Chemistry Coiled and extended sodium dodecyl sulfate conformations in the water-soluble cavitand. Nuclear magnetic resonance evidence supports the "strained" structure in which 8 carbon atoms adopt a coiled conformation to maximize interaction with the host. Artwork was done by Michael P. Schramm, Ph.D., The Skaggs Institute for Chemical Biology. Julius Rebek, Jr., Ph.D. Professor, Department of Chemistry Director, The Skaggs Institute for Chemical Biology CHEMISTRY DEPAR TMENT OF CHEMISTRY S TA F F K.C. Nicolaou, Ph.D.* Professor and Chairman Aline W. and L.S. Skaggs Professor of Chemical Biology Darlene Shiley Chair in Chemistry Phil Baran, Ph.D. Associate Professor Dale L. Boger, Ph.D.* Richard and Alice Cramer Professor of Chemistry Tobin J. Dickerson, Ph.D. Assistant Professor Albert Eschenmoser, Ph.D.* Professor Sheng Ding, Ph.D. Assistant Professor M.G. Finn, Ph.D.* Associate Professor Valery Fokin, Ph.D. Associate Professor M. Reza Ghadiri, Ph.D.* Professor William A. Greenberg, Ph.D. Assistant Professor Inkyu Hwang, Ph.D. Assistant Professor Hayato Ishikawa, Ph.D. Assistant Professor Kim D. Janda, Ph.D.** Professor Ely R. Callaway, Jr., Chair in Chemistry Director, The Worm Institute for Research and Medicine Jeffery W. Kelly, Ph.D.* Lita Annenberg Hazen Professor of Chemistry Dean, Graduate and Postgraduate Studies 2006 Ramanarayanan Krishnamurthy, Ph.D. Associate Professor Richard A. Lerner, M.D.*** President, The Scripps Research Institute Lita Annenberg Hazen Professor of Immunochemistry Cecil H. and Ida M. Green Chair in Chemistry Masayuki Matsushita, Ph.D.**** Chugai Pharmaceutical Co., LTD. Tokyo, Japan Michael Meijler, Ph.D. Assistant Professor Jorge J. Nieva, M.D. Assistant Professor Evan Powers, Ph.D. Assistant Professor Julius Rebek, Jr., Ph.D.* Professor Director, The Skaggs Institute for Chemical Biology Ed Roberts, Ph.D. Professor THE SCRIPPS RESEARCH INSTITUTE Andrew Bin Zhou, Ph.D. Assistant Professor S TA F F S C I E N T I S T S Byeong D. Song, Ph.D. Lubica Supekova, Ph.D. Peter G. Schultz, Ph.D.* Professor Scripps Family Chair K. Barry Sharpless, Ph.D.* Professor W.M. Keck Professor of Chemistry Vaughn Smider, Ph.D. Assistant Professor Anita Wentworth, Ph.D. Assistant Professor Paul Wentworth, Jr., Ph.D. Professor Chi-Huey Wong, Ph.D.* Ernest W. Hahn Professor and Chair in Chemistry Stellios Arseniyadis, Ph.D.**** Ecole Supérieure de Physique et de Chimie Industrielle Paris, France Gonen Ashkenasy, Ph.D.**** Ben-Gurion University of the Negev Beer-Sheeva, Israel Wen Xiong, Ph.D. I N S T R U M E N TAT I O N / SERVICE FACILITIES Raj K. Chadha, Ph.D. Director, X-Ray Crystallography Facility Dee H. Huang, Ph.D. Director, Nuclear Magnetic Resonance Facility Gary E. Siuzdak, Ph.D. Director, Mass Spectrometry Facility SENIOR RESEARCH A S S O C I AT E S Ashraf Brik, Ph.D. Yanping Chen, Ph.D. Floyd E. Romesberg, Ph.D. Associate Professor 63 Gunnar Kaufmann, Ph.D. Nurit Ashkenasy, Ph.D.**** Ben-Gurion University of the Negev Beer-Sheva, Israel Masato Atsumi, Ph.D.**** Kyocera JMM Osaka, Japan Elizabeth Barrett, Ph.D. Christoph Behrens, Ph.D.**** University of Gottingen Gottingen, Germany Clay Bennett, Ph.D. Jan Bieschke, Ph.D.**** Max Delbrück Center for Molecular Medicine Berlin, Germany Babu Boga, Ph.D.**** Schering Plough Redminster, New Jersey Anthony Boitano, Ph.D. R E S E A R C H A S S O C I AT E S Ramzey Abujarour, Ph.D.**** Genomics Institute of the Novartis Research Foundation San Diego, California Venkataiah Bollu, Ph.D. Brant Boren, Ph.D. Dariush Ajami, Ph.D. Daryl Bosco, Ph.D.**** Serono, Inc. Boston, Massachusetts Lital Alfonta, Ph.D. Andrew Brogan, Ph.D. Xaiver Alvarez-Mico, Ph.D. Adrian Brunkhorst, Ph.D.***** Rahesh Ambasudhan, Ph.D. Paul Bulger, Ph.D. Narendra B. Ambhaikar, Ph.D.**** Vertex Pharmaceuticals San Diego, California Kevin Bunker, Ph.D. Antonio Burtoloso, Ph.D. 64 CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE Steven Johnson, Ph.D. Nil Emre, Ph.D. Sayam Sen Gupta, Ph.D.**** Technical University Munich Munich, Germany Alexandre Carella, Ph.D. Lisa Eubanks, Ph.D. Richard Guy, Ph.D. Michael Cassidy, Ph.D.**** Bristol-Myers Squibb New Brunswick, New Jersey Cyrine Ezzili, Ph.D. Clemens Haas, Ph.D.***** Michael Kelso, Ph.D.**** University of Wollongong Wollongong, Australia Akiyuki Hamasaki, Ph.D. Sukbok Chang, Ph.D. Raffaella Faraoni, Ph.D.