop methods for predicting biomolecular interactions, analyzing biomolecular structure and function, and presenting the biomolecular world in education and outreach. We have applied these methods to several important systems in human health and welfare. In a novel distributed computing network, we continue the search for HIV protease inhibitors to fight the growing problem of drug resistance in HIV disease. We used AutoDock, a suite of programs for predicting bound conformations and binding energies for biomolecular complexes, in the virtual screening of large databases of compounds and ultimately identified new compounds for use in the treatment of cancer. We used methods for predicting protein interaction to probe the mechanism of blood coagulation. TA N G I B L E I N T E R FA C E S I N S T R U C T U R A L B I O L O G Y We have continued to develop autofabricated physical models (“solid printing”) of biological molecules and the components and assemblies of the molecules; our goal is to use the models in both research and education. We integrated computer graphics and computation with these physical models by using augmented reality to create custom interfaces to facilitate exploration and computation of molecular interactions. We have begun to use a self-assisted protein-folding model to teach elements of protein structure and assembly to our graduate students. We are continuing to develop the software that will enable the control of interactive computations through manipulation of the tangible models. In collaboration with T. Herman, Milwaukee School of Engineering, Milwaukee, Wisconsin, we created a model of the active site of acetylcholinesterase that can be opened to show the buried active site and bound substrates (Fig. 1). This model was used, along with an interactive Internet guide to the structure, as part of a Waksman Challenge at Rutgers, the State University of New Jersey. Groups of teachers and students were asked to use the models and associated materials to explore problems with insecticide resistance in compounds that act on mosquito acetylcholinesterase. Recently, we worked with Biomedical Graphics at Scripps Research to establish a solid-model printing service for researchers at Scripps and elsewhere. This service is now in operation and has made a number of molecular models for scientists working with molecular structures. The users’ responses have been positive, and 196 MOLECULAR BIOLOGY 2006 F i g . 1 . A tangible model of the active site of acetylcholinesterase. The model separates into 4 pieces, allowing students to fit different substrates into the buried active site. the research community is beginning to see how a solid 3-dimensional model can provide tangible, multimodal feedback that mouse, keyboard, and image behind a glass screen cannot provide. A D VA N C E S I N C O M P U TAT I O N A L D O C K I N G We have just completed developing and testing a semiempirical free energy force field for use in AutoDock and similar grid-based docking methods. The force field is based on a comprehensive thermodynamic model that allows incorporation of intramolecular energies into the predicted free energy of binding. The model also incorporates a charge-based method for evaluating desolvation designed to use a typical set of atom types. The method was calibrated by using a set of 188 diverse proteinligand complexes of known structure and binding energy and was tested by using a set of 100 complexes of ligands with retroviral proteases. Compared with the previous AutoDock force field, the new force field provides an improvement in redocking simulations. AutoDock 4 has been modified to support more atom types and to use an improved atom-typing mechanism. Importantly, AutoDock 4 also now simulates the molecular system being docked in the unbound state, by generating and evaluating the extended conformation of the ligand and moving side chains in the receptor before the docking occurs. The unbound state is now considered in the calculation of the change in free energy upon binding. AutoDock’s companion graphical user interface, AutoDockTools, has been modified to support preparation of input files for AutoDock 4, in particular to allow the definition of flexible side chains in macromolecules. AutoDockTools has also been made easier to use by simplifying the menus. On the basis of the AutoDock force field, we developed a method for locating and characterizing the opti- THE SCRIPPS RESEARCH INSTITUTE mal binding site for ligands on the surface of a protein of known structure. The method identifies the contiguous constant-volume region with the most favorable binding affinity. The optimal binding sites identify regions of primary binding affinity and regions of suboptimal binding strength, which can be used to predict the function of proteins if the function is unknown or to identify target locations for the design of new inhibitors. We showed the usefulness of the method in the design of inhibitors for HIV type 1 protease, and we are applying the method to a large database of protein structures. C O M P O N E N T - B A S E D V I S U A L I Z AT I O N E N V I R O N M E N T S To facilitate the integration and interoperation of computational models and techniques from a wide variety of scientific disciplines, we continue to expand our component-based software environment. The environment is centered on Python, a high-level, object-oriented, interpretive programming language. This approach allows the compartmentalization and reuse of software components. Python provides a powerful computation “glue” for assembling computational components and, at the same time, a flexible language for the interactive scripting of new applications. We released version 1.4.1 of our software components in March 2006. This release contains substantial enhancements, including a completely rewritten interface to the adaptive Poisson-Bolzman solver APBS, making it easy to produce high-quality pictures of electrostatic potentials on molecular surfaces. A new control panel provides a high-level interface for rapidly displaying molecular models in a variety of representations. This new release is also distributed with installer programs for computers running the Windows and Macintosh OS X operating systems. We also added a parser for macromolecular Crystallographic Information File that allow users to read and write files in the macromolecular Crystallographic Information File format. This addition helps overcome limitations in the Protein Data Bank format such as maximum number of atoms or chain IDs. MODELING PROTEIN FLEXIBILITY IN DOCKING We have developed a hierarchical and multiresolution representation of the flexibility of biological macromolecules that can be used in computational simulations. This treelike structure enables the computationally tractable encoding of a small subset of a protein’s conformational subspace. After implementing the core infrastructure of the Flexibility Tree and developing intuitive graphical interfaces for building such trees, we have MOLECULAR BIOLOGY 2006 started exploring the use of this data structure in the context of automated docking. We reproduced a cross-docking experiment carried out earlier with AutoDock in which 20 inhibitors of HIV protease I where docked systematically into the 20 conformations of the receptor. We showed that by adding receptor flexibility, we could increase the rate of successful cross docking from 72% to 98%. PROTEIN DOCKING In collaboration with C. Bajaj, University of Texas, Austin, we are investigating a novel fast Fourier transform–based method for predicting the association of protein in complexes. In parallel with this docking method, we evaluated the effect on blurring molecular surfaces on the shape complementarity at the interface between proteins in a complex. We characterized the level of distortion introduced by blurring atomic spheres by using gaussian distributions and determined an optimal blurring level for docking purposes. In addition, we added software components for the calculating the curvature of meshes that are used in the docking procedure. F I G H T I N G D R U G R E S I S TA N C E I N H I V D I S E A S E As part of a program project, we continue our work on inhibitors to fight drug resistance in the treatment of AIDS. In collaboration with K.B. Sharpless and C.-H. Wong, Department of Chemistry, we have designed and optimized a series of inhibitors built around a triazole formed in a click chemistry reaction. We are also exploring larger issues of resistance via docking experiments with large chemical databases and large sets of mutant protease structures. These massive docking experiments are made possible by the resources available in the FightAIDS@Home distributed computing system. FightAIDS@Home enlists the worldwide community in a large computational effort to design effective therapeutic agents to fight AIDS. Personal computers are used by the program when the computers are not in use by their owners, providing an enormous, and largely untapped, computational resource. The current goal is to identify inhibitors that are effective against the wild-type virus and against common mutant forms of the virus. In the past year, we moved FightAIDS@Home to the IBM World Community Grid. This transition involved working closely with the team at IBM to board our automated docking software, AutoDock, to be able to run on the Windows United Devices client and the Linux and Macintosh OS X Berkeley Open Infrastructure for Network Computing clients. This move has increased the number of available processors to about 300,000 THE SCRIPPS RESEARCH INSTITUTE 197 and has enabled us to compute a complete scan of the National Cancer Institute diversity set (2000 compounds) against a panel of 200 mutant HIV proteases in a matter of 4 months. This computation required more than 2 quadrillion energy evaluations of ligand vs protein. INTERACTIONS OF TISSUE FACTOR We used our ligand-protein and protein-protein model to study the interactions of tissue factor (TF) in the initiation of blood coagulation and the related regulatory roles of the factor. TF plays a potential role in metastasis, growth, and angiogenesis of tumor cells via 2 distinct mechanisms: interaction of the complex consisting of TF and factor VIIa with protease-activated receptor 2 (PAR2) and interaction of the complex consisting of TF, factor VIIa, and factor Xa with PAR1 or PAR2. However, no PAR structures are available for studying these mechanisms. Because PAR2 is involved in both pathways, we performed protein homology modeling studies of this receptor. We found 8 unique PAR2 sequences. For each unique sequence, we searched its homology sequences in the Protein Data Bank and chose as the homology template the sequence that has the highest-resolution x-ray crystal structure. Sequence alignment was then performed between the PAR2 sequence and the template sequence. The alignment was then input to MODELLER for building 10 homology structures, from which we chose the structure with the best quality as the homology model. The homology models have enabled us to use AutoDock to perform docking studies of PAR2-activating peptides and small molecules on PAR2. The discovered binding modes are being confirmed by our collaborator, W. Ruf, Department of Immunology. STRUCTURE-BASED DRUG DESIGN IN GAUCHER DISEASE Gaucher disease is the most common lipid-storage disorder caused by activity-compromising mutations in glucosylceramidase and is the most common genetic disease affecting Ashkenazi Jews. In addition to causing great pain, anemia, and massive enlargement of the liver and spleen, Gaucher disease can lead to neurologic impairment or early demise. Among the most promising treatments, small-molecule chemical chaperones can rescue the enzyme activity of the misfolded glucosylceramidase. The experimentally identified deoxynojirimycin-type inhibitors have a narrow concentration range and cause a mild improvement in the activity of the mutant enzyme. We did a molecule fragment–based virtual screening with the National Cancer Institute diversity data set. 198 MOLECULAR BIOLOGY 2006 A total of 72 compounds identified as the best compounds of interest by virtual screening were tested in the enzyme assays by our collaborator, J. Kelly, Department of Chemistry. A total of 13 compounds are insoluble in dimethyl sulfoxide at up to 10 mM; 25 precipitate in the assay buffer. Among the remaining 34 compounds examined by using in vitro enzyme assays, 16 show significant inhibition of the enzyme. The top compounds of interest have almost doubled the activities of the mutant enzymes N370S and G202R in vivo. P R O T E I N P H O S P H ATA S E 2 C I N H I B I T O R S In collaboration with P. Greengard, Rockefeller University, New York, New York, we used AutoDock to screen the National Cancer Institute diversity set against protein phosphatase 2C (PP2C), an enzyme that must remain active for tumor growth in breast cancer. Several compounds were identified as inhibitors of PP2C in the computational screen. The compounds were ordered from the National Cancer Institute and were assayed experimentally. Several were inhibitory at micromolar concentrations; the potency of the best was between 5 and 10 µM. The lead compounds discovered in this study are the first nonphosphate-based PP2C inhibitors reported. THE SCRIPPS RESEARCH INSTITUTE to science education and outreach with a combination of presentations, popular and professional illustrations and animations, 3-dimensional tangible models, and a presence on the Worldwide Web. In these projects, we use the diverse visualization tools developed in the Molecular Graphics Laboratory to disseminate results that range from atomic structure to cellular function. We also continued several regular features that informally present molecular structure and function. The “Molecule of the Month” at the Protein Data Bank entered its seventh year of providing an accessible introduction to the central database of biomolecular structure. Each month, a new molecule is presented with a description of the molecule’s structure, function, and relevance to health and welfare (Fig. 2). Visitors are M E C H A N I S T I C S T U D I E S O F B I O C ATA LY S T S I N COCAINE ANTIBODIES Currently, no effective treatment of cocaine addiction approved by the Food and Drug Administration is available. One possible treatment based on receptor design entails thorough investigation of cocaine hydrolysis by catalytic antibodies. Between 2 possible reaction pathways (an oxyanion hole for carbonyl or an hydrogen-bond trap for hydroxide ion formed by tyrosines at positions H50 and L94), quantum mechanical, molecular docking, and molecular dynamics free-energy calculations have shown no conclusive evidence for a dominant pathway between 2 possible ones. An explanation for the low turnover rate of the antibody is the failure of the antibody to promote any mechanism selectively because of a homogeneous microenvironment generated by eliciting against a hapten with 2 equivalent phosphorusoxygen bonds. Computational modeling would help improve hapten design and thus improve the efficiency of the catalytic antibody. V I S U A L M E T H O D S F R O M AT O M S T O C E L L S Understanding structural molecular biology is essential to foster progress and critical decision making among students, policy makers, and the general public. In the past year, we continued our longstanding commitment F i g . 2 . ATP synthase was presented as the Molecule of the Month in 2005. The illustration of this complex molecular machine was constructed from 4 separate entries in the Protein Data Bank: 1c17, 1e79, 2a7u, and 1l2p. then given suggestions about to how to begin their own exploration of the structures in the data bank. Other projects include “The Molecular Perspective,” articles in the journal The Oncologist that present structures of interest to clinical oncologists and provide a source of continuing education for physicians; “Recognition in Action,” a new series at the Journal of Molecular Recognition; and work with the Nanoscale Informal Science Network supported by the National Science Foundation MOLECULAR BIOLOGY 2006 to develop new materials for presenting the science of nanotechnology. PUBLICATIONS Beuscher, A., Olson, A.J., Goodsell, D.S. Identifying protein binding sites and optimal ligands. Lett. Drug Des. Discov. 2:483, 2005. Cheng, T.-J., Goodsell, D.S., Kan, C.-C. Identification of sanguinarine as a novel HIV protease inhibitor from high-throughput screening of 2,000 drugs and natural products with a cell-based assay. Lett. Drug Des. Discov. 2:364, 2005. Dickerson, T.J., Beuscher, A.E. IV, Rogers, C.J., Hixon, M.S., Yamamoto, N., Xu, Y., Olson, A.J., Janda, K.D. Discovery of acetylcholinesterase peripheral anionic site ligands through computational refinement of a directed library. Biochemistry 44:14845, 2005. Goodsell, D.S. Computational docking of biomolecular complexes with AutoDock. In: Protein-Protein Interactions: A Molecular Cloning Manual, 2nd ed. Golemis, E., Adams, P. (Eds.). Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2005, p. 885. Goodsell, D.S. The molecular perspective: c-Abl tyrosine kinase. Oncologist 10:758, 2005; Stem Cells 24:209, 2006. Goodsell, D.S. The molecular perspective: cisplatin. Oncologist 11:316, 2006; Stem Cells 24:514, 2006. Goodsell, D.S. The molecular perspective: double-stranded DNA breaks. Oncologist 10:361, 2005; Stem Cells 23:1021, 2005. Goodsell, D.S. The molecular perspective: RAD51 and BRCA2. Oncologist 10:555, 2005; Stem Cells 23:1434, 2005. Goodsell, D.S. The molecular perspective: tumor necrosis factor. Oncologist 11:83, 2006. Goodsell, D.S. Recognition in action: DNA mimicry. J. Mol. Recognit. 18:427, 2005. Goodsell, D.S. Representing structural information. In: Current Protocols in Bioinformatics. Baxeranis, A.D., Davison, D.B. (Eds.). Wiley & Sons, Hoboken, NJ, 2005, p. 5.4.1. Huey, R., Morris, G.M., Olson, A.J., Goodsell, D.S. A semi-empirical free energy force field with charge-based desolvation. J. Comput. Chem., in press. Rogers, J.P., Beuscher, A.E. IV, Flajolet, M., McAvoy, T., Nairn, A.C., Olson, A.J., Greengard, P. Discovery of protein phosphatase 2C inhibitors by virtual screening. J. Med. Chem. 49:1658, 2006. Sanner, M., Stolz, M., Burkhard, P., Kong, X.-P., Min, G., Sun, T.-T., Driamov, S., Aebi, U., Stoffler, D. Nature at work from the nano to the macro scale. Nanobiotechnology 1:7, 2005. Predicting Protein Structure, Association, and Inhibitors R. Abagyan, J. An,* W. Bisson, A. Cheltsov, K. Hyun, J. Kovacs, I. Kufareva, P. Lam,** G. Nicola, A. Saldanha * Genome Sciences Centre, Vancouver, British Columbia ** Molsoft L.L.C., La Jolla, California oday the Protein Data Bank contains more than 37,000 structures and is growing at a rate of 20 per day. These structures provide a unique opportunity for functional studies and rational design of therapeutic agents. We continue to focus on anno- T THE SCRIPPS RESEARCH INSTITUTE 199 tating and characterizing the protein structures in terms of their interaction interfaces and flexibility; predicting protein associations; modeling homologous structures and membrane proteins; predicting conformational rearrangements; and, finally, using ligand docking and virtual screening to detect inhibitors of specific molecular targets. This past year, our efforts in the last area led to new or improved inhibitors against the receptor for epidermal growth factor (EGFR), anthrax lethal factor, dynamin, α1-antitrypsin, and the androgen receptor. B I O I N F O R M AT I C S A N D C H E M I N F O R M AT I C S We helped G. Siuzdak and his group, Department of Molecular Biology, build a cheminformatics system for characterizing metabolites on the basis of liquid chromatography–mass spectrometry data. The resulting software (XCMS) incorporates novel nonlinear retention time alignment, matched filtration, peak detection, and peak matching and is freely available from http://metlin.scripps.edu/download/. The software helps identify changes in specific endogenous metabolites, such as potential biomarkers. We have proposed a method for sharing chemical information in conjunction with data on experimental compounds without revealing the identity of the compounds. Privacy of chemical structure is of paramount importance in the industrial sector, and the proposed solution opens a way to transfer a rich knowledge base from the pharmaceutical industry to academia. Finally, we collaborated with scientists at the Structural Genomics Consortium, Oxford, England, to improve the way the new structures are annotated, distributed, and animated by using internal coordinates–based methods. LIGAND DISCOVERY Small-molecule therapeutic agents can be discovered by using docking and virtual chemical library screening. The docking technology can also help in understanding structural mechanisms of action of small molecules and rational design of better molecules. However, modeling protein flexibility and ligand-induced conformational changes is a major challenge. We modeled the induced receptor rearrangements at several levels, including relevant normal modes combined with full side-chain sampling and “minus-one” calculations. In particular, we used the developed ligand-induced receptor simulation techniques to identify new antagonists of the androgen receptor and the first small-molecule inhibitors of α1-antitrypsin polymerization. Our docking-based in silico chemical library screening against the EGFR tyrosine kinase and the consequent 200 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE experimental validation allowed identification of several compounds with antiproliferative effects on cancer cells. Among them, a C(4)-N(1)-substituted pyrazolo[3,4d]pyrimidine inhibits EGFR tyrosine kinase activity at micromolar concentrations. We screened a library of tyrphostins against the GTPase activity of dynamin I and performed optimization of discovered compounds. The results yielded a number of promising inhibitors that are effective at micromolar concentrations. Using a fragment-based approach, we developed inhibitors of the lethal factor metalloproteinase of Bacillus anthracis. The discovered compounds are highly potent and selective against lethal factor in in vitro assays, including cell-based assays. PEPTIDE DOCKING AND STRUCTURE PREDICTION Predicting partial protein structure or molecular association is a critical task of computational biology, which remains a focus of our research. In particular, we developed a method for ab initio prediction of peptide-MHC binding geometry for diverse class I MHC allotypes. Such models are useful for predicting specific ternary complexes with T-cell receptors and for designing new molecules that interact with these complexes. The surprisingly accurate prediction (0.75-Å backbone root mean square deviation) that we achieved by using our method for cross-docking of a highly flexible decapeptide, dissimilar to the original bound peptide, and docking predictions with homology models for 2 allotypes with mean backbone root mean square deviations of less than 1.0 Å illustrate the effectiveness of the method. PREDICTING FUNCTIONAL SITES Functional annotation of protein structures involves identifying and characterizing protein-protein interfaces, oligomerization states, and binding sites for small ligands. We developed a method called protein interface recognition that can be used to predict interfaces on the basis of an isolated protein structure and does not depend on evolutionary information. The method was benchmarked by using a diverse set of 748 protein interfaces. The accuracy and efficiency make the method a suitable tool for automated high-throughput annotation of protein structures discovered in structural proteomics studies (Fig. 1). Some protein interfaces can safely be targeted for drug discovery. We developed a systematic approach to assessing the “druggability” of a protein interface. The approach includes detecting a suitable ligand-binding pocket with maximal confidence in a functionally F i g . 