**** Ambit Biosciences San Diego, California Shuo Chen, Ph.D. Simon Ficht, Ph.D. Yanping Chen, Ph.D.***** Laura Flatauer, Ph.D.**** Genomics Institute of the Novartis Research Foundation San Diego, California Mark Bushey, Ph.D. Jan Elsner, Ph.D. Sara Butterfield, Ph.D. Jodie Chin, Ph.D. Charles Cho, Ph.D. Wooseok Han, Ph.D.**** Chiron Emeryville, California Frank Hauke, Ph.D.**** University of Erlangen Erlangen, Germany Antonella Converso, Ph.D.**** Merck & Co., Inc. West Point, Pennsylvania Jeromy Cottell, Ph.D. James Crawford, Ph.D. Matthew Cremeens, Ph.D. Jesse Dambacher, Ph.D. Sandra De Lamo Marin, Ph.D. Ross Denton, Ph.D. Caroline Desponts, Ph.D. David Diaz-Diaz, Ph.D.**** Universidad Autónoma de Madrid Madrid, Spain F. Scott Kimball, Ph.D. Ravinder Reddy Kondreddi, Ph.D. Larisa Krasnova, Ph.D. Ted Foss, Ph.D. Kevin Cole, Ph.D. Gyungyoun Kim, Ph.D.**** SK Biopharmaceuticals Fairfield, New Jersey Jason Hein, Ph.D. So-Hye Cho, Ph.D. Scott Cockroft, Ph.D. Jaroslaw Kalisiak, Ph.D. Jane Kuzelka, Ph.D. Joseph Rodolph Fotsing, Ph.D. Mark Hixon, Ph.D.**** Takeda San Diego, California Rebecca Fraser, Ph.D.**** Novartis Pharma AG Horsham, England Jiyong Hong, Ph.D.**** Duke University Durham, North Carolina Graeme Freestone, Ph.D. Jim Fuchs, Ph.D. Sukwon Hong, Ph.D.**** University of Florida Gainsville, Florida Amelia Fuller, Ph.D. Zhangyong Hong, Ph.D. Jianmin Gao, Ph.D. Richard Hooley, Ph.D. Muyun Gao, Ph.D. Ola Ghoneim, Ph.D. Daniel Horne, Ph.D.**** Amgen Thousand Oaks, California Nathan Gianneschi, Ph.D. Tsui-Ling Hsu, Ph.D. Romelo Gibe, Ph.D. Institute of Chemical and Engineering Sciences Jurong Island, Singapore, China Zheng-Zheng Huang, Ph.D. Andreas Lanver, Ph.D. Brian Lawhorn, Ph.D. Jinq-Chyi Lee, Ph.D. Jongkook Lee, Ph.D. JongSeok Lee, Ph.D. Ki-Bum Lee, Ph.D. Kooyeon Lee, Ph.D. Sang Hyup Lee, Ph.D.**** LG Life Sciences Daejeon, South Korea Sejin Lee, Ph.D. Lucas Leman, Ph.D. Alexandre Lemire, Ph.D. Amy Hurshman, Ph.D.**** Joint Science Department of the Claremont Colleges Claremont, California Edward Lemke, Ph.D. Achim Lenzin, Ph.D. Christine Dierks, Ph.D.**** Genomics Institute of the Novartis Research Foundation San Diego, California Christina Gil-Lamaignere, Ph.D. Der-Ren Hwang, Ph.D. Hongming Li, Ph.D. Neill Gingles, Ph.D. Giltae Hwang, Ph.D. Ke Li, Ph.D. David Edmonds, Ph.D. Naran Gombosuren, Ph.D. Tetsuo Iwasawa, Ph.D. Pi-Hui Liang, Ph.D. Greg Elliott, Ph.D.**** Moore Cancer Center La Jolla, California Rajesh K. Grover, Ph.D. Michael Jahnz, Ph.D. Jan Grunewald, Ph.D. Wei Jin, Ph.D. Jiayu Liao, Ph.D.**** University of California Riverside, California CHEMISTRY Yeon-Hee Lim, Ph.D. 2006 Troy Lister, Ph.D. Robert Milburn, Ph.D.**** Amgen Thousand Oaks, California Chris Liu, Ph.D. Kyung-Hoon Min, Ph.D.***** THE SCRIPPS RESEARCH INSTITUTE Charles Papageorgiou, Ph.D.**** Amgen Cambridge, Massachusetts 65 F. Anthony Romero, Ph.D. Youngha Ryu, Ph.D. Riccardo Salvio, Ph.D. Doron Pappo, Ph.D. Haitian Liu, Ph.D.**** Senomyx, Inc. La Jolla, California Jun Liu, Ph.D.**** Genomics Institute of the Novartis Research Foundation San Diego, California Christos A. Mitsos, Ph.D.**** Athens, Greece Junguk Park, Ph.D. Gopi Kumar Mittapalli, Ph.D. Laxman Pasunoori, Ph.D. Lionel Moisan, Ph.D. Richard Payne, Ph.D. Ana Montero, Ph.D. Murali Peram Surakattula, Ph.D. Lei Liu, Ph.D. Miguel Morales, Ph.D. Wenshe Liu, Ph.D. Tingwei Mu, Ph.D. Yi Liu, Ph.D.**** Lawrence Berkeley National Laboratory Berkeley, California Mridul Mukherji, Ph.D. Ying (Cindy) Liu, Ph.D. Dimitrios Lizos, Ph.D.**** Novartis Pharmaceuticals Basil, Switzerland Jon Loren, Ph.D.**** Genomics Institute of the Novartis Research Foundation San Diego, California Hongzheng (Eric) Ma, Ph.D. Roman Manetsch, Ph.D.**** University of South Florida Tampa, Florida Enrique Mann, Ph.D. Felix Marr, Ph.D.***** Carol Lamenca Martinez, Ph.D.**** Berlin, Germany Roshan Perera, Ph.D. Goran Petrovic, Ph.D. Andrew Myles, Ph.D.**** University of Alberta Edmonton, Canada Jared Piper, Ph.D.**** Eli Lilly Indianapolis, Indiana Suresh Pitram, Ph.D. Kenichiro Nagai, Ph.D. Tülay Polat, Ph.D. Yuya Nakai, Ph.D. Damien Poliet, Ph.D. Joonwoo Nam, Ph.D. Anu Sawkar, Ph.D.**** Ropes & Gray New York, New York Patrick Schanen, Ph.D.**** ETH Zürich, Laboratory of Organic Chemistry Zurich, Switzerland Stefan Schiller, Ph.D. Daniel Schlawe, Ph.D.