1 . Predicting protein oligomerization geometry by using protein interface recognition. sensitive location on the biomolecule and assessing the reliability of the local structure. This approach was applied to the Skp1-Cullin-F-box protein ubiquitin ligase interface. It can be used before high-throughput or virtual library screening. CD59 is a membrane glycoprotein with therapeutic potential for treatment of inflammatory conditions. Using scanning mutagenesis, refined nuclear magnetic resonance models, and additional site-specific mutations, we identified a binding interface on CD59 that is much broader than previously thought. We identified substitutions that decreased CD59 activity and a surprising number of substitutions that enhanced it. On the basis of these findings, we prepared clinically relevant soluble mutant CD59-based proteins that had up to a 3-fold increase in complement inhibitory activity. PUBLICATIONS Abagyan, R. Problems in computational structural genomics. In: Structural Proteomics. Sundstrom, M., Norin, M., Edwards, A. (Eds.). CRC Press, Boca Raton, FL, 2006, p. 223. Abagyan, R., Lee, W.H., Raush, E., Budagyan, L., Totrov, M., Sundstrom, M., Marsden, B.D. Disseminating structural genomics data to the public: from a data dump to an animated story. Trends Biochem. Sci. 31:76, 2006. Bordner, A., Abagyan, R.A. Ab initio prediction of peptide-MHC binding geometry for diverse class I MHC allotypes. Proteins 63:512, 2006. Cardozo, T., Abagyan, R. Druggability of SCF ubiquitin ligase-protein interfaces. Methods Enzymol. 399:634, 2005. Cavasotto, C.N., Orry, A.J.W., Abagyan, R. Receptor flexibility in ligand docking. In: Handbook of Theoretical and Computational Nanotechnology. Rieth, M., Schommers, W. (Eds.). American Scientific Publishers, Stevenson Ranch, CA, 2006, Vol.6, p. 217. Cavasotto, C.N., Orry, A.J.W., Abagyan, R.A. The challenge of considering receptor flexibility in ligand docking and virtual screening. Curr. Comput. Aided Drug Des. 1:423, 2005. MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 201 Cavasotto, C.N., Ortiz, M.A., Abagyan, R.A., Piedrafita, F.J. In silico identification of novel EGFR inhibitors with antiproliferative activity against cancer cells. Bioorg. Med. Chem. Lett. 16:1969, 2006. Forino, M., Johnson, S., Wong, T.Y., Rozanov, D.V., Savinov, A.Y., Li, W., Fattorusso, R., Becattini, B., Orry, A.J., Jung, D., Abagyan, R.A., Smith, J.W., Alibek, K., Liddington, R.C., Strongin, A.Y., Pellecchia, M. Efficient synthetic inhibitors of anthrax lethal factor. Proc. Natl. Acad. Sci. U. S. A. 102:9499, 2005. Hill, T., Odell, L.R., Edwards, J.K., Graham, M.E., McGeachie, A.B., Rusak, J., Quan, A., Abagyan, R., Scott, J.L., Robinson, P.J., McCluskey, A. Small molecule inhibitors of dynamin I GTPase activity: development of dimeric tyrphostins. J. Med. Chem. 48:7781, 2005. Huang, Y., Smith, C.A., Song, H., Morgan, B.P., Abagyan, R., Tomlinson, S. Insights into the human CD59 complement binding interface toward engineering new therapeutics. J. Biol. Chem. 280:34073, 2005. Kovacs, J.A., Cavasotto, C.N., Abagyan, R.A. Conformational sampling of protein flexibility in generalized coordinates: application to ligand docking. J. Comput. Theor. Nanosci. 2:354, 2005. Kufareva, I., Budagyan, L., Raush, E., Totrov, M., Abagyan, R. PIER: protein interface recognition for structural proteomics. Proteins, in press. Orry, A.J., Abagyan, R.A., Cavasotto, C.N. Structure-based development of targetspecific compound libraries. Drug Discov. Today 11:261, 2006. F i g . 1 . A novel nonlinear approach for correcting and analyzing mass spectrometry data for characterization of metabolites. V I R A L C H A R A C T E R I Z AT I O N We have developed novel methods for characterizing viruses that have applications to whole viruses, viral proteins, and viral metabolites. Our results have enabled us to examine both local and overall viral structure, gaining insight into the dynamic changes of proteins on the viral surface and the changes that occur during viral infection (Fig. 2). Smith, C.A., O’Maille, G., Want, E.J., Qin, C., Trauger, S.A., Brandon, T.R., Custodio, D.E., Abagyan, R., Siuzdak, G. METLIN: a metabolite mass spectral database. Ther. Drug Monit. 27:747, 2005. Smith, C.A., Want, E.J., O’Maille, G., Abagyan, R., Siuzdak, G. XCMS: processing mass spectrometry data for metabolite profiling using nonlinear peak alignment, matching, and identification. Anal. Chem. 78:779, 2006. Tetko, I.V., Abagyan, R., Oprea, T.I. Surrogate data: a secure way to share corporate data. J. Comput. Aided Mol. Des. 19:749, 2005. Mass Spectrometry F i g . 2 . A comprehensive approach for studying viral infection by G. Siuzdak, J. Apon, H.P. Benton, E. Go, K. Harris, L. Hoang, R. Lowe, A. Meyers, H. Morita, A. Nordstrom, T. Northen, G. O’Maille, C. Qin, Z. Shen, C. Smith, M. Sonderegger, S. Trauger, W. Uritboonthai, E. Want, W. Webb, W. Wikoff, D. Wong M E TA B O L I T E P R O F I L I N G ndogenous small-molecule metabolites, ubiquitous in biofluids, are crucial elements in understanding living organisms whether in fundamental biochemistry, disease diagnosis, or drug toxicity. The inherent advantage of monitoring small molecules rather than proteins is the relative ease of quantitative analysis of the molecules with mass spectrometry. We are implementing novel mass spectrometry and bioinformatics techniques (Fig. 1) to investigate the profile of small-molecule metabolites. Our purposes are to correlate metabolite activity with protein regulation and to develop metabolite analysis as a diagnostic method. Our ultimate goal is to create analytical and chemical technologies and data management approaches to identify and structurally characterize metabolites of physiologic importance. E using a combination of mass spectrometry techniques. Three different aspects of viral infection within an infected cell—the expression kinetics of the viral proteins, changes in the expression levels of cellular proteins, and changes in cellular metabolites—were monitored. These analyses reveal the complexity of the protein and metabolite regulation involved in cellular transformations that occur during viral infection. MASS SPECTROMETRY IN SILICO We are also developing ultra-high-sensitivity approaches in mass spectrometry with a new strategy that involves pulsed laser desorption/ionization from a silylated silicon surface. In desorption/ionization on silicon, silicon is used to capture analytes and laser radiation is used to vaporize and ionize these molecules. Using this technology, we can analyze a wide range of molecules with unprecedented sensitivity, in the yoctomole range. PUBLICATIONS Cohen, L., Go, E.P., Siuzdak, G. Small-molecule desorption/ionization mass analysis. In: A Practical Guide to MALDI MS: Instrumentation, Methods and Applications. Hillenkamp, F., Peter-Katalinic, J. (Eds.). Wiley & Sons, New York, in press. 202 MOLECULAR BIOLOGY 2006 Lee, J.-C.,Wu, C.-Y., Apon, J.V., Siuzdak, G., Wong, C.-H. Reactivity-based onepot synthesis of tumor-associated antigen N3 minor octasaccharide for the development of a cleavable DIOS-MS sugar array. Angew. Chem. Int. Ed. 45:2753, 2006. Lowe, R., Tong, G., Voelcker, N.H., Siuzdak, G. Monitoring EDTA and endogenous metabolite from serum with mass spectrometry. Spectroscopy 19:137, 2005. Luo, G., Chen, Y., Siuzdak, G., Vertes, A. Surface modification and laser pulse length effects on internal energy transfer in DIOS. J. Phys. Chem. B. 109:24450, 2005. Nordstrom, A., Apon, J.V., Uritboonthai, W., Go, E.P., Siuzdak, G. Surfactant enhanced desorption/ionization on silicon mass spectrometry. Anal. Chem. 78:272, 2006. Nordstrom, A., He, L., Siuzdak, G. Desorption/ionization on silicon (DIOS). In: Hyphenation Methods. Niessen, W. (Ed.). Elsevier, St. Louis, in press. Encyclopedia of Mass Spectrometry, Vol 8. Gross, M.L., Caprioli, R.M. (Eds. in Chief). Nordstrom, A., O’Maille, G., Qin, C., Siuzdak, G. Nonlinear data alignment for UPLC-MS and HPLC-MS based metabolomics: quantitative analysis of endogenous and exogenous metabolites in human serum. Anal. Chem. 78:3289, 2006. Siuzdak, G. The Expanding Role of Mass Spectrometry in Biotechnology, 2nd ed. MCC Press, San Diego, CA, 2006. Smith, C.A., O’Maille, G., Want, E.J., Qin, C., Trauger, S.A., Brandon, T.R., Custodio, D.E., Abagyan, R., Siuzdak, G. METLIN: a metabolite mass spectral database. Ther. Drug Monit. 27:747, 2005. Smith, C.A., Want, E.J., O’Maille, G., Abagyan, R., Siuzdak, G. XCMS: processing mass spectrometry data for metabolite profiling using nonlinear peak alignment, matching, and identification. Anal. Chem. 78:779, 2006. Talkington, M.W., Siuzdak, G., Williamson, J.R. An assembly landscape for the 30S ribosomal subunit. Nature 438:628, 2005. Want, E., Cravatt, B.F., Siuzdak, G. The expanding role of mass spectrometry in metabolite profiling and characterization. Chembiochem 6:1941, 2005. THE SCRIPPS RESEARCH INSTITUTE We using a wide variety of biophysical techniques to study the mechanism of assembly of the 30S ribosome in vitro. The 30S ribosome can be reconstituted from purified components in vitro with extremely high efficiency, a characteristic that has enabled detailed mechanistic studies. The 30S ribosome is composed of a single RNA chain of approximately 1500 nucleotides and 20 small proteins of 8–20 kD. Pioneering work by Nomura more than 30 years ago led to the development of an assembly map that outlines the basic order of protein binding. However, the mechanistic basis for these early observations was unknown. Using nuclear magnetic resonance, x-ray crystallography, calorimetry, and fluorescence methods, we have studied the details of assembly of small RNA-protein complexes derived from the 30S subunit to elucidate these molecular events. A guiding principle for ribosome assembly is that each protein recognizes a small local region of the RNA as its binding site. The protein cannot bind until the RNA structure in its binding site is properly folded. We showed that the assembly reaction can be considered an alternating series of RNA conformational changes and protein binding events (Fig. 1). RNA helices must Want, E.J., O’Maille, G., Smith, C.A., Brandon, T.R., Uritboonthai, W., Qin, C., Trauger, S.A., Siuzdak, G. Solvent-dependent metabolite distribution, clustering, and protein extraction for serum profiling with mass spectrometry. Anal. Chem. 78:743, 2006. Assembly Landscape of the 30S Ribosome J.R. Williamson, F. Agnelli, A. Beck, C. Beuck, A. Bunner, A. Carmel, J. Chao, S. Edgcomb, M. Hennig, E. Johnson, D. Kerkow, E. Kompfner, S. Kwan, P. Mikulecky, W. Ridgeway, H. Schultheisz, L.G. Scott, E. Sperling, B. Szymczyna he ribosome is a large molecular machine that is responsible for synthesis of all proteins in the cell. It is composed of 2 multicomponent subunits that bind mRNA, tRNAs, and other factors to carry out translation of the genetic code from RNA into protein product. In bacteria, the large, or 50S, subunit is responsible for catalyzing the formation of peptide bonds, whereas the small, or 30S, subunit is responsible for reading out the genetic code. An elaborate process exists for biogenesis of the ribosome machinery in cells to assemble the ribosome from individual components. T F i g . 1 . A model for ribosome assembly. The RNA chain is shown as cylinders representing helical regions of RNA structure. In the first step, the RNA changes conformation, which creates a protein-binding site for the protein S15. Next, a subsequent RNA folding event occurs, which in turn creates a binding site for the proteins S6 and S18. Assembly appears to proceed as an alternating series of folding and binding events. be properly arranged to create the binding site for the first protein, which is protein S15 in Figure 1. Binding of S15 effectively consolidates the gains from RNA folding in the previous step. Furthermore, after S15 binding, the next RNA conformational change is facilitated; this change sets up the binding site for the next proteins, which are S6 and S18 in Figure 1. Thus, each protein serves as a local reporter for RNA folding in a specific region of the 30S subunit. The overall assembly reaction can be schematically illustrated as shown in Figure 2, where an unfolded RNA chain is combined with 20 different proteins, a change that after a complex series of RNA conforma- MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 203 a number of parallel pathways exist by which the ribosome can assemble. In addition to the functional restrictions placed on the sequence of the RNA, most likely the sequence is also selected under evolutionary pressure to fold efficiently under a variety of conditions encountered by bacteria in the environment. F i g . 2 . The 30S ribosome assembly reaction. The RNA chain is represented as a thin line that is disordered at the beginning of the experiment. The 20 small proteins that bind to the RNA are represented as circles. The final assembled subunit is composed of highly folded RNA with each protein bound at a specific location. tional changes and protein-binding events results in the structured 30S subunit. Our previous analyses involved fragments of the overall structure, and we were interested in monitoring the kinetics of binding and assembly of the intact 30S subunits. Monitoring the simultaneous binding of 20 different proteins to an RNA molecule is a serious technical challenge. To surmount this challenge, we developed an isotope-pulse chase assay in which mass spectrometry is used to indicate binding of the proteins to the RNA. Assembly is initiated by using a pulse of a mixture of the 20 15 N-labeled proteins; after a short assembly time, a mixture of 14 N-proteins is added as the chase. The fully assembled subunits are isolated, and the fraction of 15N for each protein is measured as a function of the pulse time by using quantitative mass spectrometry. In this way, the time course of binding for each protein can be measured. The strength of the method is that the binding rates can all be measured simultaneously. Using this method, we can perform mechanistic experiments on 30S ribosome assembly by using the kinetic tools of physical chemistry. We have varied the protein concentration to show that the binding rates correspond to a bimolecular association, not to a ratelimiting RNA conformational change. We have varied the magnesium ion concentration to show that ions can play 2 opposing roles during assembly. Some parts of the 30S subunit speed up at lower magnesium concentrations, and different parts slow down. Perhaps most important, we have measured the rates as a function of temperature and performed Arrhenius analysis of the activation energies for binding. The main conclusion from these studies is that 30S assembly has no single global rate-limiting step. Rather, PUBLICATIONS Chao, J.A.., Lee, J.H., Chapados, B.R., Debler, E.W., Schneemann, A., Williamson, J.R. Dual modes of RNA-silencing suppression by Flock House virus protein B2. Nat. Struct. Mol. Biol. 12:952, 2005. Davis, J.H., Tonelli, M., Scott, L.G., Jaeger, L., Williamson, J.R., Butcher, S.E. RNA helical packing in solution: NMR structure of a 30 kDa GAAA tetraloop-receptor complex [published correction appears in J. Mol. Biol. 760:742, 2006]. J. Mol. Biol. 351:371, 2005. Hennig, M., Munzarova, M.L., Bermel, W., Scott, L.G., Sklenar, V., Williamson, J.R. Measurement of long-range 1H-19F scalar coupling constants and their glycosidic torsion dependence in 5-fluoropyrimidine-substituted RNA. J. Am. Chem. Soc. 128:5851, 2006. Scott, L.G., Williamson, J.R. The binding interface between Bacillus stearothermophilus ribosomal protein S15 and its 5′-translational operator mRNA. J. Mol. Biol. 351:280, 2005. Talkington, M.T., Siuzdak, G., Williamson, J.R. An assembly landscape for the 30S ribosomal subunit. Nature 438:628, 2005. Development of the Genetic Code and Its Connection to Human Disease P. Schimmel, J. Bacher, K. Beebe, Z. Druzina, K. Ewalt, M. Kapoor, E. Merriman, C. Motta, L. Nangle, F. Otero, J. Reader, R. Reddy, M. Swairjo, K. Tamura, E. Tzima, W. Waas, X.-L. Yang he genetic code is thought to have developed in the putative RNA world and thereby enabled the transition to the modern world of proteins. The early code was primitive and over many eons was refined. This refinement came from the acquisition of new activities by a group of proteins known as aminoacyl-tRNA synthetases. These proteins established the rules of the code through aminoacylation reactions, whereby each of the 20 amino acids is covalently joined to its cognate tRNA. The tRNA harbors the genetic code triplet associated with the specific amino acid that is joined to the tRNA. Each amino acid has a single tRNA synthetase. The synthetases are thought to be among the earliest proteins, essential components of the translation apparatus that established the genetic code and that were present in the last common ancestor of the universal T 204 MOLECULAR BIOLOGY 2006 tree of life. As the tree developed and branched into the 3 great kingdoms—archaebacteria, bacteria, and eukaryotes—the enzymes were incorporated into every cell type of every organism. Detailed investigations of the structures and evolution of the aminoacyl-tRNA synthetases have provided a picture of the development of the genetic code and how the development was directed by the evolution of the synthetases and tRNAs. In previous research, we focused on the specifics of the molecular recognition of tRNAs and how the enzymes distinguish one tRNA from another to achieve accurate aminoacylation for a precise genetic code. During these studies, examination of a recent crystal structure of human tryptophanyl-tRNA synthetase (TrpRS) in complex with the tRNA for tryptophan (tRNATrp) revealed 2 states of the enzyme-tRNA complex (Fig. 1). In one state, the tRNA is entering the THE SCRIPPS RESEARCH INSTITUTE Recently, we showed how the error-correction activity is essential for maintaining cell viability and how defects in this activity can lead to disease. In collaboration with S.L. Ackerman, Jackson Laboratories, Bar Harbor, Maine, we found that a single point mutation in mice leads to neurodegeneration (Fig. 2). In particular, Purkinje cells F i g . 1 . Crystal structures of uncharged tRNA Trp associating with TrpRS (A) and charged tRNATrp dissociating from the enzyme (B). The location of the bound free amino acid (Trp) in the 2 active sites of the homodimer is indicated. The tRNA binds across both subunits. active site. In the other state, it has been charged (that is, tryptophan has been joined to tRNATrp in the aminoacylation reaction) and is dissociating from the enzyme. During the long evolutionary development of aminoacyl-tRNA synthetases and their populating of every cell type, the enzymes adopted novel functions while keeping their canonical role as determinants of the genetic code. Related to their central role, the enzymes acquired novel domains enabling them to correct errors of aminoacylation and thereby ensure the stringent accuracy of the code. Unrelated to the canonical activities of the enzymes in translation, the expanded functions include regulation of transcription and translation in bacteria, RNA splicing in fungal organisms, and cytokine signaling in mammalian cells. These novel functions connect translation to other central pathways that control growth, development, and regulation of all cell types. F i g . 2 . Pathologic changes in sticky mutant mice (A). B-D, Cal- bindin D-28 (Calb) immunohistochemistry of sagittal sections of cerebella from 3-week-old (B), 6-week-old (C), and 12-month-old (D) sti/sti mutant and 12-month-old wild-type (WT; E) mice. Cerebellar lobules are indicated by roman numerals. F-H, Hematoxylin and eosin staining of Purkinje cells (arrowheads) in lobule II of cerebella from 1-month-old (F) or 12-month-old (G) sti/sti mutant and 12-month-old wild-type (H) mice. I-N, Cleaved caspase 3 (Casp3) immunohistochemistry (I-K) and TUNEL analysis (L-N) of cerebella from 4-week-old mutant mice. Scale bars: For B-E, 500 µm; F-H, 50 µm; I-N, 10 µm. in the brain deteriorate and ataxia develops. This simple, heritable mutation is due to small amounts of misacylation of alanine-specific tRNA (tRNAAla) to generate, for example, serine attached to the tRNA. In this instance, small amounts of serine are incorporated in place of alanine in the polypeptides that are produced. Thus, a mild MOLECULAR BIOLOGY 2006 editing defect can lead to heritable neurologic disorders. A more severe defect in editing would doubtless be lethal and not sustained in the population. In other collaborative studies with R. Burgess, Jackson Laboratories, we established a connection between glycyl-tRNA synthetase and Charcot-Marie-Tooth disease. A single point mutation in the synthetase leads to the disease. This mutation does not affect the aminoacylation activity of glycyl-tRNA synthetase. Instead, the results suggest that glycyl-tRNA synthetase has an additional function, possibly in neurologic development. Several examples of Charcot-Marie-Tooth disease due to mutations in glycyl-tRNA synthetase in humans have been found. To better understand the molecular origins of this disease, we obtained crystals of human glycyl-tRNA synthetase that diffract to about 3 Å. A structure is being determined, and mutations found in humans will be mapped on the structure. This information will be used in conjunction with other assays and experiments to understand the connection between neurologic development, Charcot-Marie-Tooth disease, and glycyl-tRNA synthetase. These results with glycyl-tRNA synthetase support the hypothesis that aminoacyl-tRNA synthetases in mammals are not only components of the translation apparatus but also a reservoir of cytokines with activities that are unmasked by specific activation events, such as alternative splicing or generation of specific fragments by proteolysis. Examples we are studying include tyrosyl- and tryptophanyl-tRNA synthetases. These are both procytokines that when split by alterative splicing or natural proteolysis, result in fragments that are active in signal transduction pathways. For example, a fragment of tryptophanyl-tRNA synthetase is a potent angiostatic agent. PUBLICATIONS Lee, J.W., Beebe, K., Nangle, L.A., Jang, J., Longo-Guess, C.M., Cook, S.A., Davisson, M.T., Sundberg, J.P., Schimmel, P., Ackerman, S.L. Editing-defective tRNA synthetase causes protein misfolding and neurodegeneration in the sticky mouse. Nature 443:50, 2006. Nangle, L.A., Motta, C.M., Schimmel, P. Global effects of mistranslation from an editing defect in mammalian cells. Chem. Biol. 13:1091, 2006. Reader, J.S., Ordoukhanian, P.T., Kim, J.-G., de Crécy-Lagard, V., Hwang, I., Farrand, S., Schimmel, P. Major biocontrol of plant tumors targets tRNA synthetase [published correction appears in Science 310:54, 2005]. Science 309:1533, 2005. Schimmel, P., Beebe, K. From the RNA world to the theatre of proteins. In: The RNA World, 3rd ed. Gesteland, R.R., Cech, T.R. Atkins, J.F. (Eds.). Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2005, p. 227. Seburn, K.L., Nangle, L.A., Cox, G.A., Schimmel, P., Burgess, R.W. An active dominant mutant of glycyl-tRNA synthetase causes neuropathy in Charcot-MarieTooth 2D mouse model. Neuron 51:715, 2006. THE SCRIPPS RESEARCH INSTITUTE 205 Swairjo, M.A., Reddy, R.R., Lee, B., Van Lanen, S.G., Brown, S., de CrécyLagard, V., Iwata-Reuyl, D., Schimmel, P. Crystallization and preliminary x-ray characterization of the nitrile reductase QueF: a queosine-biosynthesis enzyme. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 61(Pt. 10):945, 2005. Tamura, K., Schimmel, P.R. Chiral-selective aminoacylation of an RNA minihelix: mechanistic features and chiral suppression. Proc. Natl. Acad. Sci. U. S. A. 103:13750, 2006. Tzima, E., Schimmel, P. Inhibition of tumor angiogenesis by a natural fragment of a tRNA synthetase. Trends Biochem. Sci. 31:7, 2006. Waas, W.F., de Crécy-Lagard, V., Schimmel, P. Discovery of a gene family critical to wyosine base formation in a subset of phenylalanine-specific transfer RNAs. J. Biol. Chem. 280:37616, 2005. Yang, X.-L., Otero, F.J., Ewalt, K.L., Liu, J., Swairjo, M.A., Kohrer, C., RajBhandary, U.L., Skene, R.J., McRee, D., Schimmel, P. Two conformations of a crystalline human tRNA synthetase-RNA complex: implications for protein synthesis. EMBO J. 25:2919, 2006. Mechanisms of RNA Assembly and Catalysis M.J. Fedor, E.M. Calderon, J.W. Cottrell, C.P. Da Costa, S. Daudenarde, J.W. Harger, Y.I. Kuzmin, E.M. Mahen, M. Roychowdhury-Saha ur goal is to generate basic insights into catalysis by RNA and RNA-protein enzymes, RNA folding, and RNA interactions with small molecules. In addition to contributing basic knowledge of RNA structure and function in normal growth and development, results of our studies provide a framework for developing technical and therapeutic applications involving RNAs as targets and reagents. Apart from the ribosome, which catalyzes peptidyl transfer, the naturally occurring ribozymes catalyze transfer of phosphate groups. The small RNA enzymes that we study catalyze reversible phosphodiester cleavage reactions that generate 5′ hydroxyl and 2′,3′-cyclic phosphate termini (Fig. 1). Possible strategies for catalysis of phosphoryl transfer reactions include aligning reactive groups in an optimal orientation for an in-line attack mechanism, general acid-base catalysis of proton transfer to activate nucleophilic oxygens or to stabilize oxyanionleaving groups, electrostatic stabilization of negative charge that accumulates in the transition state, and destabilizing the ground state. Our goal is to understand which of these catalytic strategies RNA enzymes use. In contrast to the chemical versatility of the amino acid side chains that make up the active sites of protein enzymes, just 4 nucleotides are available for the construction of ribozyme active sites. Nucleotides are well suited to faithful storage and transmission of genetic O 206 MOLECULAR BIOLOGY 2006 F i g . 1 . Chemical mechanism of RNA cleavage mediated by the family of small catalytic RNAs that includes the hairpin ribozyme. Cleavage of the phosphodiester bond occurs through an SN2-type mechanism that involves in-line attack of the 2′ oxygen nucleophile on the adjacent phosphorus to form a trigonal bipyramidal transition state. Breaking of the 5′ oxygen-phosphorus bond generates products with 5′ hydroxyl and 2′,3′-cyclic phosphate termini. information through complementary base pairing, but they are not particularly adept at catalytic chemistry. Protonation and deprotonation of nucleotides occur at high or low pH extremes, a situation that would make it difficult to mediate general acid or base catalysis at neutral pH. No positively charged nucleotide functional groups are expected to be available at neutral pH to function as Lewis acids to activate a nucleophile or stabilize an electronegative transition state or an oxyanion-leaving group. Recent high-resolution structures of self-cleaving RNAs lay the groundwork for experiments to probe fundamental questions about how RNA enzymes use their functional groups for catalysis. Like all enzymes, hairpin ribozymes combine several strategies to enhance catalytic rate. One important strategy, apparent from the crystal structures, is the alignment of nucleophilic and leaving-group oxygens in the optimal orientation for an in-line SN2-type nucleophilic attack. The structure of the hairpin ribozyme active site places guanine 8, adenine 9, adenine 10, and adenine 38 nucleobases near the reactive phosphate. Guanine 8 and adenine 38 occupy positions reminiscent of 2 histidine residues in the active site of ribonuclease A, a protein enzyme that catalyzes the same reaction. Histidine residues perform general acidbase catalysis during ribonuclease A catalysis, so the similarity between hairpin ribozyme and ribonuclease A active sites raised the possibility that guanine 8 and adenine 38 nucleobases might perform functions similar to those of histidine residues. Hairpin ribozyme activity increases with increasing pH, consistent with the notion that activity depends on the availability of the deprotonated form of guanine 8 to accept a proton from the 2′ hydroxyl nucleophile as predicted by the general acid-base catalysis model. To THE SCRIPPS RESEARCH INSTITUTE test this model, we replaced guanine 8 with an abasic residue, a substitution that eliminates the nucleobase but leaves the phosphodiester backbone intact. However, this abasic variant had the same pH dependence as an unmodified ribozyme, arguing that the pH transition does not involve guanine 8. Replacing adenine 38 with an abasic residue, on the other hand, did eliminate the pH-dependent transition in activity, implicating adenine 38 in a catalytically important deprotonation. These and other results are consistent with 2 models of the hairpin ribozyme catalytic mechanism in which adenine 38 contributes either general acid-base catalysis (Fig. 2A) or electrostatic stabilization of negative F i g . 2 . Two models of hairpin ribozyme catalysis. Results of mech- anistic studies of the hairpin ribozyme are consistent with 2 models in which the functional form of adenine 38 is either protonated or unprotonated. In the first model (A), protonated adenine 38 would act as a general acid by donating a proton to the 5′ oxygen, acting in concert with hydroxide ion that activates the 2′ oxygen nucleophile during cleavage, and unprotonated adenine 38 would act as a general base to activate the 5′ oxygen nucleophile during ligation. In the second model (B), unprotonated adenine 38 accepts a hydrogen bond from the 5′ hydroxyl nucleophile during ligation and accepts a hydrogen bond from a protonated bridging 5′ oxygen during cleavage, providing electrostatic stabilization to developing negative charge. In both models, the amidine group of guanine 8, in its protonated form, donates hydrogen bonds to the 2′ and phosphoryl oxygens that stabilize negative charge that develops in the transition state and positions reactive groups in the orientation appropriate for an SN2 in-line nucleophilic attack. Reproduced with permission from Fedor, M.J., Williamson, J.R. The catalytic diversity of RNAs. Nat. Rev. Mol. Cell Biol. 6:399, 2005. Copyright 2005 Nature Publishing Group/Macmillan Magazines Ltd. charge that develops in the transition state as 5 electronegative oxygen atoms from transient bonds with MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 207 phosphorus (Fig. 2B) and guanine 8 donates hydrogen bonds to stabilize the transition state electrostatically. Directed Evolution of Nucleic Acid Enzymes G.F. Joyce, S.E. Hamilton, D.P. Horning, T.A. Jackson, G.C. Johns, B.J. Lam, B.M. Paegel, G.G. Springsteen, S.B. Voytek ll life known to exist on Earth today is based on DNA genomes and protein enzymes, but most likely it was preceded by a simpler form of life based on RNA. This earlier era is referred to as the “RNA world.” During that time, genetic information resided in the sequence of RNA molecules and phenotype was derived from the catalytic behavior of RNA. By studying the properties of RNA in the laboratory, especially with regard to the evolution of catalytic function, we can gain insight into the RNA world. In addition, we can develop novel nucleic acid enzymes that have applications in biology and medicine. A CONVERTING AN RNA ENZYME TO A DNA ENZYME The transfer of sequence information between 2 different classes of nucleic acid–like molecules, for example between RNA and DNA, is straightforward because it relies on the 1-to-1 correspondence of Watson-Crick pairing. Nearly 50 years ago, in articulating the central dogma of molecular biology, Francis Crick referred to this property as “sequentialization.” Sequentialization also applies to the transfer of information from RNA to protein via the genetic code. The transfer of function, however, is more difficult because function is an overall property of a macromolecule and cannot be conveyed in a sequential manner. There is no known example of an RNA enzyme that retains catalytic activity when prepared as the corresponding DNA molecule, and vice versa. We used in vitro darwinian evolution to convert an RNA enzyme to a DNA enzyme of the same function, after the acquisition of a few critical mutations. The starting RNA had the ability to join 2 RNA substrates in a template-directed manner, with a catalytic rate of 0.14 min–1 (Fig. 1A). A corresponding DNA molecule in which ribose was replaced by deoxyribose and uracil was replaced by thymine had no detectable activity. The DNA molecule was used as a starting point to generate trillions of randomized variants, which were selected for the ability to catalyze the RNA-joining reaction. After 10 F i g . 1 . Composition of an RNA enzyme (A) and a DNA enzyme (B) related by evolutionary descent. Both enzymes contain about 50 nucleotides and catalyze the joining of 2 RNA substrates (S1 and S2). The evolved DNA enzyme contains 10 mutations relative to the starting RNA enzyme (highlighted with black circles). generations of evolution, we obtained a population of DNA enzymes with the desired activity. A typical example contains 10 mutations relative to the starting sequence and has a catalytic rate of 0.052 min–1 (Fig. 1B). When this DNA enzyme was prepared as the corresponding RNA enzyme, it had no detectable activity. Thus, the evolutionary transition from an RNA enzyme to a DNA enzyme represents a switch in the chemical basis of catalytic function. Evolutionary pathways such as this one for conversion of an RNA enzyme to a DNA enzyme may exist between other classes of nucleic acid–like molecules. The RNA world may have been preceded by a simpler “pre-RNA world” based on a nucleic acid–like molecule that would have occurred more readily on the primitive Earth. Our findings suggest that the catalytic function of a pre-RNA molecule might have been transferred to a corresponding RNA enzyme through darwinian evolution. CONTINUOUS EVOLUTION OF RNA ENZYMES Processes of darwinian evolution are fundamental to understanding biological form and function but are difficult to appreciate on the human timescale. During the past decade, we have developed methods for evolving molecules rapidly and under controlled laboratory conditions. One of the most powerful of these methods, and the one that most closely resembles biological evolution, is a system for the continuous in vitro evolution of RNA enzymes. It involves a population of RNA enzymes that catalyze an RNA-joining reaction. Any molecule in the population that performs the reaction becomes amplified to produce “progeny” molecules, which then have the opportunity to perform the reaction again. The entire process takes place within a common reaction mixture and can be continued indefinitely, so long as an adequate supply of reaction materials is maintained. 208 MOLECULAR BIOLOGY 2006 Recently, we developed a novel approach for the continuous evolution of RNA enzymes that uses microfluidic technology. With this approach, evolution can be carried out in an automated fashion under computer control, with continuous monitoring of the population size and precise control over critical parameters such as mutation frequency and selection pressure. We have used the microfluidic device to conduct evolution experiments, beginning with a reaction mixture containing about 1 billion RNA enzymes and carrying out repeated rounds of RNA catalysis and selective amplification in an automated fashion. The amount of RNA is monitored continuously by using a confocal laser fluorescence microscope. When a predetermined threshold concentration is reached, the computer initiates an automated dilution and provides a fresh supply of reagents. This process of selective amplification and dilution was carried out for 70 successive dilutions of 10-fold each during a period of 6.5 hours. The microfluidic system is now being used to address fundamental questions of macromolecular evolution, such as the role of genetic diversity in escaping evolutionary bottlenecks and the maximum frequency of mutation that can be tolerated by an evolving population. THE SCRIPPS RESEARCH INSTITUTE Nature’s strategies to prepare novel molecules that perform specific functional tasks, such as regulating a gene, destroying cancer, or catalyzing a reaction with enzymelike efficiency. We hope to apply these novel insights, technologies, methods, and products to provide solutions to human diseases, including cancer, HIV disease, and genetic diseases. D I R E C T I N G T H E E V O L U T I O N O F C ATA LY T I C F U N C T I O N Using our concept of reactive immunization, we have developed antibodies that catalyze aldol as well as retro-aldol reactions of a wide variety of molecules. The catalytic proficiency of the best of these antibodies approaches 1014, a value 1000 times that of the best catalytic antibodies reported to date and overall the best of any synthetic protein catalyst. We have shown the efficient asymmetric synthesis and resolution of a variety of molecules, including tertiary and fluorinated aldols, and have used these chiral synthons to synthesize natural products (Fig. 1). The results highlight the poten- PUBLICATIONS Joyce, G.F., Orgel, L.E. Progress toward understanding the origin of the RNA world. In: The RNA World, 3rd ed. Gesteland, R.F., Cech, T.R., Atkins, J.F. (Eds.). Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 2006, p. 23. Oberhuber, M., Joyce, G.F. A DNA-templated aldol reaction as a model for the formation of pentose sugars in the RNA world. Angew. Chem. Int. Ed. 44:7580, 2005. Paul, N., Springsteen, G., Joyce, G.F. Conversion of a ribozyme to a deoxyribozyme through in vitro evolution. Chem. Biol. 13:329, 2006. Studies at the Interface of Molecular Biology, Chemistry, and Medicine C.F. Barbas III, M. Ahmad, K. Albertshofer, L. Asawapornmongkul, N.S. Chowdari, S. Eberhardy, R. Fuller, B. Gonzalez, R. Gordley, J. Guo, D.H. Kim, R.L. Lerner, C. Lund, J. Mandell, S. Mitsumori, W. Nomura, M. Popkov, D.B. Ramachary, S.S.V. Ramasastry, L.J. Schwimmer, D. Shabat,* F. Silva, J. Suri, F. Tanaka, U. Tschulena, N. Utsumi, Y. Ye, Y. Yuan, H. Zhang * Tel Aviv University, Tel Aviv, Israel W e are concerned with problems in molecular biology, chemistry, and medicine. Many of our studies involve learning or improving on F i g . 1 . A variety of compounds synthesized with the world’s first commercially available catalytic antibody, 38C2, produced at Scripps Research. tial synthetic usefulness of catalytic antibodies as artificial enzymes in addressing problems in organic chemistry that are not solved by using natural enzymes or more traditional synthetic methods. Other advances in this area include the development of the first peptide aldolase enzymes. By using both design and selection, we have created small peptide catalysts that recapitulate many of the kinetic features of large enzyme catalysts. These smaller enzymes allow us to address the relationship between the size of natural proteins and the proteins’ catalytic efficiency. MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 209 O R G A N O C ATA LY S I S : A B I O O R G A N I C A P P R O A C H T O C ATA LY T I C A S Y M M E T R I C S Y N T H E S I S To further explore the principles of catalysis, we are studying amine catalysis as a function of catalytic scaffold. Using insights garnered from our studies of aldolase antibodies, we determined the efficacy of simple chiral amines and amino acids for catalysis of aldol and related imine and enamine chemistries such as Michael, Mannich, Knoevenagel, and Diels-Alder reactions. Although aldolase antibodies are superior catalysts in terms of the kinetic parameters, these more simple catalysts are enabling us quantify the importance of pocket sequestration in catalysis. Furthermore, many these catalysts are cheap, environmentally friendly, and practical for large-scale synthesis. With this approach, we showed the scope and usefulness of the first efficient amine catalysts of direct asymmetric aldol, Mannich, Diels-Alder, and Michael reactions. The organocatalyst approach is a direct outcome of our studies of catalytic antibodies and provides an effective alternative to organometallic reactions that use severe reaction conditions and often-toxic catalysts. We think that our discovery that simple naturally occurring amino acids such as L-proline and other amines can effectively catalyze a variety of enantioselective intermolecular reactions will change the way many reactions will be performed. As a testament to the mild nature of this approach, we developed the first catalytic asymmetric Aldol, Mannich, Michael, and fluorination reactions involving aldehydes as nucleophiles. Previously, such reactions were considered out of the reach of traditional synthetic methods. In extensions of these concepts, we designed novel amino acid derivatives that direct the stereochemical outcome of reactions in ways not possible with proline (Fig. 2). In other studies, we created the first asymmetric small-molecule aldol catalysts that are highly effective with water and seawater as solvent. We think that our results are also relevant to the prebiotic synthesis of the molecules of life. For example, we have shown that our amino acid strategy can be used to synthesize carbohydrates directly, thereby providing a provocative prebiotic route to the sugars essential for life. THERAPEUTIC ANTIBODIES, IN AND OUT OF CELLS We developed the first human antibody phage display libraries and the first synthetic antibodies and methods for the in vitro evolution of antibody affinity. The ability to manipulate large libraries of human antibodies and to evolve such antibodies in the laboratory provides tremendous opportunities to develop new medi- F i g . 2 . Design of the new catalyst (3R,5R)-5-methyl-3-pyrrolidinecarboxylic acid (right) allows efficient access to anti-Mannich products not accessible through proline catalysis (left). cines. Laboratories and pharmaceutical companies around the world now apply the phage display technology that we developed for antibody Fab fragments. In our laboratory, we are targeting cancer and HIV disease. One of our antibodies, IgG1-b12, protects animals against primary challenge with HIV type 1 (HIV-1) and has been further studied by many researchers. We improved this antibody by developing in vitro evolution strategies that enhanced its neutralization activity. By coupling laboratory-evolved antibodies with potent toxins, we showed that immunotoxins can effectively kill infected cells. We are also developing genetic methods to halt HIV by gene therapy. We created unique human antibodies that can be expressed inside human cells to make the cells resistant to HIV infection. In the future, these antibodies might be delivered to the stem cells of patients infected with HIV-1, allowing the development of a disease-free immune system that would obviate the intense regimen of antiviral drugs now required to treat HIV disease. Using our increased understanding of antibody-antigen interactions, we extended our efforts in cancer therapy and developed rapid methods for creating human antibodies from antibodies derived from other species. We produced human antibodies that should enable us to selectively starve a variety of cancers by inhibiting angiogenesis and antibodies that will be used to deliver radionuclides to colon cancers to destroy the tumors. We hope that these antibodies will be used in clinical trials done by our collaborators at the Sloan-Kettering Cancer Center in New York City. On the basis of our studies on HIV-1, we used intracellular expression of antibodies directed against angio- 210 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE genic receptors to create a new gene-based approach to cancer. Our studies indicate that this type of gene therapy can be successfully applied to the treatment of cancer. our approach can become a key tool in selective chemotherapeutic strategies. To see a movie illustrating this approach, visit http://www.scripps.edu/mb/barbas/ antibody/antibody.mov. T H E R A P E U T I C A P P L I C AT I O N S O F C ATA LY T I C ADAPTOR IMMUNOTHERAPY: THE ADVENT OF ANTIBODIES CHEMOBODIES The development of highly efficient catalytic antibodies opens the door to many practical applications. One of the most fascinating is the use of such antibodies in human therapy. We think that use of this strategy can improve chemotherapeutic approaches to diseases such as cancer and AIDS. Chemotherapeutic regimens are typically limited by nonspecific toxic effects. To address this problem, we developed a novel and broadly applicable drug-masking chemistry that operates in conjunction with our unique broad-scope catalytic antibodies. This masking chemistry is applicable to a wide range of drugs because it is compatible with virtually any heteroatom. We showed that generic drug-masking groups can be selectively removed by sequential retroaldol–retro-Michael reactions catalyzed by antibody 38C2 (Fig. 3). This reaction cascade is not catalyzed by any known natural enzyme. We think that combining the chemical diversity of small synthetic molecules with the immunologic characteristics of antibody molecules will lead to therapeutic agents with superior properties. Therefore, we developed a conceptually new device that equips small synthetic molecules with both the immunologic effector functions and the long serum half-life of a generic antibody molecule. For a prototype, we developed a targeting device based on the formation of a covalent bond of defined stoichiometry between (1) a 1,3-diketone derivative of an arginine–glycine–aspartic acid peptidomimetic that targets the integrins αvβ3 and αvβ5 and (2) the reactive lysine of aldolase antibody 38C2 (Fig. 4). The resulting F i g . 3 . Targeting cancer and HIV with prodrugs activated by catalytic antibodies. A bifunctional antibody is shown targeting a cancer cell for destruction. A nontoxic analog of doxorubicin, prodoxorubicin, is being activated by an aldolase antibody to the toxic form of the drug. Application of this masking chemistry to the anticancer drugs doxorubicin, camptothecin, and etoposide produced prodrugs with substantially reduced toxicity. These prodrugs are selectively unmasked by the catalytic antibody when the antibody is applied at therapeutically relevant concentrations. The efficacy of this approach has been shown in in vivo models of cancer. Currently, we are developing more potent drugs and novel antibodies that will allow us to target breast, colon, and prostate cancer as well as cells infected with HIV-1. On the basis of our preliminary findings, we think that F i g . 4 . Designed small-molecule targeting agents (SCS-873 as shown) program the specificity of the antibody 38C2 (A). The resulting chemobodies (cp38C2, B) have characteristics that are often superior to either those of either the small molecule or the antibody alone. complex spontaneously assembled in vitro and in vivo, selectively retargeted antibody 38C2 to the surface of cells expressing integrins αvβ3 and αvβ5, dramatically increased the circulatory half-life of the peptidomimetic, and effectively reduced tumor growth in animal models of human Kaposi sarcoma, colon cancer, and melanoma. ZINC FINGER GENE SWITCHES The solution to many diseases might be simply turning genes on or off in a selective way. In order to pro- MOLECULAR BIOLOGY 2006 duce switches that can turn genes on or off, we are studying molecular recognition of DNA by zinc finger proteins and methods of creating novel zinc finger DNAbinding proteins (Fig. 5). Because of their modularity F i g . 5 . A designed polydactyl zinc finger binds 18 bp of DNA. A single zinc finger domain is highlighted. With this approach, we can now construct more than a billion gene switches and use them to specifically turn genes on or off in multiple organisms. Further elaboration of the approach allows every gene in the genome to be either turned on or upregulated or downregulated, providing a new approach to probe gene function across the genome. and well-defined structural features, zinc finger proteins are particularly well suited for use as DNA-binding proteins. Each finger forms an independently folded domain that typically recognizes 3 nucleotides of DNA. We showed that proteins can be selected or designed that contain zinc fingers that recognize novel DNA sequences. These studies are aiding the elucidation of rules for sequence-specific recognition within this family of proteins. We selected and designed specific zinc finger domains that will constitute an alphabet of 64 domains that will allow any DNA sequence to be bound selectively. The prospects for this “second genetic code” are fascinating and promise a major impact on basic and applied biology. We showed the potential of this approach in multiple mammalian and plant cell lines and in whole organisms. With the use of characterized modular zinc finger domains, polydactyl proteins capable of recognizing an 18-nucleotide site can be rapidly constructed. Our results suggest that zinc finger proteins might be useful as genetic regulators for a variety of human aliments and provide the basis for a new strategy of gene therapy. Our goal is to develop this class of therapeutic proteins to inhibit or enhance the synthesis of proteins, providing a direct strategy for fighting diseases of either somatic or viral origin. We are also developing proteins that will inhibit the growth of tumors and others that will inhibit the THE SCRIPPS RESEARCH INSTITUTE 211 expression of a protein known as CCR5, which is a key to infection of human cells by HIV-1. We developed an HIV-1–targeting transcription factor that strongly suppresses HIV-1 replication. Genetic diseases such as sickle cell anemia are also being targeted with this approach. Using a library of transcription factors, we developed a strategy that effectively allows us to turn on and off every gene in the genome. With this powerful new strategy, we can quickly regulate a target gene or discover other genes that have a key role in disease. In the future, we hope to use novel DNA-modifying enzymes directed by zinc fingers to manipulate chromosomes themselves. PUBLICATIONS Alwin, S., Gere, M.B., Guhl, E., Effertz, K., Barbas, C.F. III, Segal, D.J., Weitzman, M.D., Cathomen, T. Custom zinc-finger nucleases for use in human cells. Mol. Ther. 12:610, 2005. Blancafort, P., Chen, E.I., Gonzalez, B., Bergquist, S., Zijlstra, A., Guthy, D., Brachat, A., Brakenhoff, R.H., Quigley, J.P., Erdmann, D., Barbas, C.F. III. Genetic reprogramming of tumor cells by zinc finger transcription factors. Proc. Natl. Acad. Sci. U. S. A. 102:11716, 2005. Blau, C.A., Barbas, C.F. III, Bomhoff, A.L., Neades, R., Yan, J., Navas, P.A., Peterson, K.R. γ-Globin gene expression in chemical inducer of dimerization (CID)dependent multipotential cells established from human β-globin locus yeast artificial chromosome (β-YAC) transgenic mice. J. Biol. Chem. 280:36642, 2005. Cheong, P.H.