**** Syncom BV Groningen, the Netherlands Michael Schramm, Ph.D. Laura Segatori, Ph.D. Mary Sever, Ph.D. Alex Shaginian, Ph.D. Guido Pontremoli, Ph.D. David Shaw, Ph.D. Sridhar Narayan, Ph.D.**** Eisai Research Institute Wilmington, Massachusetts Mariceli Puga, Ph.D. Daniel Nicoletti, Ph.D. Sreenivas Punna, Ph.D.**** ChemoCentryx, Inc. Mountain View, California Junhwa Shin, Ph.D.**** Korea Atomic Energy Research Institute Seoul, Korea Alain Noncovich, Ph.D.**** Senomyx Inc. La Jolla, California Yasuo Norikane, Ph.D.**** Nanotechnology Research Institute, AIST Tsukuba, Japan Mehdi Numa, Ph.D.**** Vertex Pharmaceuticals Incorporated San Diego, California Shigeo Matsuda, Ph.D. Severin Odermatt, Ph.D. Klaus-Dieter Michael Maue, Ph.D. Barun Okram, Ph.D. Laura McAllister, Ph.D. Peter Orahovats, Ph.D. Kathleen McKenzie, Ph.D. Yazmin Osornio, Ph.D. Sebastian Steiniger, Ph.D. Daniela Radu, Ph.D. Nicole Rahe, Ph.D.**** Tesa AG Hamburg, Germany Shai Rahimipour, Ph.D.**** Bar-Ilan University Ramat-Gan, Isreal Shula Stokols, Ph.D.**** W. L. Gore & Associates Flagstaff, Arizona Ji Young Suk, Ph.D.**** Merck Research Laboratories Boston, Massachusetts Daniel Summerer, Ph.D. Praveen Rao, Ph.D. Takahiro Suzuki, Ph.D. Dalit Rechavi-Robinson, Ph.D.**** University of Geneva Geneva, Switzerland Stefanie Roeper, Ph.D.***** Vertex Pharmaceuticals Cambridge, Massachusetts Leo Takaoka, Ph.D. Eric Tippmann, Ph.D. Jonathan Tripp, Ph.D. Meng-Lin Tsao, Ph.D. 66 CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE Craig Turner, Ph.D. Heiko Wurdak, Ph.D. V I S I T I N G I N V E S T I G AT O R S Jim Turner, Ph.D.**** Fish & Richardson P.C. San Diego, California Jian Xie, Ph.D. Mohammad Al-Sayah, Ph.D. American University of Sharjah Sharjah, United Arab Emirates Andrew Udit, Ph.D. Yue Xu, Ph.D. Ryu Yamasaki, Ph.D. Taiki Umezawa, Ph.D. Yasuyuki Ura, Ph.D. Shuyan Yao, Ph.D.**** Sangamo BioScience, Inc. Richmond, California Kenji Usui, Ph.D. Juraj Velcicky, Ph.D.**** Novartis Basel, Switzerland Alessandro Volonterio, Ph.D.**** Politencino di Milano Milano, Italy Robert Yeh, Ph.D. **** Siemens Torrance, California Yong Sik Yoo, Ph.D.**** Cheil Industries, Inc. Seoul, Korea Zhanqian Yu, Ph.D. Hong Wang, Ph.D. Jiangyun Wang, Ph.D. Weidong Wang, Ph.D. Mark Zak, Ph.D.**** Genentech San Francisco, California Xiaolong Wang, Ph.D. Felix Zelder, Ph.D.**** Timo Weide, Ph.D. Huaqiang Zeng, Ph.D.**** National University of Singapore Singapore Lisa Whalen, Ph.D.**** University of New Mexico Albuquerque, New Mexico Matthew Whiting, Ph.D. Aarron Willingham, Ph.D.**** Affymetrix Santa Clara, California Albert Willis, Ph.D. Chung-Yi Wu, Ph.D.**** Academia Sinica The Genomics Research Institute Taipei, Taiwan Qisheng Zhang, Ph.D. Yingchao Zhang, Ph.D. Yuanxiang Zhao, Ph.D.**** Cellular Dynamics International Inc. Madison, Wisconsin Heyue Henry Zhou, Ph.D. Min Zhou, Ph.D. Douglass Wu, Ph.D. Optimer Pharmaceuticals San Diego, California Xiuwen Zhu, Ph.D. Margarita Wuchrer, Ph.D.**** Merck KgaA Frankfurt, Germany Caterina Zoni, Ph.D. Masaaki Sawa, Ph.D. **** Dainippon Pharmaceuticals Co., Ltd Osaka, Japan Masakazu Sugiyama, Ph.D. Ajinomoto Co., Inc. Kawasaki-Shi, Japan Luda Bazhenova, Ph.D.**** University of California San Diego, California Shin-Ichi Takanashi, Ph.D.**** Sheila Fleming, Ph.D.**** University of California Los Angeles, California Erich Uffelman, Ph.D.**** Washington and Lee University Lexington, Virginia Masakazu Fujio, Ph.D. Mitsubishi Pharma Corporation Yokohama, Japan Luigi Gomez Paloma, Ph.D.**** University di Salerno Fisciano, Italy Andrew S. Grant, Ph.D.**** Mount Allison University Sackville, New Brunswick, Canada Yoshiyuki Hari, Ph.D. Nagoya City University Nagoya, Japan Akira Ino, Ph.D. Shionogi & Co., LTD. Osaka, Japan Lisa Landino, Ph.D. College of William and Mary Williamsburg, Virginia Jason Moss, Ph.D.**** PDL BioPharma, Inc. Fremont, California Poul Nielsen, Ph.D. University of Southern Denmark Odense, Denmark Joerg Zimmermann, Ph.D. Manuela Rodriguez, Ph.D.**** University di Salerno Fisciano, Italy Makoto Yamashita, Ph.D.