-Y., Zhang, H., Thayumanavan, R., Tanaka, F., Houk, K.N., Barbas, C.F. III. Pipecolic acid-catalyzed direct asymmetric Mannich reactions. Org. Lett. 8:811, 2006. Corte-Real, S., Collins, C., Aires da Silva, F., Simas, P., Barbas, C.F. III, Chang, Y., Moore, P., Goncalves, J. Intrabodies targeting the Kaposi sarcoma-associated herpesvirus latency antigen inhibit viral persistence in lymphoma cells. Blood 106:3797, 2005. Dreier, B., Fuller, R.P., Segal, D.J., Lund, C.V., Blancafort, P., Huber, A., Koksch, B., Barbas, C.F. III. Development of zinc finger domains for recognition of the 5′-CNN3′ family DNA sequences and their use in the construction of artificial transcription factors. J. Biol. Chem. 280:35588, 2005. Eberhardy, S.R., Goncalves, J., Coelho, S., Segal, D.J., Berkhout, B., Barbas, C.F. III. Inhibition of human immunodeficiency virus type 1 replication with artificial transcription factors targeting the highly conserved primer-binding site. J. Virol. 80:2873, 2006. Lund, C.V., Popkov, M., Magnenat, L., Barbas, C.F. III. Zinc finger transcription factors designed for bispecific coregulation of ErbB2 and ErbB3 receptors: insights into ErbB receptor biology. Mol. Cell. Biol. 25:9082, 2005. Mandell, J., Barbas, C.F. III. Zinc Finger Tools: custom DNA-binding domains for transcription factors and nucleases. Nucleic Acids Res. 34(Web server issue):W516, 2006. Mase, N., Nakai, Y., Ohara, H., Yoda, H., Takabe, K., Tanaka, F., Barbas, C.F. III. Organocatalytic direct asymmetric aldol reactions in water. J. Am. Chem. Soc. 128:734, 2006. Mitsumori, S., Zhang, H., Cheong, P.H.-Y., Houk, K.N., Tanaka, F., Barbas, C.F. III. Direct asymmetric anti-Mannich-type reactions catalyzed by a designed amino acid. J. Am. Chem. Soc. 128:1040, 2006. Nathan, S., Rader, C., Barbas, C.F. III. Neutralization of Burkholderia pseudomallei protease by Fabs generated through phage display. Biosci. Biotechnol. Biochem. 69:2302, 2005. Popkov, M., Rader, C., Gonzelez, B., Sinha, S.C., Barbas, C.F. III. Small molecule drug activity in melanoma models may be dramatically enhanced with an antibody effector. Int. J. Cancer 119:1194, 2006. 212 MOLECULAR BIOLOGY 2006 Suri, J.T., Mitsumori, S., Albertshofer, K., Tanaka, F., Barbas, C.F. III. Dihydroxyacetone variants in the organocatalytic construction of carbohydrates: mimicking tagatose and fuculose aldolases. J. Org. Chem. 71:3822, 2006. Suri, J.T., Steiner, D.D., Barbas, C.F. III. Organocatalytic enantioselective synthesis of metabotropic glutamate receptor ligands. Org. Lett. 7:3885, 2005. Swan, C.H., Buhler, B., Tschan, M.P., Barbas, C.F. III, Torbett, B.E. T-cell protection and enrichment through lentiviral CCR5 intrabody gene delivery. Gene Ther. 13:1408, 2006. Tan, W., Dong, Z., Wilkinson, T.A., Barbas, C.F. III, Chow, S.A. Human immunodeficiency virus type 1 incorporated with fusion proteins consisting of integrase and the designed polydactyl zinc finger protein E2C can bias integration of viral DNA into a predetermined region in human cells. J. Virol. 80:1939, 2006. Tanaka, F., Barbas, C.F. III. Enamine-based reactions using organocatalysts: from aldolase antibodies to small amino acid and amine catalysts. J. Synth. Org. Chem. Jpn. 63:27, 2005. Tanaka, F., Fuller, R., Barbas, C.F. III. Development of small designer aldolase enzymes: catalytic activity, folding, and substrate specificity. Biochemistry 44:7583, 2005. Weinstain, R., Lerner, R.A., Barbas, C.F. III, Shabat, D. Antibody-catalyzed asymmetric intramolecular Michael addition of aldehydes and ketones to yield the disfavored cis-product. J. Am. Chem. Soc. 127:13104, 2005. Synthetic Enzymes, Catalytic Antibodies, Ozone Scavengers in Asthma, Organometallic Chemistry, and Biomolecular Computing E. Keinan, O. Reany, C.H. Lo, S. Bauer, N. Metanis, E. Kossoy, M. Soreni, R. Piran, M. Sinha, I. Ben-Shir, T. Ratner, T. Shekhter, T. Mejuch, E. Solel e focus on synthetically modified enzymes, antibody-catalyzed reactions, anticancer and antiasthma agents, and biomolecular computation, as illustrated in the following examples. W SYNTHETIC ENZYMES Efforts to generate new enzymatic activities from existing protein scaffolds may not only provide biotechnologically useful catalysts but also lead to better understanding of the natural process of evolution. We profoundly changed the catalytic activity and mechanism of the enzyme 4-oxalocrotonate tautomerase by means of rationally designed synthetic mutations. For example, a single amino acid substitution that corresponds to a mutation in a single base pair led to a dramatic change in the catalytic activity. Although the wild-type enzyme catalyzes only the tautomerization of 4-oxalocrotonate, the mutant P1A catalyzes both the THE SCRIPPS RESEARCH INSTITUTE original tautomerization reaction via a general acid-base mechanism and the decarboxylation of oxaloacetate via a nucleophilic mechanism. We also showed that the electrostatic manipulation of an enzyme’s active site can alter the substrate specificity of the enzyme in a predictable way. We replaced 1, 2, or all 3 active-site arginine residues with citrulline analogs to maintain the steric features of the active site of 4-oxalocrotonate tautomerase while changing its electronic properties. These synthetic changes revealed that the wild-type enzyme binds the natural substrate predominantly through electrostatic interactions. This and other mechanistic insights led to the design of a modified enzyme that was specific for a new substrate that had different electrostatic properties and that bound the enzyme via hydrogen-bonding complementarity rather than electrostatic interactions. This research on synthetic enzymes is being done in collaboration with P.E. Dawson, Department of Cell Biology. C ATA LY T I C A N T I B O D I E S Engineering herbicide resistance in crops facilitates control of weed species, particularly weeds that are genetically related to the crop, and may be useful in selecting lines that have undergone multiple transformation events. We showed that herbicide-resistant plants can be engineered by designing both a herbicide and a catalytic antibody that destroys the herbicide within the plants. First, we developed a carbamate herbicide that can be catalytically destroyed by the aldolase antibody 38C2. Then we targeted the light chain and half of the heavy chain (Fab) of the catalytic antibody to the endoplasmic reticulum in 2 lines of Arabidopsis thaliana transformants. Finally, we crossed the 2 transgenic plants to produce a herbicide-resistant F1 hybrid (Fig. 1). Our results suggest that in vivo expression of catalytic antibodies could become a general strategy to achieve phenotype modifications not only in plants but also in other organisms. O Z O N E S C AV E N G E R S A N D A N T I A S T H M A A C T I V I T Y A new hypothesis we proposed on the mechanism of asthmatic inflammation has led to an ozone-scavenging compound that prevents bronchial obstruction in rats with asthma. Previously, scientists at Scripps Research discovered that ozone can be generated not only via the antibody-mediated water oxidation pathway but also by antibody-coated activated white blood cells during inflammatory processes. This finding led us to speculate that the pulmonary inflammation in asthma might be caused by ozone production by white blood cells in MOLECULAR BIOLOGY 2006 F i g . 1 . Influence of herbicide (1) on the rooting and development of seedlings of F1 hybrids and control A thaliana plants. The control plants are shown in A and C; the hybrid plant lines (F1) expressing both light and heavy chains of the catalytic antibody 38C2, in B and D. Plantlets grown on medium without the herbicide are shown in A and B; those grown with the herbicide are shown in C and D. lungs and that inhalation of electron-rich olefins, which are known ozone scavengers, might have antiasthmatic effects. In experiments in rats, inhalation of such a compound, limonene, caused a significant improvement in signs of asthma. These results could have consequences in the management of asthma. O R G A N O M E TA L L I C C H E M I S T R Y Rhenium oxide, which is known primarily as a strong oxidant, is a highly selective Lewis acid catalyst that affects the heteroacylative dimerization of tetrahydrofuran at room temperature. This multicomponent reaction, which involves tetrahydrofuran, trifluoroacetic anhydride, and a carboxylic acid, produces a nonsymmetrical diester (compound 3 in Fig. 2) in high yields. The proposed catalytic cycle (Fig. 2) involves a multistep sequence of nucleophilic attacks, metal-oxygen bond metathesis, and electrophilic cleavage by trifluoroacetic anhydride. This synthetically useful reaction highlights the unique, frequently avoided Lewis acidity of transition-metal oxides. In study with the platinum complex TpPt(CO)CH3 (Tp = hydridotrispyrazolylborate), we found that the proton exchange between water and the methyl group involves the formation and deprotonation of a “sticky” σ-methane ligand. The efficiency of this nontrivial process is attributed to the spatial orientation of functional groups that operate in concert to achieve a multistep proton walk. The key role played by the free pyrazolyl nitrogen, acting as a proton carrier, is reminiscent of the dual functionality of the histidine in the catalytic triad of natural serine proteases. THE SCRIPPS RESEARCH INSTITUTE 213 F i g . 2 . The catalytic cycle of the rhenium oxide–catalyzed heteroa- cylative dimerization of tetrahydrofuran (THF), which is proposed on the basis of isotope-labeling experiments, starts with an attack of a rhenium oxo ligand on a coordinated tetrahydrofuran, then an attack of the resultant alkoxide ligand on a second coordinated tetrahydrofuran, nucleophilic addition of the resultant alkoxide ligand to the coordinated carboxylic acid, and finally, electrophilic cleavage of the other coordinated alkoxide by trifluoroacetic anhydride (TFAA). BIOMOLECULAR COMPUTING DEVICES Previously, we described a programmable finite automaton with 2 symbols and 2 states that computed autonomously. All of the components of the device, including hardware, software, input, and output, were biomolecules mixed together in solution. The hardware consisted of a restriction nuclease and a ligase; the software (transition rules) and the input were doublestranded DNA oligomers. Computation was carried out by processing the input molecule via repetitive cycles of restriction, hybridization, and ligation reactions to produce a final-state output in the form of a doublestranded DNA molecule. More recently, we markedly increased the levels of complexity and mathematical power of these automata by the design of a 3-state–3-symbol automaton, thus increasing the number of syntactically distinct programs from 765 to 1 billion. We have further amplified the applicability of this design by using surface-anchored input molecules and surface plasmon resonance technology to monitor the computation steps in real time. This technology allowed parallel computation with DNA chips that carry multiple input molecules and can be used as pixel arrays for image encryption. PUBLICATIONS Lo, H.C., Han, H., D‚Souza, L.J., Sinha, S.C., Keinan, E. Rhenium(VII) oxide-catalyzed heteroacylative ring-opening dimerization of tetrahydrofuran. J. Am. Chem. Soc., in press. 214 MOLECULAR BIOLOGY 2006 Lo, H.C., Iron, M.A. Martin, J.M.L., Keinan, E. Proton walk in the aqueous platinum complex TpPtMeCO via a sticky σ-methane ligand. Chem. Eur. J., in press. Metanis, N., Keinan, E., Dawson, P.E. Synthetic seleno-glutaredoxin 3: highly reducing oxidoreductases with enhanced catalytic efficiency. J. Am. Chem. Soc., in press. Tuttle, T., Keinan, E., Thiel, W. Understanding the enzymatic activity of 4-oxalocrotonate tautomerase and its mutant analogues: a computational study. J. Phys. Chem. B Condens. Matter Mater. Surf. Interfaces Biophys. 110:19685, 2006. Weiss, Y., Rubin, B., Shulman, A., Ben Shir, I., Keinan, E., Wolf, S. Determination of plant resistance to carbamate herbicidal compounds inhibiting cell division and early growth by seed and plantlets bioassays. Nat. Protoc., in press. Weiss, Y., Shulman, A., Ben Shir, I., Keinan, E., Wolf, S. Herbicide-resistance conferred by expression of a catalytic antibody in Arabidopsis thaliana. Nat. Biotechnol. 24:713, 2006. Functional Characterization of Proteases via Combinatorial Libraries J.L. Harris, J. Alves* ith the complete sequencing of genomes from multiple organisms, information on the repertoire of genes can be readily established. However, large gaps still remain in our knowledge of the biological role of most genes. These gaps are mainly due to the fact that most biological functions are regulated not at the gene or transcript level, but at the posttranslational level. In contrast to the situation in genomics, in which the changes in the content or amount of cellular DNA or RNA can be readily examined, monitoring translational and posttranslational dynamics of functional proteins on a genome-wide level is more difficult. Progress in our current understanding of biological processes is limited by the available tools that can be used to probe function at the posttranslational level. We are developing and applying technologies based on small-molecule protein modifiers to profile the active state of enzymes. In collaboration with N. Winssinger, Université Louis Pasteur, Strasbourg, Germany, we have developed an encoding strategy that uses peptide nucleic acid (PNA) sequences. Encoding combinatorial libraries with PNA tags allows not only for the synthetic history of the library to be captured in the resulting molecule but also for spatial deconvolution of the molecules on DNA microarrays. Using this technology, we have created encoded protease inhibitor and substrate libraries of thousands of molecules. These libraries have been applied to var- W THE SCRIPPS RESEARCH INSTITUTE ious biological systems and have resulted in the identification and characterization of proteases within those systems. For example, a cysteine protease from the house dust mite Dermatophagoides pteronyssinus was identified by using a 4000-member PNA-encoded inhibitor library. The identified protease plays a key role in allergic hypersensitivity through the selective degradation of CD25 from T cells. Another example of functional characterization of protein activity is the profiling of the substrate specificity of proteases from Dengue virus, the etiologic agent of dengue fever, dengue hemorrhagic fever, and dengue shock syndrome. Using 2 substrate libraries of approximately 160,000 members, we characterized the structure-activity relationship of the NS3 protease from the 4 Dengue virus serotypes and facilitated the development of inhibitors of the virus. PUBLICATIONS Harris, J.L. Protease substrate profiling. In: Enzyme assays: high-throughput screening, genetic selection and fingerprinting. Reymond, J.-L. (Ed.). Wiley-VCH, New York, 2006, p. 303. Harris, J.L., Winssinger, N. PNA encoding (PNA = peptide nucleic acid): from solution-based libraries to organized microarrays. Chem. Eur. J. 11:6792, 2005. Li, J., Lim, S.P., Beer, D., Patel, V., Wen, D., Tumanut, C., Tully, D.C., Williams, J.A., Jiricek, J., Priestle, J.P., Harris, J.L., Vasudevan, S.G. Functional profiling of recombinant NS3 proteases from all four serotypes of Dengue virus using tetrapeptide and octapeptide substrate libraries. J. Biol. Chem. 280:28766, 2005. Petrassi, H.M., Williams, J.A., Li, J., Tumanut, C., Ek, J., Nakai, T., Masick, B., Backes, B.J., Harris, J.L. A strategy to profile prime and non-prime proteolytic substrate specificity. Bioorg. Med. Chem. Lett. 15:3162, 2005. Winssinger, N., Harris, J.L. Microarray-based functional protein profiling using peptide nucleic acid-encoded libraries. Expert Rev. Proteomics 2:937, 2005. Organic Synthesis and Selective Drug Delivery S.C. Sinha, R.A. Lerner, Z. Chen, S. De, S. Das, S. Abraham, F. Guo ur main research interests are synthesis of biologically important natural and nonnatural molecules, synthetic methods, and antibody catalysis in organic synthesis and selective drug delivery. During the past year, we focused on 3 different classes of compounds: the anticancer adjacent bis-tetrahydrofuran annonaceous acetogenins, the antibacterial macrocyclic lactones sorangiolides, and nonnatural small-molecule drugs that target G protein–coupled receptors. In our work on antibody catalysis, we developed a proadapter approach for production of the chemically programmed O MOLECULAR BIOLOGY 2006 aldolase antibody 38C2 and new doxorubicin prodrugs that are catalyzed by antibody 38C2 faster than the previously reported drugs are. S E L E C T I V E C H E M O T H E R A P Y W I T H C ATA LY T I C ALDOLASE ANTIBODIES For selective chemotherapy, we intend to develop drug conjugates and prodrugs that will target cell-surface receptors, such as the glycoprotein integrins αvβ3 and α v β 5 . These integrins are directly implicated in tumor angiogenesis; they are overexpressed in the vasculature of angiogenic tumors and in numerous cancer cells but are less expressed on quiescent blood vessels. Using antibody 38C2 and small-molecule antagonists of αvβ3 and αvβ5 (or a targeting agent), we developed antagonist-38C2 conjugates, also known as chemically programmed 38C2 (Fig. 1). F i g . 1 . Schematic drawings of the diketone and the proadapter strategies used to produce chemically programmed antibody constructs that target cells expressing the integrins αvβ3 and αvβ5. Abbreviations: Ab, antibody; TA, targeting agent. The conjugation between the targeting agents and the antibody takes place in the binding sites though the diketone or vinyl ketone linkers. Because the vinyl ketones are highly reactive, we used the corresponding acetone adduct as the prolinker, which undergoes 38C2catalyzed reaction to produce the active linker before the active linker reacts. This strategy has been termed the proadapter approach. The conjugates prepared by both approaches bound efficiently to cells expressing αvβ3 and αvβ5, including human breast cancer cell lines MDA-MB-435 and MDA-MB-231, and inhibited the growth of both the primary tumors and secondary metastasis in distant organs. Development of these strategies for the formation of antibody constructs can have a large effect on the treatment of various diseases, including cancer. In the alternative approach, we are developing prodrugs that can be efficiently activated by the aldolase antibodies 38C2 and 93F3. These antibodies will be targeted to tumor cells or the tumor vasculature by using antagonists of αvβ3 and αvβ5. In the past year, we devel- THE SCRIPPS RESEARCH INSTITUTE 215 oped new doxorubicin prodrugs that are not only more stable than the previously reported analogous prodrugs but also activated faster by using antibody 38C2. We also produced 38C2-antagonist conjugates. The conjugates bound efficiently to MDA-MB-231 cells expressing αvβ3 and αvβ5. We also found that the modified antibody can activate new doxorubicin prodrugs. Therefore, we have all the tools to investigate prodrug therapy in animal models. The selective chemotherapy studies are carried out in collaboration with C.F. Barbas, Department of Molecular Biology, and B. Mueller, La Jolla Institute for Molecular Medicine, San Diego, California. S Y N T H E S I S O F N AT U R A L A N D N O N N AT U R A L S M A L L MOLECULES Total synthesis of naturally occurring and biologically important compounds is important not only for confirming their structures but also for producing the compounds and their analogs for comprehensive biological evaluations. To synthesize these compounds, we are developing methods that involve both antibody catalysis and common synthetic routes. In the past year, in addition to synthesizing sorangiolides, which are naturally occurring macrocyclic lactones, and the library of bis-tetrahydrofuran annonaceous acetogenins, we focused on small-molecule nonnatural ligands of G protein–coupled receptors. The studies on the synthesis of these ligands are carried out in collaboration with E. Roberts, Department of Chemistry. Sorangiolides (Fig. 2) are weakly active antibacterial compounds. Our goal is to synthesize the highly F i g . 2 . Structure of sorangiolides A and B (top) and a general structure of bis-tetrahydrofuran annonaceous acetogenins (bottom). active sorangiolide analogs. Thus, we have developed synthetic routes that can provide the macrocyclic structure of sorangiolides. Using an intermediate, we will synthesize both the natural and nonnatural molecules. For other bis-tetrahydrofuran acetogenins, which are among the most active cancer agents and are toxic to several human cancer cell lines at much lower concen- 216 MOLECULAR BIOLOGY 2006 trations than doxorubicin is, we developed methods that can provide all the stereoisomers of asimicin and bullatacin. The new methods involve a bidirectional approach. Now, we are pursuing synthesis of the 64 stereoisomers of asimicin and bullatacin. PUBLICATIONS Das, S., Li, L.-S., Abraham, S., Chen, Z., Sinha, S.C. A bidirectional approach to the synthesis of a complete library of adjacent-bis-THF annonaceous acetogenins. J. Org. Chem. 70:5922, 2005. Li, L.-S., Babendure, J.L., Sinha, S.C., Olefsky, J.M., Lerner, R.A. Synthesis and evaluation of photolabile insulin prodrugs. Bioorg. Med. Chem. Lett. 15:3917, 2005. Popkov, M., Rader, C., Gonzalez, B., Sinha, S.C., Barbas, C.F. III. Small molecule drug activity in melanoma models may be dramatically enhanced with an antibody effector. Int. J. Cancer 119:1194, 2006. Structure, Function, and Applications of Virus Particles J.E. Johnson, M. Banerjee, A. Chatterji, Z. Chen, I. Gertsman, R. Huang, R. Khayat, G. Lander, J. Lanman, K.K. Lee, T. Matsui, P. Natarajan, A. Odegard, J. Speir e investigate model virus systems that provide insights for understanding assembly, maturation, entry, localization, and replication. We have also developed viruses as reagents for applications in nanomedicine, chemistry, and biology. We investigate viruses that infect bacteria, insects, plants, and the extreme thermophile Sulfolobus. These viruses have genomes of single-stranded RNA and doublestranded DNA. We use a variety of physical methods to investigate structure-function relationships, including single-crystal and static and time-resolved solution x-ray diffraction, electron cryomicroscopy and image reconstruction, mass spectrometry, structure-based computational analyses, and methods associated with thermodynamic characterization of virus particles and their transitions. Biological methods we use include genetic engineering of viral genes and their expression in Escherichia coli, mammalian cells, insect cells, and yeast and the characterization of these gene products by physical methods. For cytologic studies of viral entry and infection, we use fluorescence and electron microscopy and particles assembled in heterologous expression systems. Our studies depend on extensive consultations and collaborations with others at Scripps Research, including groups led by C.L. Brooks, D.A. Case, B. Carragher, M.G. Finn, M. Manchester, D.R. Millar, R.A. Milligan, W THE SCRIPPS RESEARCH INSTITUTE C. Potter, V. Reddy, A. Schneemann, G. Siuzdak, J.R. Williamson, and M.J. Yeager, and a variety of groups outside of Scripps. DOUBLE-STRANDED DNA VIRUSES HK97 is a double-stranded DNA virus similar to phage λ. It undergoes a remarkable morphogenesis in its assembly and maturation, and this process can be recapitulated in vitro. We determined the atomic resolution structure of the 650-Å mature, head II particle and discovered the mechanism used to concatenate the subunits of the particle into a chain-mail fabric similar to that seen in armor of medieval knights. In the past year, we focused on the dynamics of maturation. Prohead II is a 500-Å metastable intermediate at pH 7 that can be induced to begin maturation by lowering the pH to 4. Solution x-ray scattering and single-molecule fluorescence showed that the initial transition to a particle of about 560 Å occurs as a highly cooperative, stochastic event with no detectable intermediates that takes place in less than 1 second for an individual particle. A quorum of cross-links must form in this particle to generate the second expansion intermediate (about 650 Å), which also forms cooperatively with no detectable intermediates. At pH 4, formation of crosslinks continues, with 360 formed per particle. Limited pentamer dynamics (established from crystallography and electron cryomicroscopy) prevents the last 60 crosslinks from forming, but pentamer trajectories extend at pH 7, allowing these cross-links to form, completing maturation. Bacteriophage P22 is the prototype of the Podoviridae, which are characterized by a T = 7 capsid with a short tail structure incorporated into a unique 5-fold vertex. We determined an asymmetric reconstruction of this particle that revealed spooled DNA, the dodecameric portal, and the location of the 9 gene products known to be in the particle. Sulfolobus turreted icosahedral virus is an archaeal virus isolated from Sulfolobus, which grows in the acidic hot sulfur springs (pH 2–4, 72°C–92°C) in Yellowstone National Park. An electron cryomicroscopy reconstruction of the virus showed that the capsid has pseudo T = 31 quasi symmetry and is 1000 Å in diameter, including the pentons. We solved the x-ray structure of the major capsid protein of the virus, and it revealed a fold nearly identical to the major capsid proteins of the eukaryotic adenoviruses and PRD-1, a virus that infects bacteria. These findings indicate a virus phylogeny that spans the 3 domains of life. Difference electron density maps in which the x-ray model is subtracted from the elec- MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 217 tron cryomicroscopy density clearly shows an internal membrane in which the capsid proteins are anchored. Ochoa, W., Chatterji, A., Lin, T., Johnson, J.E. Generation and structural analysis of reactive empty particles derived from an icosahedral virus. Chem. Biol. 13:771, 2006. SINGLE-STRANDED RNA VIRUSES Prasad, T., Turner, M., Falkner, J., Mittleman, D., Johnson, J.E., Lin, T., Colvin, V. Nanostructured virus crystals for x-ray optics. IEEE Trans. Nanotechnol. 5:93, 2006. Flock House virus is a T = 3, single-stranded RNA virus that infects Drosophila. We are studying viral entry and early expression and assembly of the capsid protein. Recently, studies on viral entry indicated the presence of an “eluted” particle early in infection that has initiated its disassembly program but is then eluted back into the medium. We did a phenotypic characterization of the particles, and we are using electron cryomicroscopy to study them. For studies on the expression and assembly of the capsid protein, we are using tags inserted genetically in the capsid protein that allow the freshly made proteins to be optically visualized with a fluorophore and in the electron microscope with photoconversion of the fluorophore. Recently, high-pressure freezing of infected cells revealed exceptionally detailed features of viral entry and regions of replication within the cell. Refined atomic models of tetravirus structures and structure-based mutagenesis combined with highly sensitive assays for defining phenotypes have revealed the electrostatic principals of maturation for the T = 4 tetraviruses. Cowpea mosaic virus is a 30-nM reagent that we use for chemistry and nanomedicine. We found that particles of the virus with doxorubicin bound internally can be specifically targeted to tumor cells via peptides on the viral surface that recognize receptors for vascularization signals that are highly expressed on tumor cells. PUBLICATIONS du Plessis, L., Hendry, D.A., Dorrington, R.A., Hanzlik, T.N., Johnson, J.E., Appel, M. Revised RNA2 sequence of the tetravirus nudaurelia capensis ω virus (NωV): annotated sequence record. Arch. Virol. 150:2397, 2005. Khayat, R., Tang, L., Larson, E.T., Lawrence, C.M., Young., M., Johnson, J.E. Structure of an archaeal virus capsid protein reveals a common ancestry to eukaryotic and bacterial viruses. Proc. Natl. Acad. Sci. U. S. A. 102:18944, 2005. Lander, G.C., Tang, L., Casjens, S.R., Gilcrease, E.B., Prevelige, P., Poliakov, A., Potter, C.S., Carragher, B., Johnson, J.E. The structure of an infectious P22 virion shows the signal for headful DNA packaging. Science 312:1791, 2006. Lee, K.K., Tsuruta, H., Hendrix, R.W., Duda, R.L., Johnson, J.E. Cooperative reorganization of a 420 subunit virus capsid. J. Mol. Biol. 352:723, 2005. Reddy, V.S., Johnson, J.E. Structure-derived insights into virus assembly. Adv. Virus Res. 64:45, 2005. Sapsford, K.E., Soto, C.M., Blum, A.S., Chatterji, A., Lin, T., Johnson, J.E., Ligler, F.S., Ratna, B.R. A cowpea mosaic virus nanoscaffold for multiplexed antibody conjugation: application as an immunoassay tracer. Biosens. Bioelectron. 21:1668, 2006. Shepherd, C.M., Borelli, I.A., Lander, G., Natarajan, P., Siddavanahalli, V., Bajaj, C., Johnson, J.E., Brooks, C.L. III, Reddy, V.S. VIPERdb: a relational database for structural virology. Nucleic Acids Res. 34:386, 2006. Soto, C.M., Blum, A.S., Vora, G.J., Lebedev, N., Meador, C.E., Won, A.P., Chatterji, A., Johnson, J.E., Ratna, B.R. Fluorescent signal amplification of carbocyanine dyes using engineered viral nanoparticles. J. Am. Chem. Soc. 128:5184, 2006. Speir, J.A., Bothner, B., Qu, C., Willits, D.A., Young, M.J., Johnson, J.E. Enhanced local symmetry interactions globally stabilize a mutant virus capsid that maintains infectivity and capsid dynamics J. Virol. 80:3582, 2006. Tang, J., Johnson, J.M., Dryden, K.A., Young, M.J., Zlotnick, A., Johnson, J.E. The role of subunit hinges and molecular “switches” in the control of viral capsid polymorphism. J. Struct. Biol. 154:59, 2006. Tang, L., Gilcrease, E.B., Casjens, S.R., Johnson, J.E. Highly discriminatory binding of capsid-cementing proteins in bacteriophage L. Structure 14:837, 2006. Taylor, D.J., Speir, J.A., Reddy, V., Cingolani, G., Pringle, F.M., Ball, L.A., Johnson, J.E. Preliminary x-ray characterization of authentic providence virus and attempts to express its coat protein gene in recombinant baculovirus. Arch. Virol. 151:155, 2006. Walukiewicz, H.E., Johnson, J.E., Schneemann, A. Morphological changes in the T = 3 capsid of Flock House virus during cell entry. J. Virol. 80:615, 2006. Wikoff, W.R., Conway, J.F., Tang, J., Lee, K.K., Gan, L., Cheng, N., Duda, R.L., Hendrix, R.W., Steven, A.C., Johnson, J.E. Time-resolved molecular dynamics of HK97 capsid maturation interpreted by electron cryo-microscopy and x-ray crystallography. J. Struct. Biol. 153:300, 2006. Nanomanufacturing on an Icosahedral Scaffold and Neutralization of Avian H5N1 Influenza Viruses T. Lin, J.E. Johnson, A. Censullo, A. Chatterji MOLECULAR ELECTRONICS ON AN ICOSAHEDRAL SCAFFOLD Lin, T., Lomonossoff, G.P., Johnson, J.E. Structure-based engineering of an icosahedral virus for nanomedicine and nanotechnology. In: Nanotechnology in Biology and Medicine: Methods, Devices, and Applications. Vo-Dinh. T. (Ed.). CRC Press, Boca Raton, FL, in press. Medintz, I.L., Sapsford, K.E., Konnert, J.H., Chatterji, A., Lin, T., Johnson, J.E., Mattoussi, H. Decoration of discretely immobilized cowpea mosaic virus with luminescent quantum dots. Langmuir 21:5501, 2005. Natarajan, P., Lander, G.C., Shepherd, C.M., Reddy, V.S., Brooks, C.L. III, Johnson, J.E. Exploring icosahedral virus structures with VIPER. Nat. Rev. Microbiol. 3:809, 2005. Molecular manufacturing, the essence of nanotechnology, involves the manipulation of molecules as the self-assembling components at the nanometer scale to build devices in mesoscale. Although small molecules with novel electronic properties can be synthesized, making functional connectivity among the different components in designed patterns is generally difficult. In contrast, biological macromolecules are more amenable 218 MOLECULAR BIOLOGY 2006 for self-assembly because of their versatility, programmability through genetic engineering, and propensity to form arrays and can be used either directly as devices or as scaffolds for patterning small molecules. We have shown that cowpea mosaic virus (CPMV), an icosahedral plant virus, can be used as the template for nanochemistry by introducing unique cysteine residues and exploiting the native lysine residues. In collaborative studies with B.R. Ratna, Naval Research Laboratory, Washington, D.C., the virus capsid was exploited as a nano circuit board, and the reactive groups were used as anchoring points for the assembly of the electronic molecules oligophenylene-vinylene and 1,4-C6H 4[trans-(4AcSC6H 4≡CPt(Pbu3)2≡C] 2. The establishment of the molecular network was shown by measuring electronic conductance with scanning tunnel microscopy. THE SCRIPPS RESEARCH INSTITUTE analysis of escape mutants in conjunction with the vaccine development. Structural studies of antibody interactions with the H5N1 viruses are also being carried out to shed light on the mechanism of neutralization of the viruses. PUBLICATIONS Medintz, I.L., Sapsford, K.E., Konnert, J.H., Chatterji, A., Lin, T., Johnson, J.E., Mattoussi, H. Decoration of discretely immobilized cowpea mosaic virus with luminescent quantum dots. Langmuir 21:5501, 2005. Prasad, T., Turner, M., Falkner, J., Mittleman, D., Johnson, J.E., Lin, T., Colvin, V. Nanostructured virus crystals for x-ray optics. IEEE Trans. Nanotechnol., in press. Design and Informatics in Structural Virology N E U T R A L I Z AT I O N O F AV I A N H 5 N 1 I N F L U E N Z A V.S. Reddy, S. Kumar, M. Tripp, P. Singh, R. Mannige, VIRUSES I. Borelli, J. Loo, C.L. Brooks III, J.E. Johnson, Influenza is one of the most important viral diseases in humans. It has caused morbidity and mortality in millions of people in frequent epidemics and pandemics throughout the centuries. Human influenza virus is typically associated with 3 H subtypes: H1, H2, and H3. In recent years, an avian H5 (H5N1) influenza virus crossed the species barrier to infect humans with high virulence. To date, the avian virus has not been efficient in transmission from human to human, and the disease has not spread in the human population. However, the continuous circulation and spreading of H5N1 viruses in avian species across the globe leads to more human infections and increases the likelihood that the virus will acquire the necessary characteristics for efficient human-to-human transmission through genetic mutation or reassortment with a prevailing human influenza A virus. The possible emergence of an H5N1 virus highly contagious to humans is a serious pandemic threat. Therefore, producing effective vaccines to counter the threat posed by the H5N1 viruses is important. CPMV is an effective scaffold for the development of subunit vaccines. We are developing a novel combinatorial strategy in which the CPMV system is used to identify vaccine candidates. In another study in collaboration with scientists in Hong Kong and Southern China, the epicenter of the influenza outbreaks, we have produced more than 100 monoclonal antibodies against the avian influenza viruses and have shown that many of these antibodies are neutralizing. These neutralizing antibodies are used in the M. Manchester, G. Nemerow, A. Schneemann e are interested in understanding the structural underpinnings and requirements for formation and function of viral capsids and in designing novel protein shells that polyvalently display molecules of interest. To this end, we use structural, computational, informatics, and genetic methods. Viruses are highly evolved macromolecular machines that perform a variety of functions during their life cycle, including selective packaging of the genome, self-assembly into uniform capsids, binding to host cells, and delivery of the genome to the targeted cells. Simple viruses, such as nonenveloped viruses, form closed protein shells of uniform size and character by the self-association of structural and functional components: proteins and the nucleic acid genome. Hence, these viruses are useful for structural and functional analyses. In collaboration of with G.R. Nemerow, Department of Immunology, we are using x-ray crystallographic methods to determine the structure of the entire human adenovirus particle, currently at about 9-Å resolution. We are continuing to collect diffraction data at higher resolution. We continue to maintain and expand the virus structure database, namely VIPERdb (http://viperdb.scripps.edu), where the coordinates of the characterized spherical capsid structures are stored and organized in terms of viral taxonomy and capsid architecture. We are developing structural analysis tools to “mine” the capsid structures in terms of protein-protein interactions, contacting residue pairs, association ener- W MOLECULAR BIOLOGY 2006 gies, contributions of individual residues, and surface characteristics. VIPERdb is being developed as part of the Multiscale Modeling Tools for Structural Biology, a National Institutes of Health research resource headed by C.L. Brooks, Department of Molecular Biology. In addition, on the basis of the structural similarity that occurs within a virus family, we are building homology models for the uncharacterized members of virus families. These models will be useful for molecular virologists investigating structural and functional relationships in viruses. We are generating decoys of pathogenic molecules on the surfaces of viral capsids that can be used as vaccines against cytotoxins such as ricin. Currently, tomato bushy stunt virus–like capsids are our display platform of choice; the platform consists of multiple copies of a 2-domain capsid protein subunit with the C-terminal P-domain exposed on the surface. Such a unique subunit structure is useful for attaching peptides or proteins of interest at the end of the C terminus of the capsid protein or for replacing the external P-domain with the proteins of interest rather than inserting them in a loop. PUBLICATIONS Hsu, C., Singh, P., Ochoa, W., Manayani, D.J., Manchester, M., Schneemann, A., Reddy, V.S. Characterization of polymorphism displayed by the coat protein mutants of tomato bushy stunt virus. Virology 349:222, 2006. Natarajan, P., Lander, G.C., Shepherd, C.M., Reddy, V.S., Brooks, C.L. III, Johnson, J.E. Exploring icosahedral virus structures with VIPER. Nat. Rev. Microbiol. 3:809, 2005. Shepherd, C.M., Borelli, I.A., Lander, G., Natarajan, P., Siddavanahalli, V., Bajaj, C., Johnson, J.E., Brooks, C.L. III, Reddy, V.S. VIPERdb: a relational database for structural virology. Nucleic Acids Res. 34(Database Issue):D386, 2006. Taylor, D.J., Speir, J.A., Reddy, V., Cingolani, G., Pringle, F.M., Ball, L.A., Johnson, J.E. Preliminary x-ray characterization of authentic providence virus and attempts to express its coat protein gene in recombinant baculovirus. Arch. Virol. 151:155, 2006. Biology and Applications of Icosahedral Virus Capsids A. Schneemann, B. Groschel, C. Hsu, J. Lee, D.J. Manayani, D. Marshall, J.E. Petrillo, M.E. Siladi, P.A. Venter oat proteins of nonenveloped, icosahedral viruses perform multiple functions during the course of viral infection, including capsid assembly, specific encapsidation of the viral genome, binding to a cellular receptor, and uncoating. In some viruses, a single type of protein is sufficient to carry out these functions; C THE SCRIPPS RESEARCH INSTITUTE 219 we are interested in the determinants that endow a polypeptide chain with such versatility. We seek to harness this versatility for novel applications of viruses in biotechnology and nanotechnology. We focus on a structurally and genetically well-characterized virus family, the T = 3 nodaviruses. Nodaviruses are composed of 180 copies of a single coat protein and 2 strands of positive-sense RNA. Currently, we are elucidating the mechanism by which the 2 genomic RNAs are packaged into a single virion. Our long-term goal is to develop nodaviruses as RNA packaging and delivery vectors. Our data indicate that the 2 viral RNAs are recognized separately, but it is not yet known whether packaging occurs sequentially and whether one or more coat protein subunits are involved in this process. Interestingly, we found that RNA genome packaging is coupled to genome replication, suggesting potential approaches for packaging of foreign RNAs. In other studies, we are investigating the mechanism by which nodaviral protein B2 suppresses RNA silencing in infected cells. In collaboration with J.R. Williamson, Department of Molecular Biology, we showed that B2 binds to double-stranded RNA in a sequence-independent manner and that it interferes with cleavage of double-stranded RNA substrates by the cellular protein Dicer. Moreover, in collaboration with J.L. Imler, University of Strasbourg, Strasbourg, France, we showed that B2 is critical for nodaviral infection of Drosophila and that Dicer plays an essential role in host defense against nodaviruses in vivo. We are also collaborating with several investigators at Scripps Research, the Salk Institute, and Harvard University to develop nodaviruses as platforms for delivery of anthrax antitoxins. To this end, we are using particles to display the VWA domain of capillary morphogenesis protein 2, the cellular receptor for anthrax toxin, in a multivalent fashion on the surface of the virion. Two insertion sites yielding different patterns of 180 copies of the VWA domain were selected on the basis of computational modeling of the high-resolution crystal structure of the insect nodavirus Flock House virus. The resulting chimeric viruslike particles functioned as a potent anthrax antitoxin in cell culture and protected rats from challenge with lethal toxin. This research is important because it shows that protein domains containing more than 150 amino acids can be displayed on Flock House virus in a biologically functional form, suggesting numerous additional applications. 220 MOLECULAR BIOLOGY 2006 Flock House virus particles are also good candidates for novel materials in nanotechnology applications. The particles are stable, easily manipulated, biocompatible, and nontoxic in vivo and can be produced easily and in high quantities. The high-resolution x-ray structure of the virus revealed the potential for using chemical approaches to attach ligands to the surface of the virus and for using genetic strategies to modify the capsid. In collaboration with M. Manchester, Department of Cell Biology, and M. Ozkan, University of California, Riverside, we used conjugation chemistry to couple inorganic nanotubes and quantum dots to Flock House virus particles to produce an array of novel hybrid structures. This approach may one day be used to fabricate unique materials for a variety of applications, including biofilms with tunable pore size, 3-dimensional scaffolds for production of nanoelectronic devices, and drug delivery. PUBLICATIONS Chao, J.A., Lee, J.H., Chapados, B.R., Debler, E.W., Schneemann, A., Williamson, J.R. Dual modes of RNA-silencing suppression by Flock House virus protein B2. Nat. Struct. Mol. Biol. 12:952, 2005. Destito, G., Schneemann, A., Manchester, M. Biomedical nanotechnology using virus-based nanoparticles. Curr. Top. Microbiol. Immunol., in press. Galiana-Arnoux, D., Dostert, C., Schneemann, A., Hoffmann, J.A., Imler, J.L. Essential function in vivo for Dicer-2 in host defense against RNA viruses in Drosophila. Nat. Immunol. 7:590, 2006. Hsu, C., Singh, P., Ochoa, W., Manayani, D.J., Manchester, M., Schneemann, A., Reddy, V.S. Characterization of polymorphism displayed by the coat protein mutants of tomato bushy stunt virus. Virology 349:222, 2006. Schneemann, A. The structural and functional role of RNA in icosahedral virus assembly. Annu. Rev. Microbiol. 60:51, 2006. Singh, P., Destito, G., Schneemann, A., Manchester, M. Canine parvovirus-like particles, a novel nanomaterial for tumor targeting. J. Nanobiotechnol. 4:2, 2006. Walukiewicz, H.E., Johnson, J.E., Schneemann, A. Morphological changes in the T = 3 capsid of Flock House virus during cell entry. J. Virol. 80:615, 2006. Molecular Biology of Retroviruses J.H. Elder, A.P. de Parseval, Y.-C. Lin, S. de Rozieres, M. Sundstrom, K. Tam, M. Giffin,* H. Heaslet,* C.D. Stout, B.E. Torbett* * Department of Molecular and Experimental Medicine, Scripps Research ur research centers on the molecular characterization of retroviruses, with emphasis on feline immunodeficiency virus (FIV) and development of ways to interfere with the viral life cycle. FIV causes O THE SCRIPPS RESEARCH INSTITUTE an AIDS-like syndrome in domestic cats and has structural and functional similarities to HIV, the causative agent of AIDS in humans. Discovery of ways to interfere with FIV infection may ultimately result in development of treatments for infections in both cats and humans. In recent studies, we continued to focus on the molecular characterization of receptor interactions and the molecular basis for the development of drug resistance in the aspartic protease encoded by FIV. RECEPTOR STUDIES Like many strains of HIV, FIV uses the chemokine receptor CXCR4 to enter the primary target cell, the CD4 + T cell. However, unlike HIV, FIV does not use the cell-surface protein CD4 as a primary binding receptor. Rather, the feline lentivirus uses the activation antigen CD134 to initially bind to CD4+ T cells. CD134 is expressed on activated CD4 + T cells, a finding that explains why FIV can infect and kill CD4+ T cells, even though the virus does not bind CD4. As reported last year, we showed that interaction of the FIV surface glycoprotein gp95 with a soluble version of CD134 allows productive infection of cells that bear the entry receptor CXCR4 but lack cell-surface CD134. This finding is consistent with the notion that binding of CD134 causes a conformational change in gp95, which in turn increases the affinity of interaction with CXCR4 to facilitate infection of the target cell. These effects are similar to the effects of binding of soluble CD4 by gp120, the surface glycoprotein of HIV and indicate that although different primary receptors are involved, the actual mechanism of infection of FIV and HIV is strikingly similar. We speculate that the benefit of this type of binding cascade is to limit exposure of critical regions of the surface glycoproteins to the immune system until the primary binding event has already occurred, thus reducing the likelihood of virus neutralization. Using chimeric proteins consisting of feline and human CD134 (the human homolog does not bind FIV glycoprotein) and site-directed mutagenesis, we have mapped regions of feline CD134 involved in interaction with gp95. The results indicated that as few as 3 amino acids in the C-terminal part of outer domain 1 of feline CD134 are sufficient to impart FIV gp95 binding and receptor function to human CD134. Studies are in progress to map the regions of gp95 that bind CD134. Importantly, we have now a panel of antibodies that bind and neutralize FIV only after CD134 is bound; we have used peptides to map the region in MOLECULAR BIOLOGY 2006 which these CD134-dependent neutralizing antibodies react. These studies effectively map regions of the viral glycoprotein critical for CD134 interaction. Cocrystallization studies are under way to determine the structure of the region surrounding the antibody-binding site. These experiments will contribute to our understanding of the nature of receptor binding and will define targets for vaccine development. P R O T E A S E D R U G R E S I S TA N C E The aspartic protease of lentiviruses is responsible for processing the viral Gag and Pol polyproteins into the final gene products required for viral replication and must function efficiently to generate infectious virus. Drugs against HIV protease are keys to the success of highly active antiretroviral therapy used to treat, but not cure, patients infected with HIV. The substrate and inhibitor specificities of FIV differ from those of HIV. We investigated the nature of these differences to better understand the structural basis of development of resistance to therapy, an ongoing problem with current drugs used to treat HIV disease. In certain instances, similarities exist between amino acid positions that dictate differences in substrate specificity between FIV and HIV aspartic protease and those that mutate in response to drug treatment. Mutations in these sites increase the dissociation constant for a given drug, but at a cost in catalytic efficiency for the viral protease. Compensatory amino acid substitutions can then occur that increase the catalytic efficiency of the drug-resistant protease, thus increasing expression of virus despite drug treatment. We prepared mutants of FIV protease in which amino acids found in drug-resistant HIV protease were placed in the equivalent positions in the FIV enzyme. Then, using cells transduced with gag/pol gene expression vectors encoding HIV-FIV hybrid proteases, we tested the mutants for relative drug sensitivity. We found that the Gag/Pol polyproteins are processed by the hybrid proteases and have drug sensitivity profiles similar to those of HIV protease. However, the order of site cleavage, which is critical to generation of infectious virus, is altered by these specific changes. Studies are under way to establish a structural basis for this phenomenon. The findings highlight yet another potential approach to interrupting the viral life cycle. PUBLICATIONS Brik, A., Alexandratos, J., Lin, Y.