**** Takeda Chemical Industries, Ltd. Osaka, Japan S C I E N T I F I C A S S O C I AT E S Jon Ashley Gina Dendle Suresh Mahajan, Ph.D. * Joint appointment in The Skaggs Institute for Chemical Biology ** Joint appointments in the Skaggs Institute for Chemical Biology and the Department of Immunology *** Joint appointments in The Skaggs Institute for Chemical Biology and the Department of Molecular Biology **** Appointment completed; new location shown ***** Appointment completed CHEMISTRY 2006 K.C. Nicolaou, Ph.D. Chairman's Overview s the "central science," chemistry stands between biology and medicine and between physics and materials science and provides the crucial bridge for drug discovery and development. But chemistry has a much more profound and useful role in science and society. It is the discipline that continually creates the myriad of new materials that we all encounter in our everyday lives: pharmaceuticals, high-tech materials, polymers and plastics, insecticides and pesticides, fabrics and cosmetics, fertilizers, and vitamins—basically everything we can touch, feel, and smell. Chemists at Scripps Research focus on chemical synthesis and chemical biology, the areas most relevant to biomedical research and materials science. The members of our faculty are distinguished teacher-scholars who maintain highly visible and independent research programs in areas as diverse as biological and chemical catalysis, synthesis of natural products, combinatorial chemistry, molecular design, supramolecular chemistry, chemical evolution, materials science, and chemical biology. The chemistry graduate program attracts some of the best-qualified candidates from both the United States and abroad. Our major research facilities, under the direction of Dee H. Huang (nuclear magnetic resonance), Gary Siuzdak (mass spectrometry), and Raj A THE SCRIPPS RESEARCH INSTITUTE 67 Chadha (x-ray crystallography), are second to none and continue to provide crucial support to our research programs. In addition, the Mabel and Arnold Beckman Center for the Chemical Sciences constantly receives high praise from visitors from around the world for its architectural design and operational aspects, both highly conducive to research. Research in the Department of Chemistry goes on unabated, establishing international visibility and attracting attention as evidenced by numerous lecture invitations, visits by outside scholars, and headline news in the media. As of 2005, the Institute for Scientific Information ranked 3 members of our department as highly cited researchers (in the top 100 worldwide); 2 of the 3 are among the top 51 positions. Richard Lerner and his group continue to make advances in catalytic antibodies, with new antibodies that catalyze important synthetic and biological reactions and novel applications in chemical synthesis. The group’s research has recently expanded to include the fundamental chemistry of the polyoxygen species. Barry Sharpless and his group continue endeavors to discover and develop better catalysts for organic synthesis and to construct, through innovative chemistry and biology, libraries of novel compounds for biological screening. Scientists in Albert Eschenmoser’s La Jolla-based group advance their experimental studies on the chemical etiology of nucleic acid structure by investigating nucleic acid alternatives that have novel backbones and recognition elements unrelated to the canonical phophodiesterbased oligonucleotide systems. Members of my own group continue explorations of chemical synthesis and chemical biology, focusing on the total synthesis of new anticancer agents, antibiotics, marine-derived neurotoxins, antimalarial compounds, antifeedant agents, and other biologically active natural and designed molecules. The members of Julius Rebek’s group devise biomimetic receptors for studies in molecular recognition. These include molecules that bind neurotransmitters and membrane components. Larger host receptors can surround 3 or more molecular guests and act as chambers where the chemical reactions of the guests are accelerated. The group synthesizes small molecules that act as protein helix mimetics for pharmaceutical applications. Peter Schultz and his group have continued to expand the number of genetically encoded amino acids to include fluorescent, photocaged, metal binding, thioester, sulfated, and long-chain alkane side chains. They have 68 CHEMISTRY 2006 also adapted this technology to mammalian cells and are applying it to a number of basic and applied