-C., Elder, J.H., Olson, A.J., Wlodawer, A., Goodsell, D.S., Wong, C.-H. 1,2,3-Triazole as a peptide surrogate in the rapid synthesis of HIV-1 protease inhibitors. Chembiochem 6:1167, 2005. THE SCRIPPS RESEARCH INSTITUTE 221 de Parseval, A., Bobardt, M.D., Chatterji, A., Chatterji, U., Elder, J.H., David, G., Zolla-Pazner, S., Farzan, M.R., Lee, T.-H., Gallay, P.A. A highly conserved arginine in gp120 governs HIV-1 binding to both syndecans and CCR5 via sulfated motifs. J. Biol. Chem. 280:39493, 2005. de Parseval, A., Grant, C.K., Sastry, K.J., Elder, J.H. Sequential CD134-CXCR4 interactions in feline immunodeficiency virus (FIV): soluble CD134 activates FIV Env for CXCR4-dependent entry and reveals a cryptic neutralization epitope. J. Virol. 80:3088, 2006. Gonzalez-Lira, B., Rueda-Orozco, P.E., Galicia, O., Montes-Rodriguez, C.J., Guzman, K., Guevara-Martinez, M., Elder, J.H., Prospero-Garcia, O. Nicotine prevents HIVgp120-caused electrophysiological and motor disturbances in rats. Neurosci. Lett. 394:136, 2006. Heaslet, H., Kutilek, V., Morris, G.M., Lin, Y.-C., Elder, J.H., Torbett, B.E., Stout C.D. Structural insights into the mechanisms of drug resistance in HIV-1 protease NL4-3. J. Mol. Biol. 356:967, 2006. Liang, F.-S., Brik, A., Lin, Y.-C., Elder, J.H., Wong, C.-H. Epoxide in water and screening in situ for rapid discovery of enzyme inhibitors in microtiter plates. Bioorg. Med. Chem. 14:1058, 2006. Whiting, M., Muldoon, J., Lin, Y.-C., Silverman, S.M., Lindstrom, W., Olson, A., Kolb, H.C., Finn, M.G., Sharpless, K.B., Elder, J.H., Fokin, V.V. Inhibitors of HIV-1 protease by using in situ click chemistry. Angew. Chem. Int. Ed. 45:1435, 2006. Metalloenzyme Engineering D.B. Goodin, C.D. Stout, S. Vetter, E.C. Glazer, R.F. Wilson, A. Annalora, A.-M. Hays ur goals are to understand the diverse reactivity of heme enzymes and to use that information to generate engineered forms with novel catalytic properties. The primary hypothesis that has driven these studies is that the chemical reactivity displayed by these enzymes resides partially within the heme cofactor. One role of the protein is to limit or direct the access of substrates to the active site in ways that result in specific catalysis. In addition, many important examples exist in which the protein directly modulates the activity of the heme. Thus, our goals are to delineate the boundaries between these 2 roles for the protein and then use this information to introduce sites where nonnative substrates interact with the heme cofactor in ways that will induce new catalytic reactions. We use a number of techniques in structural biology and spectroscopy and strategies of rational protein redesign and molecular evolution. One area of emphasis is the basic physical, spectroscopic, and functional properties of heme enzymes. For example, the FeIII/FeII and FeII/FeI redox couples of inducible nitric oxide synthase have recently been measured by using direct cyclic voltammetry in organic films on graphite electrodes. These studies allow easy measurement of electron transfer between the enzyme and the electrode surface and have revealed the inter- O 222 MOLECULAR BIOLOGY 2006 conversion of several coordination states of the heme. The results will complement ongoing studies in which the enzymes are directly and homogeneously coupled to electrode surfaces by using molecular wires. In other research, we used cavity complementation to introduce small-molecule binding sites near the active site of a protein environment. This approach has provided new ways to test ideas about the diversity of the functions of heme enzymes and is a useful tool for testing predictions of protein-ligand interactions. For example, in a collaboration with B. Shoichet, University of California, San Francisco, we completed a study in which compounds in a database were docked into a buried engineered cavity that has an unusual specificity for charged ligands. Using x-ray crystallography, we verified the accuracy of the docking predictions for 15 of the top 16 compounds. In other studies, we used synthetic molecular wires, substrate analogs linked to photochemical or redox-active sensitizers, to bind at the active site of cytochrome P450, peroxidases, and nitric oxide synthase. These wires will be useful as reporters of the active-site environment and as triggers to study reaction mechanisms. In collaboration with H.B. Gray, California Institute of Technology, Pasadena, California, we recently solved structures of cytochrome P450cam bound to 2 such wires. Marked changes in the protein structure occurred near 2 helices that are similar to structural variations seen in mammalian P450s, suggesting that the degree of structural plasticity in prokaryotic P450s is similar to that of mammalian forms. In other research, we removed the proposed electron-transfer pathway from a peroxidase and replaced it with a solvent-filled channel. We have designed surrogate molecular wires to replace the native pathway, and we have shown that one of these binds the channel in a mode that is completely analogous to the native structure. This technique will provide new ways to test proposals about the specificity and structural requirements of this important structural element. Finally, we are designing and synthesizing a class of cofactor-linked ruthenium-diamine photosensitizers that are designed to specifically target nitric oxide synthase at the pterinbinding site, allowing the role of the cofactor in catalysis to be probed by direct-charge injection-withdrawal through the wire. PUBLICATIONS Brenk, R., Vetter, S., Boyce, S.E., Goodin, D.B., Shoichet, B. Probing molecular docking in a charged model binding site. J. Mol. Biol. 357:1449, 2006. THE SCRIPPS RESEARCH INSTITUTE Control of Cell Division S.I. Reed, C. Baskerville, L.-C. Chuang, S. Ekholm-Reed, M. Henze, J. Keck, V. Liberal, K. Luo, B. Olson, S. Rudyak, D. Tedesco, F. van Drogen, J. Wohlschlegel iological processes of great complexity can be approached by beginning with a systematic genetic analysis in which the relevant components are first identified and the consequences of selectively eliminating the components via mutations are investigated. We have used yeast, which is uniquely tractable to this type of analysis, to investigate control of cell division. In recent years, it has become apparent that the most central cellular processes throughout the eukaryotic phylogeny are highly conserved in terms of both the regulatory mechanisms used and the proteins involved. Thus, it has been possible in many instances to generalize from yeast cells to human cells. B CONTROL IN YEAST In recent years, we have focused on the role and regulation of the Cdc28 protein kinase (Cdk1). Initially identified by means of a mutational analysis of the yeast cell cycle, this protein kinase and its analogs are ubiquitous in eukaryotic cells and are central to a number of aspects of control of cell-cycle progression. One current area of interest is regulation of cellular morphogenesis by Cdk1. The activity of Cdk1 driven by mitotic cyclins modulates polarized growth in yeast cells. Specifically, these activities depolarize growth by altering the actin cytoskeleton. We found that several proteins that modulate actin structure are targeted by Cdk1, and we are investigating whether these phosphorylation events control actin depolarization. A second major area of interest is in the regulation of mitosis. A key aspect of mitotic regulation in yeast is the accumulation of Cdc20, which triggers the transition from metaphase to anaphase. Cdc20 is an essential cofactor of the protein-ubiquitin ligase known as the anaphase-promoting complex or APC/C. It is through the ubiquitin-mediated proteolysis of a specific anaphase inhibitor, securin (Pds1 in yeast), that anaphase is initiated. We found that cells are prevented from entering mitosis when DNA replication is blocked by the drug hydroxyurea, which causes the destabilization of Cdc20 and inhibition of Cdc20 translation. While investigating mitosis, we found that a Cks1, small Cdk1-associated protein, appears to regulate the proteasome. Proteasomes are complex proteases that tar- MOLECULAR BIOLOGY 2006 get ubiquitylated proteins, including important cell-cycle regulatory proteins. Surprisingly, we found that Cks1 regulates a nonproteolytic function of proteasomes, the transcriptional activation of Cdc20. Specifically, Cks1 is required to recruit proteasomes to the gene CDC20 for efficient transcriptional elongation. Our investigations of CDC20 have led to the conclusion that Cks1 is required for recruitment of proteasomes to and transcriptional elongation of many other genes as well. Currently, we are elucidating the mechanism whereby Cks1 recruits proteasomes and facilitates transcriptional elongation. Our most recent results suggest that Cks1 and proteasomes in conjunction with Cdk1 mediate remodeling of chromatin. CONTROL IN MAMMALIAN CELLS We showed previously that the human homologs of the Cdc28 protein kinase are so highly conserved, structurally and functionally, relative to the yeast protein kinase, that they can function and be regulated properly in a yeast cell. Analyzing control of the cell cycle in mammalian cells, we produced evidence for the existence of regulatory schemes, similar to those elucidated in yeast, that use networks of both positive and negative regulators. A principal research focus is the positive regulator of Cdk2, cyclin E. Cyclin E is often overexpressed and/or deregulated in human cancers. Using a tissue culture model, we showed that deregulation of cyclin E confers genomic instability, probably explaining the link to carcinogenesis. The observation that deregulation of cyclin E confers genomic instability has led us to hypothesize a mechanism of cyclin E–mediated carcinogenesis based on accelerated loss of heterozygosity at tumor suppressor loci. We are testing this hypothesis in transgenic mouse models. We showed that a cyclin E transgene expressed in the mammary epithelium markedly increases loss of heterozygosity at the p53 locus, leading to enhanced mammary carcinogenesis. We are extending these investigations by using mouse prostate, testis, and skin models. In an attempt to understand cyclin E–mediated genomic instability, we are investigating how deregulation of cyclin E affects both S phase and mitosis. Recent data suggest that deregulation of cyclin E impairs DNA replication by interfering with assembly of the prereplication complex. Cyclin E deregulation also impairs the transition from metaphase to anaphase by promoting the accumulation of inhibitors of anaphase. Our interest in cyclin E deregulation in cancer led us to investigate the pathway for turnover of cyclin E. THE SCRIPPS RESEARCH INSTITUTE 223 We showed that phosphorylation-dependent proteolysis of cyclin E depends on a protein-ubiquitin ligase known as SCF hCdc4 . The F-box protein hCdc4 is the specificity factor that targets phosphorylated cyclin E. We are investigating how ubiquitylation of cyclin E is coordinated with other processes required for its degradation, including prolyl isomerization. We are also investigating SCF hCdc4 ubiquitylation of other important cellular proteins. Recently, we began determining the role of SCFhCdc4 in neurodegenerative disease. We found that parkin, a protein often mutated in hereditary Parkinson’s disease, regulates the stability of hCdc4, possibly leading to neuropathologic changes. Consistent with this idea, we found that SCFhCdc4 targets peroxisome proliferator–activated receptor γ coactivator-1α which protects neurons from oxidative damage. In addition, we showed that SCF hCdc4 regulates the turnover of presenilins in the brain, proteins strongly implicated in Alzheimer’s disease. Another area of interest is the role of Cks proteins in mammals, complementing our research in yeast. Mammals express 2 orthologs of yeast Cks1, known as Cks1 and Cks2. Experiments in mice lacking the gene for Cks1 and Cks2 revealed that each ortholog has a specialized function. Cks1 is required as a cofactor for Skp2-mediated ubiquitylation and turnover of inhibitors p21, p27, and p130. Cks2 is required for the transition from metaphase to anaphase in both male and female meiosis I. Nevertheless, mice nullizygous at the individual loci are viable. However, doubly nullizygous mice have not been observed because embryos die at the morula stage, a finding consistent with an essential redundant function. We found that this function most likely is involved in regulation of transcription and is linked to chromatin remodeling, as in yeast. PUBLICATIONS Jackson, L.P., Reed, S.I., Haase, S.B. Distinct mechanisms control the stability of the related S-phase cyclins Clb5 and Clb6. Mol. Cell. Biol. 26:2456, 2006. Reed, S.I. Skp’n with Cks1: revelations from the Skp1-Skp2-Cks1-p27 structure. Mol. Cell 20:1, 2005. Reed, S.I. The ubiquitin-proteasome pathway in cell cycle control. Results Probl. Cell Differ. 42:147, 2006. Smith, A.P.L., Henze, M., Lee, J.A., Osborn, K.G., Keck, J., Tedesco, D., Bortner, D.M., Rosenberg, M.P., Reed, S.I. Deregulated cyclin E promotes p53 loss of heterozygosity and tumorigenesis in the mouse mammary gland. Oncogene, in press. Spruck, C., Sun, D., Fiegl, H., Marth C., Mueller-Holzner, E., Goebel, G., Widschwendter, M., Reed, S.I. Detection of low molecular weight derivatives of cyclin E1 is a function of cyclin E1 protein levels in breast cancer. Cancer Res. 66:7355, 2006. van Drogen, F., Sangfelt, O., Malyukova, A., Matskova, L., Yeh, E., Means, A.R., Reed, S.I. Ubiquitylation of cyclin E requires the sequential function of SCF complexes containing distinct hCdc4 isoforms. Mol. Cell 23:37, 2006. 224 MOLECULAR BIOLOGY 2006 Wittenberg, C., Reed, S.I. Cell cycle-dependent transcription in yeast: promoters, transcription factors, and transcriptomes. Oncogene 24:2746, 2005. Wohlschlegel, J.A., Johnson, E.S., Reed, S.I., Yates, J.R. III. Improved identification of SUMO attachment sites using C-terminal SUMO mutants and tailored protease digestion strategies. J. Proteome Res. 5:761, 2006. THE SCRIPPS RESEARCH INSTITUTE expressed as an MBF target during late G1 phase, Nrm1 associates with MBF at target promoters and represses expression as cells enter S phase (Fig. 1). Similarly, the Nrm1 homolog, SpNrm1, in the fission yeast Schizosaccharomyces pombe regulates its only G1-specific transcription factor, MBF. Transcriptional and Proteolytic Control of Cell Proliferation and Adaptation to Environmental Stimuli C. Wittenberg, M. Ashe, R. de Bruin, B.-K. Han. M. Guaderrama, T. Kalashnikova ellular decision making and coordination of cellular events often involves the differential regulation of the expression of genes. Recently, we have focused on the mechanisms through which cells exert control over gene expression to regulate cell proliferation and the response to changes in environmental conditions. C R E G U L AT I O N O F C E L L P R O L I F E R AT I O N In most cells, commitment to a new round of cell division during the G1 phase of the cell cycle is accompanied by the activation of a large family of genes that encode activities involved in the duplication and segregation of cellular components. G1-specific genes also encode regulatory factors that promote subsequent cell-cycle events. In the budding yeast Saccharomyces cerevisiae, G1-specific genes are regulated by 2 transcription factors: SBF and MBF. Using mass spectrometry–based multidimensional protein identification technology, we have identified novel regulators of these transcription factors. SBF acts as a transcriptional activator and promotes expression of its targets specifically during the G 1 interval. We established that promoter-bound SBF associates with the Whi5 repressor during early G 1 phase and that Whi5 is inactivated via phosphorylation by a G1-specific cyclin-dependent protein kinase, thereby activating transcription (Fig. 1). This regulation is analogous to the regulation of E2F by the tumor suppressor Rb in metazoans. MBF, in conjunction with specific corepressors, acts primarily as a transcriptional repressor and limits transcription of target genes to the G1 phase. We identified Nrm1, a novel MBF-associated corepressor. When F i g . 1 . Transcriptional circuitry regulating G1-specific gene expression. G1-specific transcription in S cerevisiae is regulated by 2 heterodimeric transcription factors: SBF and MBF. Both transcription factors are bound to promoters during G1 phase before commitment to a new cell cycle. Commitment occurs when their target genes are activated by the action of the cyclin-dependent protein kinase Cln3/CDK1. For SBF, Cln3/CDK1 activates transcription by phosphorylation and inactivation of the SBF-specific repressor Whi5. Once activated, G1specific transcription leads to the accumulation of many proteins, including those that promote repression of G1-specific transcription. Nrm1 is an MBF-specific corepressor encoded by an MBF target gene. Together, these regulatory proteins can explain the confinement of G1-specific transcription to the G1 phase. The G1-specific transcriptional machinery is regulated by checkpoints that monitor the integrity of cellular structures and processes. When replication forks are stalled during S phase in the fission yeast, repression of MBF-regulated transcription is disrupted. We have shown that that response, which requires the Rad3 (ATM) and Cds1 (Chk2) checkpoint protein kinases, leads to the phosphorylation of SpNrm1 and dissociation from MBF-regulated promoters. Unexpectedly, according to the literature, derepression of MBF target genes also occurs via regulation of Nrm1 in response to activation of the DNA replication checkpoint in budding yeast. Consequently, replication stress appears to be associated with genomic instability in the absence of Nrm1. A D A P TAT I O N T O E N V I R O N M E N TA L S T I M U L I Adaptation to environmental changes generally involves remodeling of the gene expression program. We have studied the regulation of the HXT genes, which encode hexose permeases, in response to extracellular glucose. Those genes are induced by glucose and are repressed for most other carbon sources. Extracellular glucose interacts with the Snf3 and Rgt2 receptors, MOLECULAR BIOLOGY 2006 initiating a signaling cascade that culminates with the activation of HXT gene transcription. We have shown that signaling leads to the phosphorylation-dependent destruction of a transcriptional corepressor, Mth1, by the E3 ubiquitin ligase SCFGrr1. Destruction of Mth1 leads to the phosphorylation of the transcriptional repressor Rgt1 and its dissociation from HXT gene promoters. Conversely, repression of HXT gene expression requires Mth1 and is associated with Rgt1 dephosphorylation. We recently identified a type 2A protein phosphatase complex involved in Rgt1 dephosphorylation and are actively pursuing the protein kinase involved in Rgt1 phosphorylation. Interestingly, SCFGrr1, the E3 ubiquitin ligase required for HXT gene induction, is also important for destruction of phosphorylated G1 cyclins, critical regulators of cell-cycle initiation. We are investigating the basis for discrimination between targets by SCFGrr1. We found that basic residues in the leucine-rich repeat and parts of the carboxy terminus of the F-box protein Grr1 are important for recognition of phosphorylated substrates. Our recent identification of additional substrates and additional characterization of Grr1 are facilitating those studies. PUBLICATIONS de Bruin, R., Kalashnikova, T.I., Chawan, C., McDonald, W.H., Wohlschlegel, J.A., Yates, J. III, Russell, P., Wittenberg, C. Constraining G1-specific transcription to late G1 phase: the MBF-associated corepressor Nrm1 acts via negative feedback. Mol. Cell 23:483, 2006. Cell-Cycle Checkpoints, DNA Damage, and Oxidative Stress Responses P. Russell, C. Chahwan, C. Dovey, L.-L. Du, P.-H. Gaillard, V. Martin, B.A. Moser, T.M. Nakamura, M.A. Rodríguez-Gabriel, J. Williams, Y. Yamada NA damage and oxidative stress elicit cellular responses that are highly conserved throughout eukaryotic evolution. Consequently, studies of genetically tractable microorganisms such as the fission yeast Schizosaccharomyces pombe can provide a useful framework for the design and interpretation of experiments with more complex multicellular organisms. We use S pombe to study cell-cycle checkpoints, DNA repair, and stress response mechanisms. Defects in these mechanisms underlie a number of human diseases, including cancer. D THE SCRIPPS RESEARCH INSTITUTE 225 CHECKPOINTS The DNA replication and damage checkpoints prevent the onset of mitosis when DNA replication is interrupted or when DNA is damaged. A single doublestrand break is sufficient to arrest the cell cycle. One aim of our studies is to understand how cells detect DNA damage and transmit a checkpoint signal that arrests the cell cycle. Chk1 is the effector kinase of the DNA damage checkpoint. It regulates the activities of Cdc25 and Mik1/Wee1 proteins, which modulate the inhibitory phosphorylation of the cyclin-dependent kinase Cdc2. Chk1 activation by Rad3 requires the adaptor protein Crb2. Crb2 is rapidly recruited to double-strand breaks in DNA. Rad3 and Tel1 (the ATM homolog in fission yeast) stimulate Crb2 recruitment by phosphorylating a serine residue near the C terminus of histone H2A in the vicinity of double-strand breaks. Our data indicate that tandem C-terminal BRCT domains in Crb2 associate directly with phosphorylated histone H2A. Crb2 recruitment to double-strand breaks also requires the constitutive methylation of lysine at position 20 in histone H4. This step most likely involves a direct interaction with a Tudor motif in Crb2 that is located to the N-terminal side of the BRCT motifs. We recently found that these 2 histone modifications cooperate in a nonredundant mechanism to promote recruitment of Crb2 to double-strand breaks (Fig. 1). Remarkably, neither histone modification is required for recruitment of Crb2 to sustained double-strand breaks that cannot be repaired by homologous recombination. We recently discovered that the histone modification– independent recruitment of Crb2 to double-strand breaks involves association between phosphorylated threonine215 in Crb2 and another checkpoint protein known as Cut5. In future studies, we will determine whether the mechanisms that regulate Crb2 in fission yeast are conserved for the analogous proteins in human cells. DNA REPAIR Bloom, Warner, and Rothmund-Thomson syndromes in humans are typified by predisposition to cancer or premature aging. These syndromes, which all result from defects in DNA helicases, are characterized by genomic instability arising from inappropriate homologous recombination. To better understand this process, we used a 2-hybrid screen to identify novel proteins that associate with Srs2 DNA helicase in fission yeast. We discovered a previously uncharacterized protein that promotes the formation of toxic recombination struc- 226 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE current studies are aimed at providing a deeper biochemical and structural understanding of the SWS1XRCC2-RAD51D complex. O X I D AT I V E S T R E S S R E S P O N S E F i g . 1 . Parallel mechanisms of recruiting the DNA damage checkpoint protein Crb2 to sites of DNA damage. Top, A proposed mechanism of how Crb2 associates with double-strand breaks. One mode of association involves interactions with modified histones. The tandem C-terminal BRCT domains associate with the phosphory- lated C-terminal region of histone H2A. The Tudor domain interacts with the constitutive methylation of histone H4 on lysine at position 20. These modes of interaction are not redundant. A third mode of interaction involves the phosphorylated threonine at position 215 (T215) region of Crb2 and the Cut5. Cut5 is proposed to specifically bind to the single-stranded DNA region near the end of the double-strand break. This binding might involve the association of Cut5 with other proteins that bind to single-stranded DNA. Bottom, Fission yeast cells that express Crb2 were tagged with yellow fluorescent protein (YFP) and Cut5 tagged with cyan fluorescent protein (CFP). The cells were engineered to express the HO endonuclease and to contain a single HO cleavage site. The YFP-Crb2 and Cut5-CFP foci indicate large-scale accumulation of these proteins at the site of the double-strand break created by HO endonuclease. tures in yeast mutants that lack one or more DNA helicases. This protein, which we christened Sws1 because it has a SWIM-type zinc finger, is conserved from yeast to humans. In collaborative mass spectrometry and proteomics studies with J.R. Yates, Department of Cell Biology, we found that Sws1 forms a complex with 2 other proteins known as Rlp1 and Rdl1. Bioinformatic analysis revealed that these 2 proteins are Rad51 paralogs (i.e., homologous sequences derived from gene duplication) that promote the formation of the Rad51 nucleoprotein filament during homologous recombination. Rlp1 and Rdl1 are equivalent to human XRCC2 and RAD51D, proteins implicated in other human diseases characterized by genomic instability. In collaboration with C.H. McGowan, Department of Molecular Biology, we found that using small interfering RNA to silence the gene for human SWS1 reduced the occurrence of homologous recombination structures. Our Oxidative stress caused by reactive oxygen species can be highly toxic, causing damage to proteins, lipids, and nucleic acids. Oxidative stress elicits a complex gene expression response that is orchestrated in large part by MAP kinase cascades. The fission yeast Spc1 MAP kinase pathway is homologous to the p38 pathway in humans. We recently discovered Csx1, a protein that collaborates with Spc1 to control gene expression in response to oxidative stress. Csx1 is an RNA-binding protein that mediates overall control of gene expression in response to oxidative stress by binding and stabilizing mRNA that encodes Atf1, a transcription factor that is also regulated by Spc1. Most recently, we have focused on a newly discovered family of proteins that interact with Csx1. We have named these proteins Cip1 and Cip2 (for Csx1-interacting proteins 1 and 2). Remarkably, elimination of Cip1 or Cip2 results in substantial recovery of the sensitivity of Csx1 mutant cells to oxidative stress, suggesting that Cip1 and Cip2 are part of a mechanism that degrades Atf1 mRNA. PUBLICATIONS Cavero, S., Chahwan, C., Russell, P. Xlf1 is required for DNA repair by nonhomologous end-joining in Schizosaccharomyces pombe. Genetics, in press. Coulon, S., Noguchi, E., Noguchi, C., Du, L.-L., Nakamura, T.M., Russell, P. Rad22Rad52-dependent repair of ribosomal DNA repeats cleaved by Slx1-Slx4 endonuclease. Mol. Biol. Cell 17:2081, 2006. de Bruin, R.A.M., Kalashnikova, T.I., Chahwan, C., McDonald, W.H., Wohlschlegel, J., Yates III, J.R., Russell, P., Wittenberg, C. Constraining G1-specific transcription to late G1-phase: The MBF-associated corepressor Nrm1 acts via negative feedback. Mol. Cell. 23:483, 2006. Du, L.-L., Nakamura, T.M., Russell, P. Histone modification-dependent and -independent pathways for recruitment of checkpoint protein Crb2 to double-strand breaks. Genes Dev. 20:1583, 2006. Martin, V., Chahwan, C., Gao, H., Blais, V., Wohlschlegel, J., Yates, J.R. III, McGowan, C.H., Russell, P. Sws1 is a conserved regulator of homologous recombination in eukaryotic cells. EMBO J. 25:2564, 2006. Martín, V., Rodríguez-Gabriel, M.A., McDonald, W.H., Watt, S., Yates, J.R. III, Bähler, J., Russell, P. Cip1 and Cip2 are novel RNA-recognition-motif proteins that counteract Csx1 function during oxidative stress. Mol. Biol. Cell 17:1176, 2006. Matsumoto, S., Ogino, K., Noguchi, E., Russell, P., Masai, H. Hsk1-Dfp1/Him1, the Cdc7-Dbf4 kinase in Schizosaccharomyces pombe, associates with Swi1, a component of the replication fork protection complex. J. Biol. Chem. 280:42536, 2005. Nakamura, T.M., Moser, B.A., Du, L.-L., Russell, P. Cooperative control of Crb2 by ATM family and Cdc2 kinases is essential for the DNA damage checkpoint in fission yeast. Mol. Cell. Biol. 25:10721, 2005. Rodríguez-Gabriel, M.A., Russell, P. Distinct signaling pathways respond to arsenite and reactive oxygen species in Schizosaccharomyces pombe. Eukaryot. Cell 4:1396, 2005. MOLECULAR BIOLOGY 2006 Rodriguez-Gabriel, M.A., Watt, S., Bähler, J., Russell, P. Upf1, an RNA helicase required for nonsense-mediated mRNA decay, modulates the transcriptional response to oxidative stress in fission yeast. Mol. Cell. Biol. 26:6347, 2006. DNA Damage Responses in Human Cells C.H. McGowan, V. Blais, M. Duquette, E. Langley, A. MacLaren, J. Scorah, D. Slavin, E. Taylor omplex multicellular organisms, such as humans, have large numbers of mitotically competent cells that are capable of renewal, repair, and, to some extent, regeneration. The advantages of being able to replace damaged or aged cells are off set by the inherent susceptibility of mitotic cells to acquiring mutations and becoming cancerous. DNA is inherently vulnerable to many sorts of chemical and physical modification; thus, as cells duplicate and divide, they can acquire mutations. Both spontaneous and induced DNA damage must be repaired with minimal changes if growth, renewal, and repair are to be successful. Our overall objective is to understand how mammalian cells protect themselves from DNA damage and thus from developing cancer. Eukaryotic cells have evolved with a complex network of DNA repair processes and cell-cycle checkpoint responses to ensure that damaged DNA is repaired before it is replicated and becomes fixed in the genome. These pathways are highly conserved throughout evolution, and much information about human responses to DNA damage has been gained from studies of simple, genetically tractable organisms such as yeast. We use a combination of molecular, cellular, and genetic techniques to determine how these pathways operate in human cells. Checkpoints control the order and timing of events in the cell cycle; they ensure that biochemically independent processes are coupled so that a delay in a critical cell-cycle process will cause a delay in all other aspects of progression of the cycle. In addition, checkpoints also coordinate repair with delays in progression of the cell cycle and promote the use of the most appropriate repair pathway. We used genetic models to identify 2 checkpoint kinases in humans that limit progression of the cell cycle when DNA is damaged. One of these kinases, Chk2, is activated in response to DNA damage. Chk2 physically interacts with Mus81-Eme1, a C THE SCRIPPS RESEARCH INSTITUTE 227 conserved DNA repair protein that has homology to the xeroderma pigmentosum F family of endonucleases. Xeroderma pigmentosum is a cancer-prone disorder that results from a failure to appropriately repair damaged DNA. Biochemical analysis shows that Mus81-Eme1 has associated endonuclease activity against structure-specific DNA substrates, including Holliday junctions. Enzymatic analysis, immunofluorescence studies, and the use of RNA interference have all contributed to the conclusion that Mus81-Eme1 is required for recombination repair in human cells. We are also using gene targeting to study the function of the Mus81-Eme1 endonuclease in mice. Inactivation of Mus81 in mice increases genomic instability and sensitivity to DNA damage but does not promote tumorigenesis. In addition, we showed that Mus81-Eme1 is specifically required for survival after exposure to cisplatin, mitomycin C, and other commonly used anticancer drugs. As a point of interaction between checkpoint control and DNA repair, the relationship between Mus8-Eme1 and Chk2 most likely provides information critical to understanding the response to DNA damage as a whole. Anticancer therapy is largely based on the use of genotoxic agents that damage DNA and thus kill dividing cells. Coordination of cell-cycle checkpoints and DNA repair is especially important when unusually high amounts of DNA damage occur after radiation or genotoxic chemotherapy. Hence, a detailed understanding of cellular responses to DNA damage is essential in understanding both the development and the treatment of disease in humans. PUBLICATIONS Martin, V., Chawan, C., Gao, H., Blais, V., Wohlschlegel, J., Yates, J.R. III, McGowan, C.H., Russell, P. Sws1 is a conserved regulator of homologous recombination in eukaryotic cells. EMBO J. 25:2564, 2006. DNA Repair and the Maintenance of Genomic Stability M.N. Boddy, S. Pebernard, J. Prudden NA repair pathways have evolved to protect the genome from ever-present genotoxic agents. Highlighting the importance of the pathways, defects in DNA repair mechanisms strongly predispose the host to cancer and to neurologic and developmental D 228 MOLECULAR BIOLOGY 2006 disorders. The DNA repair systems we study in fission yeast are evolutionarily conserved, and therefore our studies provide a valuable framework for understanding genome maintenance in human cells. Although many DNA repair mechanisms have been described, information on how they are coordinated with necessary changes in chromatin structure is limited. We are studying the essential structural maintenance of chromosomes (SMC) complex Smc5-Smc6. The molecular functions of Smc5-Smc6 are unknown, but the complex is related to the SMC complexes that hold replicated sister chromatids together (cohesin) and condense chromatin before its segregation at mitosis (condensin). In collaboration with J.R. Yates, Department of Cell Biology, we purified the Smc5-Smc6 complex and determined the identity of the core components. The holocomplex consists of the Smc5-Smc6 heterodimer and 6 additional non-SMC elements, Nse1–Nse6. We expressed and purified individual components of the complex and determined the architecture of the holocomplex (Fig. 1). THE SCRIPPS RESEARCH INSTITUTE cation of target proteins with ubiquitin and the small ubiquitin-like protein SUMO. Such protein modifications play roles in DNA repair and chromatin remodeling. We have carried out detailed genetic and biochemical analyses of the Nse5-Nse6 heterodimer. Nse5 and Nse6 are not essential for growth; however, cells lacking either protein have high levels of spontaneous genome damage and are hypersensitive to ultraviolet light and other genotoxic agents. An important discovery is that Nse5-Nse6 prevents the deleterious engagement of an ordinarily beneficial DNA repair pathway called homologous recombination. Our studies indicate that Nse5Nse6, and by extension the Smc5-Smc6 complex, acts either to prevent initiation of homologous recombination or to separate physically linked chromosomes that arise late in this process (Fig. 1B). Abrogating homologous recombination by deleting a pivotal factor required for the process (called Rad51) reduces the sensitivity of Nse5-Nse6 mutant cells to DNA damage. The spontaneous DNA damage observed in Nse5-Nse6 mutant cells is due to the attempted separation of chromosomes into daughter cells while the chromosomes are still physically linked. Such defective chromosome separation in humans could result in cancer and other diseases. PUBLICATIONS Pebernard, S., Wohlschlegel, J., McDonald, W.H., Yates, J.R. III, Boddy, M.N. The Nse5-Nse6 dimer mediates DNA repair roles of the Smc5-Smc6 complex [published correction appears in Mol. Cell. Biol. 26:3336, 2006]. Mol. Cell. Biol. 26:1617, 2006. Raffa, G.D., Wohlschlegel, J., Yates, J.R. III, Boddy, M.N. SUMO-binding motifs mediate the Rad60-dependent response to replicative stress and self-association. J. Biol. Chem. 281:27973, 2006. F i g . 1 . Architecture and function of the Smc5-Smc6 holocomplex. A, Nse1, Nse3, and Nse4 form a stable heterotrimer that associates with Smc5. Nse2 interacts directly with Smc5 in the absence of the other Nse proteins. Smc6 interacts directly with Smc5 but with none of the other components. Nse5 and Nse6 form a stable heterodimer that also binds directly to Smc5. Double-headed arrows indicate interactions between subcomplexes. Nse5-Nse6 may recruit the holocomplex to stalled replication forks and certain DNA damage sites. B, Nse5-Nse6 might act to prevent the initiation of homologous recombination catalyzed by a number of factors, including Rad51. Nse5-Nse6 could be involved in the separation or “resolution” of physically linked chromosomes that can result from homologous recombination. Evidence suggests that Nse5-Nse6 and Smc5-Smc6 perform such functions at replication forks and DNA double-strand breaks. Nse1–Nse4 are essential for growth, and hypomorphic mutants of these proteins cause cellular sensitivity to genotoxic agents such as ultraviolet light and x-rays. Notably, Nse1 and Nse2 contain certain zinc finger domains that implicate these 2 elements in the modifi- Delineating Oncogenic and Tumor-Suppressing Signal Transduction Pathways P. Sun, Q. Deng, C. Kannemeier, R. Liao, A. Seit-Nebi, N. Yoshizuka evelopment of cancer is a result of multiple oncogenic genetic alterations, including activation of oncogenes and inactivation of tumor suppressors. Despite the essential roles of these mutations in tumor formation, normal cells usually respond to these oncogenic changes by initiating tumor-suppressing defense mechanisms such as premature senescence and apoptosis. Our main interests are delineating the signal transduction pathways that mediate these tumor-sup- D MOLECULAR BIOLOGY 2006 pressing responses and determining how oncogenes allow a cell to evade the regulation by these cellular defense mechanisms to cause cancer. Currently, we are focusing on 2 well-known oncogenes: ras and mdm2. The oncogene ras encodes a family of small GTPbinding proteins that transduce mitogenic signals from extracellular growth factors. Constitutive activation of ras is common in tumors and contributes to tumor development. In normal cells, however, the initial response to ras activation is a stable growth arrest called premature senescence. As a result, the senescence response triggered by ras must be evaded before transformation can occur. We showed that ras induces senescence through sequential activation of 2 MAP kinase pathways. Initially, ras activates the MAP kinase kinase (MEK)–extracellular signal–regulated kinase (ERK) pathway. Sustained activation of MEK-ERK turns on the stress-induced p38 pathway, which subsequently causes senescence. These studies have revealed a novel, tumor-suppressing function of p38, in addition to its known roles in inflammation and stress responses. In other studies, we identified additional signaling components, either upstream or downstream of p38, that mediate premature senescence. We found that 1 of the 4 isoforms of p38 contributes to ras-induced senescence by activating the p53 tumor suppressor protein. In addition, a serine/threonine protein kinase, which is a direct substrate of p38, also plays an essential role in ras-induced senescence. Inactivation of this protein kinase disrupts ras-induced senescence and promotes tumorigenesis both in vitro and in vivo. Our results have confirmed the tumor-suppressing function of the p38 pathway. To determine how premature senescence is bypassed in tumors, we dissected the functions of an adenovirusencoded oncoprotein, E1A, that can rescue ras-induced senescence. Our results indicated that bypassing of senescence requires binding of the cellular proteins Rb and p300/CBP by E1A. Although interference with the p16INK4A/Rb pathway or p300/CBP functions alone did not result in bypassing of senescence, these 2 types of genetic alterations cooperated to rescue cells from rasinduced senescence and lead to cellular transformation. These results indicate that p300 and CBP are integral components of the senescence pathway. Both p300 and CBP have tumor-suppressing functions. The critical role of p300 and CBP in the senescence response has provided a mechanistic basis for the tumor-suppressing function of these proteins. Another focus of our research is mdm2, an oncogene that can mediate transformation primarily through THE SCRIPPS RESEARCH INSTITUTE 229 inactivation of the tumor suppressor protein p53. However, we found that MDM2 confers resistance to a growth-inhibitory cytokine, transforming growth factor β, through a p53-independent mechanism. We are delineating this p53-independent activity of MDM2, which may play an important role in tumorigenesis. We have identified several MDM2 domains and activities that are essential for the ability of MDM2 to mediate resistance to the growth factor. In other research, we are systematically searching for genetic alterations that contribute to specific tumorassociated phenotypes, such as drug resistance, cellular immortalization, and metastasis. For these studies, we are using cDNA expression libraries or libraries of short interfering RNAs. PUBLICATIONS Lin, S., Xiao, R., Sun, P., Xu, X., Fu, X.D. Dephosphorylation-dependent sorting of SR splicing factors during mRNP maturation. Mol. Cell 20:413, 2005. Hypocretins in Arousal, Feeding Behavior, and Motivation J.G. Sutcliffe, L. de Lecea he 2 C terminally amidated hypocretin neuropeptides (also called orexins) are produced from a precursor whose expression in rats is restricted to a few thousand neurons of the lateral hypothalamus. These neurons are active during wakefulness but are quiescent during various phases of sleep. Two G protein–coupled hypocretin receptors have different distributions within the CNS. The hypocretins are found in secretory vesicles at synapses of fibers that project to areas within the posterior part of the hypothalamus that are implicated in feeding behaviors and hormone secretion. Hypocretin fibers also project to diverse targets in other brain regions and the spinal cord, including several areas implicated in cardiovascular function and sleep-wake regulation. The peptides are excitatory when applied directly in vivo. Most humans with narcolepsy have greatly reduced levels of hypocretin peptides in their cerebral spinal fluid and no or barely detectable hypocretin neurons in their hypothalami, findings suggestive of autoimmune attack. Hypocretin peptides excite noradrenergic neurons in the locus coeruleus and serotonergic neurons in the dorsal raphe to elevate muscle tone and histaminergic tuberomammillary neurons to promote wakefulness. T 230 MOLECULAR BIOLOGY 2006 These components of the ascending reticular activating system, and the hypocretin neurons themselves, project to and stimulate thalamic and basal forebrain neurons, and all of these groups contribute to the depolarization of the cerebral cortex. Arousal-related signaling occurs through both hypocretin receptors. These peptides have diverse effects on brain reward and autonomic systems related to stress that increase motivated behaviors, including feeding. The relation to feeding is complex. Acute administration of hypocretin peptides to sleeping rats increases food consumption. However, patients and animals with impaired hypocretin signaling have an increased likelihood of being obese despite reduced daily calorie intake. PUBLICATIONS de Lecea L., Sutcliffe, J.G. The hypocretins and sleep. FEBS J. 272:5675, 2005. Desplats, P.A., Kass, K.E., Gilmartin, T., Stanwood, G.D., Woodward, E.L., Head, S.R., Sutcliffe, J.G., Thomas, E.A. Selective deficits in the expression of striatalenriched mRNAs in Huntington’s disease. J. Neurochem. 96:743, 2006. Hedlund, P.B., Huitrón-Reséndiz, S., Henriksen, S.J., Sutcliffe, J.G. 5-HT7 receptor inhibition and inactivation induce antidepressantlike behavior and sleep pattern. Biol. Psychiatry 58:831, 2005. Hedlund, P.B., Sutcliffe, J.G. 5-HT7 receptors as favorable pharmacological targets for drug discovery. In: The Serotonin Receptors: From Molecular Pharmacology to Human Therapeutics. Roth, B.L. (Ed.). Humana Press, Totowa, NJ, 2006, p. 517. Hilbush, B.S., Morrison, J.H., Young, W.G., Sutcliffe, J.G., Bloom, F.E. New prospects and strategies for drug target discovery in neurodegenerative disorders. NeuroRx 2:627, 2005. THE SCRIPPS RESEARCH INSTITUTE nia of short, intermediate, and long duration and from the prefrontal cortex of matched control subjects without schizophrenia. Among many genes and pathways revealed as significantly altered in schizophrenia, we are focusing on those related to glycosphingolipid metabolism and myelination. A total of 40 genes with altered expression in patients with schizophrenia were related to these systems. To assess the effects of treatment with antipsychotic drugs on a subset of genes that encode structural components of myelin, we treated groups of mice with haloperidol, a widely prescribed “typical” antipsychotic drug. Chronic haloperidol treatment caused significant decreases in the expression levels of at least 8 myelin-related genes in several white matter regions of mouse brain as revealed by in situ hybridization analysis. In other studies, we are investigating the molecular basis for heterogeneity in schizophrenia by identifying genes that have similar expression profiles in subsets of patients with the disorder. Using weighted gene coexpression network analyses, we identified distinct subtypes in our schizophrenia cohort, most notably, dramatic differences in the expression profiles between patients with short versus long duration of illness. We are exploring subtype-specific pathways associated with these subgroups. T R A N S C R I P T I O N A L D Y S R E G U L AT I O N I N Sutcliffe, J.G. de Lecea, L. The hypocretin/orexin system. In: Handbook of Contemporary Neuropharmacology. Sibley, D.R., et al. (Eds.). Wiley-InterScience, Hoboken, NJ, in press. Sutcliffe, J.G., de Lecea, L. Hypocretins/orexins in brain function. In: Handbook of Neurochemistry and Molecular Neurobiology: Neuroactive Proteins and Peptides, 3rd ed. Lim, R. (Volume Ed.), Lajtha, A. (Series Ed.). Springer, New York, 2006, p. 499. Molecular Neurobiology of CNS Disorders E.A. Thomas, J.G. Sutcliffe, P.A. Desplats, S. Narayan, K.E. Kass, T. Gilmartin, L. Schaffer, S.R. Head GENE PROFILING IN SCHIZOPHRENIA chizophrenia is a life-long, heterogeneous mental illness with variable expression and unknown etiology. We are interested in the molecular factors that influence the course of illness in schizophrenia and how treatment modifies these factors. Using oligonucleotide microarrays, we generated gene expression profiles from tissue samples obtained at autopsy from the prefrontal cortex of patients with schizophre- S H U N T I N G T O N ’ S D I S E A S E : S T R I ATA L S P E C I F I C I T Y Much evidence supports a role for transcriptional dysregulation in Huntington’s disease. Of particular interest is how these disturbances may be specifically manifested in the striatum, the primary region of neurodegeneration in Huntington’s disease. Using microarray analysis and a transgenic mouse of Huntington’s disease, we identified a cluster of striatal-enriched genes that was downregulated in the mice. The cluster included the genes FoxP1, Bcl11b, and DRRF, which encode zinc finger–containing transcription factors, and RARB and RXRG, which encode nuclear receptors. Real-time polymerase chain reaction validated 57% and 40% reductions in levels of Bcl11b and FoxP1 mRNA, respectively, in the striatum of symptomatic transgenic mice and a 73% decrease in the expression of FoxP1 in human caudate from patients with Huntington’s disease. Transcripts for both of these factors are expressed in medium spiny projection neurons, which selectively degenerate in Huntington’s disease. Further colocalization and coimmunoprecipitation studies have suggested that Bcl11b and FoxP1 interact with polyglutamine- MOLECULAR BIOLOGY 2006 expanded N-terminal huntingtin. Sequestration of these factors into nuclear aggregates in Huntington’s disease resulting in loss of function may contribute to specific dysregulation of striatal gene expression. This mechanism may explain, in part, the specificity of the pathologic changes associated with Huntington’s disease. PUBLICATIONS Desplats, P.A., Kass, K.E., Gilmartin, T., Stanwood, G.D., Woodward, E.L., Head, S.R., Sutcliffe, J.G. Thomas. E.A. Selective deficits in the expression of striatalenriched mRNAs in Huntington’s disease. J. Neurochem. 