problems in cell biology. In addition, they have used cell-based screens to identify small molecules that selectively differentiate and expand embryonic and adult stem cells and reprogram lineage-committed cells, as well as novel genes that control cell cycle, cell migration, and developmental pathways. Chi-Huey Wong and his group further advance the fields of chemoenzymatic organic synthesis, chemical glycobiology, and the development of enzyme inhibitors. A new strategy for the synthesis of glycoproteins has been developed. The programmable 1-pot synthesis of oligosaccharides developed by this group has been further used in the assembly of glycoarrays for study of saccharides that bind to proteins. This group also developed new probes to study glycosyltransferases and their role in cancer. Members of Dale Boger’s group continue their work on chemical synthesis; combinatorial chemistry; heterocycle synthesis; anticancer agents such as vinblastine, fostriecin, and yatakemycin; and antibiotics such as vancomycin, teicoplanin, and ramoplanin. Scientists in Kim Janda’s laboratory are focusing on the impact of organic chemistry in specific biological systems. Their targeted programs span a wide range of interests from immunopharmacotherapy to biological and chemical warfare agents to filarial infections, such as "river blindness," to quorum sensing in bacteria. Their recent achievements include the discovery of a secondary nicotine metabolite that alters retinoid homeostasis, a critical component of vision and growth; small molecules that "superactivate" botulinum neurotoxin; and a virusbased system that can degrade cocaine in the central nervous system. Reza Ghadiri and his group are making significant contributions in the design and study of a new generation of antimicrobial agents, based on self-assembling peptide nanotube architecture, to combat multidrugresistant infections. In addition, they continue to make novel contributions in several ongoing basic research endeavors, such as biosensor designs, molecular computation, design of self-reproducing systems, understanding the origins of life, and design of emergent chemical systems. M.G. Finn and his group have pioneered the use of virus particles as chemical reagents and building blocks for nanochemical structures. This effort is directed toward the development of new diagnostics for disease and catalysts for organic reactions. Members of Dr. Finn’s labo- THE SCRIPPS RESEARCH INSTITUTE ratory also develop and investigate new organic and organometallic reactions and use these processes to synthesize biologically active compounds. Jeffery Kelly and his group are exploring the interface between chemistry, biology, and medicine. Their projects aim to understand the physical and biological basis of protein folding, and the misfolding and aggregation processes leading to age-onset neurodegenerative diseases. Comprehension of the latter processes is used to develop new small-molecule therapeutic strategies for a variety of neurodegenerative diseases. Anita Wentworth and her group are investigating the chemical basis of complex disease states and are synthesizing peptide and small molecule–based therapeutics. Their research is focused on disease states that have a prominent inflammatory and reactive oxygen-species chemical component, such as atherosclerosis, Alzheimer's disease, and other diseases of aging. Researchers in Floyd Romesberg’s laboratory are using diverse techniques ranging from bioorganic and biophysical chemistry to bacterial and yeast genetics to understand and manipulate the process of evolution. Major efforts include designing unnatural base pairs and the directed evolution of DNA polymerases to efficiently synthesize unnatural DNA containing the base pairs; using spectroscopy to understand biological function and how it evolves; and understanding how induced and adaptive mutations contribute to evolution in eukaryotic and prokaryotic cells. Dr. Baran and his group have recently developed extremely concise chemical solutions to the synthetic challenges posed by numerous marine natural product families, including sceptrin, ageliferin, chartelline, haouamine, welwitindolinones, and the stephacidins. These syntheses are characterized by striking brevity, new biosynthetic postulates, the invention of a new methodology, and a minimum use or complete absence of protecting groups and superfluous oxidation-state manipulations. The Frontiers in Chemistry Lecturers (17th Annual Symposium) for the 2005–2006 academic year were Richard Lerner, Scripps Research; Peter Vollhardt, University of California, Berkeley; Dieter Enders, Institute of Organic Chemistry, RWTH Aachen, Germany; and K.C. Nicolaou, Scripps Research. Thomas Scanlan (University of California, San Francisco) also visited Scripps this year as the Novartis Lecturer in Organic Chemistry, 2005. CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE 69 INVESTIGATORS’ R EPORTS Practical Total Synthesis of Natural Products P.S. Baran, N.Z. Burns, M.P. DeMartino, C.A. Guerrero, B.D. Hafensteiner, P.J. Krawczuk, K. Li, D.W. Lin, T.J. Maimone, M.K.-D. Maue, S. Nguyen, D.P. O’Malley, J.M. Richter, R.A. Shenvi, B. Whitefield rom penicillin to paclitaxel (Taxol), natural products have an unparalleled track record in the betterment of human health. In fact, 9 of the top 20 best-selling drugs were either inspired by or derived from natural products. Even the best-selling drug of all time, atorvastatin (Lipitor), was based on a natural product lead. Total synthesis, the art and science of recreating these entities in the laboratory, invariably leads to fundamental discoveries in chemistry, biology, and medicine. We focus on solving interesting challenges in the total synthesis of natural products and on bridging gaps in synthetic capabilities by inventing new reactions. Through judicious target selection and creative retrosynthetic analyses, total synthesis becomes an engine for discovery that drives the field of organic chemistry to new levels of sophistication and practicality. Synthetic organic chemistry requires tremendous ingenuity, artistic taste, experimental acumen, persistence, and character. Not surprisingly, drug development relies on the expertise of researchers who have these characteristics. Although we focus entirely on educating students in fundamental chemistry, we also collaborate with expert biologists to explore the medicinal potential of newly synthesized natural products and the products’ analogs. Recently completed total syntheses (Fig. 1) include the anticancer agents stephacidins A and B and avrainvillamide; the antibacterial agents sceptrin and ageliferin; members of the bioactive fischerindole, hapalindole, and welwitindolinone indole alkaloid family; and the anticancer agent haouamine A. Current natural product targets (Fig. 2) include chartelline C, axinellamine, strictamine, and sarcodonin. F F i g . 1 . Recently completed total syntheses. Baran, P.S., Li, K., O’Malley, D.P., Mitsos, C. Short, enantioselective total synthesis of sceptrin and ageliferin by programmed oxaquadricyclane fragmentation. Angew. Chem. Int. Ed. 45:249, 2006. PUBLICATIONS Baran, P.S., Burns, N.Z. Total synthesis of (±)-haouamine A. J. Am. Chem. Soc. 128:3908, 2006. Baran, P.S., Richter, J.M. Enantioselective total syntheses of welwitindolinone A and fischerindoles I and G. J. Am. Chem. Soc. 127:15394, 2005. Baran, P.S., Hafensteiner, B.D., Ambhaikar, N.B., Guerrero, C.A., Gallagher, J.D. Enantioselective total synthesis of avrainvillamide and the stephacidins. J. Am. Chem. Soc. 128:8678, 2006. Northrop, B.H., O’Malley, D.P., Zografos, A.L., Baran, P.S., Houk, K.N. The mechanism of the vinylcyclobutane rearrangement of sceptrin to ageliferin and nagelamide E. Angew. Chem. Int. Ed. 45:4126, 2006. 70 CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE SYNTHETIC METHODS Central to much of our work are investigations to develop and apply the hetero Diels-Alder reaction, including the use of heterocyclic and acyclic azadienes (Fig. 1), F i g . 1 . N-Sulfonyl-1-aza-1,3-butadiene Diels-Alder reaction. the thermal reactions of cyclopropenone ketals, intermolecular and intramolecular acyl radical–alkene addition reactions, medium- and large-ring cyclization technology, and solution-phase combinatorial chemistry. In each instance, the development of the methods represents the investigation of chemistry projected as a key element in the synthesis of a natural or designed agent. T O TA L S Y N T H E S I S O F N AT U R A L P R O D U C T S F i g . 