96:743, 2006. Thomas, E.A. Apolipoprotein D and arachidonic acid interactions in the treatment and pathology of schizophrenia. In: Fatty Acids and Oxidative Stress in Neuropsychiatric Disorders. Yao, J.K. (Ed.). Nova Science Publishers, Inc., Hauppauge, NY, 2006. Narayan, S., Kass, K.E., Thomas, E.A. Chronic haloperidol treatment results in a decrease in the expression of myelin/oligodendrocyte-related genes in the mouse brain. J. Neurosci. Res., in press. Thomas, E.A., Yao, J.K. Clozapine specifically alters the arachidonic acid pathway in mice lacking apolipoprotein D. Schizophr. Res., in press. Thomas, E.A. Molecular profiling of antipsychotic drug function: convergent mechanisms. In: The Pathology and Treatment of Psychiatric Disorders. Molecular Neurobiology, in press. Thomas, E.A. Striatal specificity of gene expression dysregulation in Huntington's disease. J. Neurosci. Res. 84:1151, 2006. THE SCRIPPS RESEARCH INSTITUTE 231 at the 5-HT7 receptor. Thus, both blockade and inactivation of the 5-HT7 receptor yield the same result. Sleep disturbances are common in depression. Increased amounts of REM sleep are a frequent finding. Compared with mice that have the 5-HT7 receptor mice lacking the receptor spend less time in REM sleep without alteration of other sleep parameters, further establishing the antidepressant-like profile of the animals that lack the receptor. Taken together our results suggest an important role for the 5-HT7 receptor in depression, and antagonists to this receptor should be evaluated as a treatment for depression. OBSESSIVE-COMPULSIVE DISORDER Obsessive-compulsive disorder is related to depression, at least to the extent that antidepressants are commonly used to treat both disorders. In an animal model of obsessive-compulsive disorder (marble burying), we showed that blockade or inactivation of the 5-HT7 receptor results in less compulsive behavior. Thus, the 5-HT7 receptor might be of interest as a putative target for treatment of obsessive-compulsive disorder. SCHIZOPHRENIA The 5-HT7 Receptor in Neuropsychiatric Disorders P.B. Hedlund, P.E. Danielson, S. Huitrón-Reséndiz, S.J. Henriksen, S. Semenova, M.A. Geyer, A. Markou, J.G. Sutcliffe nterest in the serotonin 5-HT7 receptor as a putative target in neuropsychiatric disorders has been growing continually. The interest was prompted by the finding that several classes of drugs used to treat disorders such as depression and schizophrenia have high affinity for the 5-HT 7 receptor. We have established evidence that supports a role for this receptor in depression, obsessive-compulsive disorder, and schizophrenia. I DEPRESSION The forced swim test and the tail suspension test are animal models of behavioral despair that have high value for predicting the antidepressant efficacy of drugs. The tests can also be used to characterize animals in which genes have been deleted. Using both of these tests, we showed that mice lacking the 5-HT7 receptor have a behavioral profile similar to that of mice treated with antidepressants. We replicated these findings by using a compound that acts as a selective antagonist Prepulse inhibition (PPI) of the acoustic startle reflex is a well-characterized animal model of schizophrenia. The model is especially relevant because similar responses can be observed in patients with schizophrenia. We showed that PPI per se is not altered in mice lacking the 5-HT7 receptor, but that when PPI is disrupted by phencyclidine, the mice are significantly less affected than are mice that have the receptor. Phencyclidine-induced disruption involves a glutamatergic component of PPI that is relevant for the action of atypical antipsychotics such as clozapine. Clozapine is a drug with relatively high affinity for the 5-HT7 receptor. PUBLICATIONS Hedlund, P.B., Huitrón-Reséndiz, S., Henriksen, S.J., Sutcliffe, J.G. 5-HT7 receptor inhibition and inactivation induce antidepressantlike behavior and sleep pattern. Biol. Psychiatry 58:831, 2005. Hedlund, P.B., Sutcliffe, J.G. 5-HT7 receptors as favorable pharmacological targets for drug discovery. In: The Serotonin Receptors: From Molecular Pharmacology to Human Therapeutics. Roth, B.L. (Ed.). Humana Press, Totowa, NJ, 2006, p. 517. Hedlund, P.B., von Euler, G. Z-analysis: a new approach to analyze stimulation curves with intrinsic basal stimulation. Biochem. Pharmacol. 70:170, 2005. 232 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE Lysophospholipid Signaling and Neural Aneuploidy J. Chun, S. Appadurai, B. Almeida, B. Anliker, E. Birgbauer, A. Dubin, S. Gardell, D. Herr, G. Kennedy, M. Kingsbury, C.W. Lee, M. Lu, M. McCreight, C. Paczkowski, S. Peterson, S. Rehen, R. Rivera, A.H. Yang, X.Q. Ye, Y. Yung, L. Zhu n the past year, we gained significant new insights into both lysophospholipid signaling and neural aneuploidy. First, we discovered that receptor-mediated lysophosphatidic acid (LPA) signaling, mediated by the cognate receptor known as LPA3, is essential for normal implantation of embryos in the uterine wall, a finding that may be relevant to the treatment of female infertility. Second, we acquired new data that indicate the potential function of genomically nonidentical brain cells in normal brain in humans. In further studies in mice, we found that aneuploid neurons can be integrated into the normal circuitry of the brain, indicating that the neurons are not simply dead or inert components but rather have the potential to modify properties of neural circuitry by virtue of their altered genomes. I F i g . 1 . Location of implantation sites in uteri at embryonic days 4.5 (E4.5) and 5.5 (E5.5). Bands indicate implantation sites. Mice lacking the gene for LPA3 have delayed implantation and at later times have reduced and abnormally spaced implantation (arrows). LY S O P H O S P H O L I P I D S Lysophospholipids such as LPA are simple phospholipids that act as extracellular signals that use cognate G protein–coupled receptors to bring about myriad effects. The 2 best studied lysophospholipids are LPA and sphingosine 1-phosphate (S1P). We continue to generate new lines of mice that lack the genes for single and multiple receptors and to characterize the mutant phenotypes. A null mutation in LPA 3 resulted in a reduced-fertility phenotype that was attributed to alterations in embryo implantation (Fig. 1). We are elucidating the downstream signaling effects of LPA3 in normal implantation. NORMAL NEURAL ANEUPLOIDY It is now clear that many cells in the brain have nonidentical genomes by virtue of being aneuploid, that is, the cells have gained and/or lost chromosomes. The initial research on aneuploidy was done in mice, raising the question of whether this phenomenon also existed in humans. Use of double labeling with point probes, which recognize a relatively discrete part of a chromosome, and “paints,” which recognize much of a given chromosome, allowed the unambiguous identification of aneuploid neurons and glia in normal human brain (Fig. 2). This finding led us to ask the additional question of whether such cells were capable of normal function. F i g . 2 . Nuclei isolated from the brains of different patients con- taining 1 (E), 2 (F), 3 (G), or 4 (H) copies of chromosome 21. The large, dark region indicates staining with DAPI (4′,6-diamidino-2phenylindole), whole-chromosome paint appears in light gray, and the chromosome 21 point probes are indicated by arrows. A complete overlap between the paint and the point probe occurs, as seen at higher magnification in the insets. Arrowheads indicate the numbers of chromosome 21 per cell. Scale bar, 5 µm. In mice, we found that indeed, aneuploid neurons can have distant connections and physiologic activities, suggesting that these genomically distinct cells can function in normal neural circuitry. Currently, we are determining the extent, forms, and roles of aneuploid neural cells in normal and diseased mammalian brains. MOLECULAR BIOLOGY 2006 PUBLICATIONS Barbeito, L., Chun, J., Binder, L.I., Neto, V.M., Perry, G., Scazzochio, C., Violini, G. The end of a Chilean institute. Science 308:792, 2005. Chun, J. Lysophospholipids in the nervous system. Prostaglandins Other Lipid Mediat. 77:46, 2005. Gon, Y., Wood, M.R., Kiosses, W.B., Jo, E., Sanna, M.G., Chun, J., Rosen, H. S1P3 receptor-induced reorganization of epithelial tight junctions compromises lung barrier integrity and is potentiated by TNF. Proc. Natl. Acad. Sci. U. S. A. 102:9270, 2005. Goparaju, S.K., Jolly, P.S., Watterson, K.R., Bektas, M., Alvarez, S., Sarkar, S., Mel, L., Ishii, I., Chun, J., Milstien, S., Spiegel, S. The S1P2 receptor negatively regulates platelet-derived growth factor-induced motility and proliferation. Mol. Cell. Biol. 25:4237, 2005. Kingsbury, M.A., Friedman, B., McConnell, M.J., Rehen, S.K., Yang, A.H., Kaushal, D., Chun, J. Aneuploid neurons are functionally active and integrated into brain circuitry. Proc. Natl. Acad. Sci. U. S. A. 102:6143, 2005. Li, H., Ye, X., Mahanivong, C., Bian, D., Chun, J., Huang, S. Signaling mechanisms responsible for lysophosphatidic acid-induced urokinase plasminogen activator expression in ovarian cancer cells. J. Biol. Chem. 280:10564, 2005. Rehen, S.K., Yung, Y.C., McCreight, M.P., Kaushal, D., Yang, A.H., Almeida, B.S.V., Kingsbury, M.A., Cabral, K.M.S., McConnell, M.J., Anliker, B., Fontanoz, M., Chun, J. Constitutional aneuploidy in the normal human brain. J. Neurosci. 25:2176, 2005. Simon, M.F., Daviaud, D., Pradere, J.P., Grès, S., Guigné, C., Wabitsch, M., Chun, J., Valet, P., Saulnier-Blache, J.S. Lysophosphatidic acid inhibits adipocyte differentiation via lysophosphatidic acid 1 receptor-dependent down-regulation of peroxisome proliferator-activated receptor γ2 J. Biol. Chem. 280:1456, 2005. Tölle, M., Levkau, B., Keul, P., Brinkmann, V., Giebing, G., Schönfelder, G., Schäfers, M., von Wnuck Lipinski, K., Jankowski, J., Jankowski, V., Chun, J., Zidek, W., Van der Giet, M. Immunomodulator FTY720 induces eNOS-dependent arterial vasodilation via the lysophospholipid receptor S1P3. Circ. Res. 96:913, 2005. Ye, X., Hama, K., Contos, J.J., Anliker, B., Inoue, A., Skinner, M.K., Suzuki, H., Amano, T., Kennedy, G., Arai, H., Aoki, J., Chun, J. LPA3 lysophosphatidic acid signalling in embryo implantation and spacing. Nature 435:104, 2005. Chemical Glycobiology J.C. Paulson, O. Blixt, L.K. Allin, H. Andersson-Sand, O.V. Bohorov, B.E. Collins, S. Han, J. Hoffman, D. Lebus, L. Liao, X. Liu, B. Ma, M. O’Reilly, N. Razi, P. Sobieszczuk, L. Stewart, H. Tateno, H. Tian, D. Vasiliu, Y. Zeng e investigate the roles of glycan-binding proteins that mediate cellular processes central to immunoregulation and human disease. We work at the interface of biology and chemistry to understand how the interaction of glycan-binding proteins with their ligands mediates cell-cell interactions, endocytosis, and cell signaling. Our multidisciplinary approach is complemented by a diverse group of chemists, biochemists, cell biologists, and molecular biologists. W BIOLOGICAL ROLES OF SIGLECS The siglecs are a family of 11 sialic acid–binding proteins that function as cell-signaling coreceptors. They are expressed on glial cells and on a variety of leukocytes that mediate acquired and innate immune THE SCRIPPS RESEARCH INSTITUTE 233 functions, including B cells, eosinophils, macrophages, dendritic cells, and natural killer cells. Siglecs are a subfamily of the immunoglobulin superfamily that have in common a unique N-terminal Ig domain that confers the ability to bind to sialic acid–containing carbohydrate groups (sialosides) of glycoproteins and glycolipids. The cytoplasmic domains of most siglecs contain tyrosine-based inhibitory motifs characteristic of accessory proteins that regulate transmembrane signaling and endocytosis of cell-surface receptor proteins. The diverse specificity for their sialoside ligands and their variable cytoplasmic regulatory elements provide siglecs with attributes for unique roles in the cell-surface biology of each cell that expresses them. The best understood siglec is CD22 (siglec-2), an accessory molecule of the B-cell receptor (BCR) complex that has both positive and negative effects on receptor signaling. The carbohydrate ligand recognized by CD22 is the sequence Siaα2-6Galbβ1-4GlcNAc found on glycoproteins of both B cells (cis ligands) and on cells that interact with B cells (e.g., T cells, trans ligands). Interactions of CD22 with cis or trans ligands regulate aspects of B-cell activation, proliferation, and development. We found that CD22 is predominately associated with clathrin-coated pits in resting B cells, whereas BCRs are minimally associated with clathrin domains. Mice deficient in the ligand for CD22 have greater colocalization of CD22 and the BCR in fused raft-clathrin domains than do mice that have the ligand, accounting for the immunosuppression in deficient mice. In wild-type mice, after antigen activation, the BCR is endocytosed via raft-clathrin domains, a logical site for the dampening of B-cell signaling by CD22. In resting cells, CD22 undergoes constitutive endocytosis, which can result in internalization of high-affinity ligands of CD22 (Fig. 1). We also study siglec-F (murine) and siglec-8 (human), which are predominately expressed on eosinophils and recognize the sialoside Siaα2-3(6-SO4=)Galβ14GlcNAc and are targets for modulating eosinophil activation. Another siglec being actively investigated is myelin-associated glycoprotein (siglec-4). This siglec is expressed on glial cells and recognizes the sialoside Siaα2-3Galβ1-3(Siaα2-6)GalNAc-R found on O-linked glycans of glycoproteins and glycolipids. Functionally, myelin-associated glycoprotein stabilizes interactions between glial cells and axons essential for normal organization of myelin and inhibits axonal regeneration, which is currently a target for pharmaceutical intervention to promote nerve regeneration. 234 MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE F i g . 1 . Relationship between microdomain localization of the BCR and CD22, a regulator of BCR signaling that binds glycan ligands. A major barrier to studying the ligand-binding properties of siglecs and their role in siglec biology is the difficulty in creating synthetic probes that compete with endogenous (cis) ligands. Even highly multivalent polymers containing the natural glycan sequence recognized by a siglec will not bind to cells unless cis ligands are first destroyed. However, we found that high-affinity analogs of the natural sialoside ligand of CD22 bind to native B cells and are carried into the cell by receptormediated endocytosis. Similar constructs with the ligand of siglec-F are also bound and endocytosed by eosinophils, but by a different endocytic mechanism. We have also developed potent inhibitors of myelin-associated glycoprotein that reverse its ability to block axon growth, and in collaborative studies with R. Schnaar, Johns Hopkins University, Baltimore, Maryland, we are investigating the potential of the inhibitors to promote nerve growth in vivo. With these successes, we have embarked on a major effort to identify high-affinity analogs of each siglec to produce ligand-based tools to investigate the biological roles of the siglecs in innate and adaptive immunity. S I A L O S I D E A N A L O G G LY C A N A R R AY S We have developed a robotically printed glycan array that displays sialoside analogs to assess the affinity of siglecs for unnatural substituents at the C-9 and C-5 positions of sialic acids. Even in the initial experiments with 65 acyl substituents at the C-9 position of sialic acid, the method was a powerful one for identifying substituents that increase the affinity of siglecs by 100-fold or more (Fig. 2). In collaboration with K.B. Sharpless, Department of Chemistry, we have created another 80 analogs by using by click chemistry to couple members of a library of alkynes to sialosides containing 9-azido-N-acetyl-neuraminic acid. Results from the array can be rapidly assimilated into the synthesis of high-affinity ligands and ligand-based probes of the F i g . 2 . Sialoside analog glycan microarray reveals high-affinity ligands for CD22. A, Sialoside ligands of CD22 with amino-terminated linkers are printed on N-hydroxyl succinimide (NHS)–activated glass slides, resulting in a covalent amide bond. B, The natural ligand (3) with various substituents (1, 2, 4, 6) and a nonligand control (5) are printed in 10 replicates at 10 two-fold diluted printing concentrations. Overlay with a fluorescence-labeled CD22-Ig chimera reveals the increased binding to various substituents compared with the natural ligand. corresponding siglec by using our flexible chemoenzymatic synthesis strategies. BIOENGINEERING OF CELL-SURFACE SIALOSIDES Sialic acids with substituents at the C-9 and C-5 positions are readily taken up by cells and incorporated into cell-surface glycans of glycoproteins and glycolipids by the natural glycosylation pathways. Taking advantage of this concept, we developed a novel method for in situ photoaffinity cross-linking of CD22 to its ligands on the same cell (cis) or adjacent cell (trans) by using a 9-aryl-azide-sialic acid. When exposed to ultraviolet light, CD22 is rapidly cross-linked to its cis ligands through protein-glycan covalent bonds (Fig. 3). The striking finding is that in addition to glycan structure, microdomain localization of CD22 strongly influences the glycoprotein ligands that CD22 interacts with. In fact, the predominant cis ligands of CD22 were glycans of neighboring CD22 molecules, showing homomultimeric complexes of CD22 mediated by CD22’s ligand-binding domain. Another application is to incorporate sialic acid analogs that increase or decrease the affinity of a siglec for its natural ligand to perturb the dynamics of interactions of the siglec with its cis ligand. For example, a 9-biphenylcarboxyl substituent (Fig. 3) increases the MOLECULAR BIOLOGY 2006 THE SCRIPPS RESEARCH INSTITUTE 235 influenza and related H1 avian influenza viruses and the more recent avian influenza virus (H5N1) to identify mutations required to switch specificity from avian receptors to human-type receptors. PUBLICATIONS Bochner, B.S., Alvarez, R.A., Mehta, P., Bovin, N.V., Blixt, O., White, J.R., Schnaar, R.L. Glycan array screening reveals a candidate ligand for siglec-8. J. Biol. Chem. 280:4307, 2005. Collins, B.E., Blixt, O., Han, S., Duong, B., Li, H., Nathan, J.K., Bovin, N., Paulson, J.C. High-affinity ligand probes of CD22 overcome the threshold set by cis ligands to allow for binding, endocytosis, and killing of B cells. J. Immunol. 177:2994, 2006. F i g . 3 . Bioengineering of cell-surface glycoproteins to carry substituents at the 9-position of N-acetyl–neuraminic acid that increase affinity (biphenylcarboxyl) or allow in situ photoaffinity cross-linking (9-aryl-azide) of CD22 to its ligands. affinity for CD22 by 100-fold, resulting in the strong CD22-mediated aggregation of B cells. These basic approaches will be of general value in elucidating the biology of other members of the siglec family. C O N S O R T I U M F O R F U N C T I O N A L G LY C O M I C S Members of our laboratory also staff 2 scientific cores for the Consortium for Functional Glycomics, organized to elucidate the mechanisms by which glycan-binding proteins mediate cell communication (http://www.functionalglycomics.org/). In the past year, scientists in the Mouse Transgenics Core, led by Peter Sobieszczuk, created 6 novel mouse strains from C57Bl/6 embryonic stem cells that are deficient in genes for key glycan-binding proteins that affect immune function. Scientists in the Glycan Array Synthesis Core, led by Ola Blixt, have produced a library of synthetic glycans by chemoenzymatic synthesis for use in numerous applications. In addition, scientists in the Scripps DNA Microarray Core, led by Steve Head, designed and conduced investigator-initiated analysis with a custom-based microarray with genes of relevance for the consortium. A major achievement by staff in the Glycan Array Synthesis Core is the development of the world largest glycan microarray, which currently has more than 300 unique structures, mostly synthetic glycans produced by chemoenzymatic synthesis. Now produced in collaboration with the DNA Microarray Core, the microarray is widely used by investigators around the world to assess the specificity of glycan-binding proteins that mediate a broad scope of biological interactions. In an exemplary collaboration with I.A. Wilson and J. Stevens, Department of Molecular Biology, this array was used to investigate the specificity of the 1918 pandemic Collins, B.E., Smith, B.A., Bengtson, P., Paulson, J.C. Ablation of CD22 in liganddeficient mice restores B cell receptor signaling. Nat. Immunol. 7:199, 2006. Comelli, E.M., Head, S.R., Gilmartin, T., Whisenant, T., Haslam, S.M., North, S.J., Wong, N.K., Kudo, T., Narimatsu, H., Esko, J.D., Drickamer, K., Dell, A., Paulson, J.C. A focused microarray approach to functional glycomics: transcriptional regulation of the glycome. Glycobiology 16:117, 2006. Comelli, E.M., Sutton-Smtih, M., Yan, Q., Amado, M., Panico, M., Gilmartin, T., Whisenant, T., Lanigan, C.M., Head, S.R., Goldberg, D., Morris, H., Dell, A., Paulson, J.C. Activation of murine CD4+ and CD8+ T lymphocytes leads to dramatic remodeling of N-linked glycans. J. Immunol. 177:2431, 2006. Han, S., Collins, B.E., Paulson, J.C. Synthesis of 9-substituted sialic acids as probes for CD22-ligand interactions on B. Oxford University Press, New York, in press. ACS Symposium Series. Leppanen, A., Stowell, S., Blixt, O., Cummings, R.D. Dimeric galectin-1 binds with high affinity to α2,3-sialylated and non-sialylated terminal N-acetyllactosamine units on surface-bound extended glycans. J. Biol. Chem. 280:5549, 2005. Paulson, J.C., Blixt, O., Collins, B.E. Sweet spots in functional glycomics. Nat. Chem. Biol. 2:238, 2006. Raman, R., Raguram, S., Venkataraman, G., Paulson, J.C., Sasisekharan, R. Glycomics: an integrated systems approach to structure-function relationships of glycans. Nat. Methods 2:817, 2005. Singh, T., Wu, J.H., Peumans, W.J., Rouge, P., Van Damme, E.J., Alvarez, R.A., Blixt, O., Wu, A.M. Carbohydrate specificity of an insecticidal lectin isolated from the leaves of Glechoma hederacea (ground ivy) towards mammalian glycoconjugates. Biochem. J. 393:331, 2005. Stevens, J., Blixt, O., Glaser, L., Taubenberger, J.K., Palese, P., Paulson, J.C., Wilson, I.A. Glycan microarray analysis of the hemagglutinins from modern and pandemic influenza viruses reveals different receptor specificities. J. Mol. Biol. 355:1143, 2006. Stevens, J., Blixt, O., Paulson, J.C., Wilson, I.A. Glycan microarray technologies: tools to survey host specificity of influenza viruses. Nat. Rev. Microbiol. 4:857, 2006. Stevens, J., Blixt, O., Tumpey, T.M., Taubenberger, J.K., Paulson, J.C., Wilson, I.A. Structure and receptor specificity of the hemagglutinin from an H5N1 influenza virus. Science 312:404, 2006. Taniguchi, N., Nakamura, K., Narimatsu, H., von der Lieth, C.W., Paulson, J.C. Human Disease Glycomics/Proteome Initiative workshop and the 4th HUPO Annual Congress. Proteomics 6:12, 2006. Tateno, H., Crocker, P.R., Paulson, J.C. Mouse siglec-F and human siglec-8 are functionally convergent paralogs that are selectively expressed on eosinophils and recognize 6′-sulfo-sialyl Lewis X as a preferred glycan ligand. Glycobiology 15:1125, 2005. van Vliet, S.J., van Liempt, E., Saeland, E., Aarnoudse, C.A., Appelmelk, B., Irimura, T., Geijtenbeek, T.B., Blixt, O., Alvarez, R., van Die, I., van Kooyk, Y. Carbohydrate profiling reveals a distinctive role for the C-type lectin MGL in the recognition of helminth parasites and tumor antigens by dendritic cells. Int. Immunol. 17:661, 2005. 236 MOLECULAR BIOLOGY 2006 Vasiliu, D., Razi, N., Zhang, Y., Jacobsen, N., Allin, K., Liu, X., Hoffmann, J., Bohorov, O., Blixt, O. Large-scale chemoenzymatic synthesis of blood group and tumor-associated poly-N-acetyllactosamine antigens. Carbohydr. Res. 3451:1447, 2006. Westerlind, U., Hagback, P., Tidback, B., Wiik, L., Blixt, O., Razi, N., Norberg, T. Synthesis of deoxy and acylamino derivatives of lactose and use of these for probing the active site of Neisseria meningitidis N-acetylglucosaminyltransferase. Carbohydr. Res. 340:221, 2005. THE SCRIPPS RESEARCH INSTITUTE