2 . Ongoing natural product total syntheses. Synthetic and Bioorganic Chemistry D.L. Boger, S.B. Boga, K. Bunker, R. Clark, D. Colby, J. Cottell, B. Crowley, J. DeMartino, G. Elliott, J. Elsner, C. Ezzili, J. Fuchs, J. Garfunkle, A. Hamasaki, W. Han, N. Haq, S. Hong, D. Horne, I. Hwang, H. Ishikawa, W. Jin, D. Kato, D. Kastrinsky, M. Kelso, G. Kim, F.S. Kimball, B. Lawhorn, S. Lee, C. Liu, K. MacMillan, J. Nam, P. Patel, A. Romero, M. Schnermann, A. Shaginian, C. Slown, L. Takaoka, H. Tao, M. Tichenor, J. Trzupek, J. Velcicky, L. Whiby, Y. Zhang he research interests of our group include the total synthesis of natural products, development of new synthetic methods, heterocyclic chemistry, bioorganic and medicinal chemistry, the study of DNA-agent interactions, and the chemistry of antitumor antibiotics. We place a special emphasis on investigations to define the structure-function relationships of natural or designed organic agents. T Efforts are under way on the total synthesis of a number of natural products that constitute agents in which we have a specific interest. Representative agents currently under study include (+)-CC-1065 and functional analogs; the duocarmycin class of antitumor antibiotics, including yatakemycin; tropoloalkaloids; prodigiosin and roseophilin; the deoxybouvardin and RA-I class of antitumor agents; vancomycin, teicoplanin, ristocetin, chloropeptins and related agents; ramoplanin; the luzopeptins, quinoxapeptins, thiocoraline, BE-22179 and sandramycin; bleomycin A2 and functional analogs; HUN-7293; chlorofusin; CI-920 (fostriecin) and cytostatin; the combretastatins; storniamide A; phomazarin; ningalins; lamellarin O; lukianol A; piericidins; nothapodytine and mappicine; rubrolone; vindoline; and vinblastine (Figs. 2 and 3). BIOORGANIC CHEMISTRY The agents listed in the previous paragraph were selected on the basis of their properties; in many instances, they are agents related by a projected property. For example, (+)-CC-1065, the duocarmycins, and yatakemycin are antitumor antibiotics and related sequence-selective DNA minor groove alkylating agents. Representative of such efforts, studies to determine the structural features of yatakemycin and the duocarmycins that contribute to the sequence-selective DNA alkylation properties of these agents have resulted CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE 71 F i g . 2 . Recent total syntheses. Fig. 3. Additional recent total syntheses. in the identification of a unique source of catalysis for the DNA alkylation reaction. Efforts are under way to develop DNA cross-linking agents of a predefined crosslink, to further understand the nature of the noncovalent and covalent interactions between agents and DNA, and to apply this understanding to the de novo design of DNA-binding and DNA-effector agents. Techniques for the evaluation of the agent-DNA binding and alkylation properties, collaborative efforts in securing biological data, nuclear magnetic resonance structures of DNAagent complexes, molecular modeling, and studies of DNA-agent interactions are integral parts of the program. 72 CHEMISTRY 2006 Additional ongoing studies include efforts to define the fundamental basis of the DNA-binding or cleavage properties of bleomycin A2, sandramycin, and the luzopeptins; to design inhibitors of the folate-dependent enzymes glycinamide ribonucleotide transformylase and aminoimidazole carboxamide ribonucleotide transformylase as potential antineoplastic agents; to establish the chemical and biological characteristics responsible for the sleep-inducing properties of the endogenous lipid oleamide; to inhibit tumor growth through inhibition of angiogenesis; to inhibit aberrant gene transcription associated with cancer; and to control intracellular signal transduction through the discovery of antagonists or agonists that affect protein-protein interactions, including receptor dimerization. PUBLICATIONS Boger, D.L., Miyauchi, H., Du, W., Hardouin, C., Fecik, R.A., Cheng, H., Hwang, I., Hedrick, M.P., Leung, D., Acevedo, O., Guimarães, C.R.W., Jorgensen, W.L., Cravatt, B.F. Discovery of a potent, selective, and efficacious class of reversible α-ketoheterocycle inhibitors of fatty acid amide hydrolase effective as analgesics. J. Med. Chem. 48:1849, 2005. Capps, K.J., Humiston, J., Dominique, R., Hwang, I., Boger, D.L. Discovery of AICAR Tfase inhibitors that disrupt requisite enzyme dimerization. Bioorg. Med. Chem. Lett. 15:2840, 2005. Cheng, H., Chong, Y., Hwang, I., Tavassoli, A., Zhang, Y., Wilson, I.A., Benkovic, S.J., Boger, D.L. Design, synthesis, and 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-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. Choi, Y., Ishikawa, H., Velcicky, J., Elliott, G.I., Miller, M.M., Boger, D.L. Total synthesis of (–)- and ent-(+)-vindoline. Org. Lett. 7:4539, 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. Chou, T.-C., Gaun, Y., Soenen, D.R., Danishefsky, S.J., Boger, D.L. Potent reversal of multidrug resistance by ningalin and its use in drug combinations against human colon carcinoma xenografts in nude mice. Cancer Chemother. Pharmacol. 56:379, 2005. Du, W., Hardouin, C., Cheng, H., Hwang, I., Boger, D.L. Heterocyclic sulfoxide and sulfone inhibitors of fatty acid amide hydrolase. Bioorg. Med. Chem. Lett. 15:103, 2005. Guimarães, C.R.W., Boger, D.L., Jorgensen, W.L. Elucidation of fatty acid amide hydrolase inhibition by potent α-ketoheterocycle derivatives from Monte Carlo simulations. J. Am. Chem. Soc. 127:17377, 2005. Hamasaki, A., Zimpleman, J.M., Hwang, I., Boger, D.L. Total synthesis of ningalin D. J. Am. Chem. Soc. 127:10767, 2005. Leung, D., Du, W., Hardouin, C., Cheng, H., Hwang, I., Cravatt, B.F., Boger, D.L. Discovery of an exceptionally potent and selective class of fatty acid amide hydrolase inhibitors enlisting proteome-wide selectivity screening: concurrent optimization of enzyme inhibitor potency and selectivity. Bioorg. Med. Chem. Lett. 15:1423, 2005. Schnermann, M.J., Boger, D.L. Total synthesis of piercidin A1 and B1. J. Am. Chem. Soc. 127:15704, 2005. THE SCRIPPS RESEARCH INSTITUTE Tse, W.C., Boger, D.L. A fluorescent intercalator displacement (FID) assay for establishing DNA binding selectivity and affinity. In: Current Protocols in Nucleic Acid Chemistry. Beaucage, S.L., et al. (Eds.). Wiley & Sons, New York, in press. Walker, S., Chen, L., Hu, Y., Rew, Y., Shin, D., Boger, D.L. Chemistry and biology of ramoplanin: a lipoglycodepsipeptide with potent antibiotic activity. Chem. Rev. 105:449, 2005. Yuan, Z., Ishikawa, H., Boger, D.L. Total synthesis of natural (+)- and ent-(–)-4desacetoxy-6,7-dihydrovindorosine [corrected] and natural and ent-minovine: oxadiazole tandem intramolecular Diels-Alder/1,3-dipolar cycloaddition reaction [published correction appears in Org. Lett. 7:2079, 2005]. Org. Lett. 7:741, 2005. Chemical and Functional Genomic Approaches to Regenerative Medicine S. Ding, R. Abu-Jarour, R. Ambasudhan, C. Desponts, N. Emre, H.S. Hahm, S. Hilcove, J. Hsu, M. Kim, Y. Shi, S. Takanashi, W. Xiong, Y. Xu, S. Yao, D. Yue, Y. Zhao, X. Zhu ecent advances in stem cell biology may make possible new approaches for the treatment of a number of diseases, including cardiovascular disease, neurodegenerative disease, musculoskeletal disease, diabetes, and cancer. These approaches could involve cell replacement therapy and/or drug treatment to stimulate the body’s own regenerative capabilities by promoting survival, migration/homing, proliferation, and differentiation of endogenous stem/progenitor cells. However, such approaches will require identification of renewable cell sources of engraftable functional cells, an improved ability to manipulate proliferation and differentiation of the cells, and a better understanding of the signaling pathways that control the fate of the cells. Equipped with large arrayed molecular libraries— combinatorial chemical libraries (>100,000 discrete and diverse small molecules), cDNA overexpression libraries (>30,000 human and mouse genes) and small interfering RNA libraries (targeting >20,000 human and mouse genes)—and a high-throughput screening platform, we are developing and integrating chemical and functional genomic tools to study stem cell biology and regeneration. We screen these libraries to identify small molecules and genes that can control the fate of stem cells in various systems, including (1) self-renewal, as well as directed neuronal, cardiac, and pancreatic differentiations of pluripotent mouse and human embryonic stem cells; (2) directed neuronal differentiation and subtype neuron specification of human and rodent neural stem cells; (3) directed dif- R CHEMISTRY 2006 ferentiation of mesenchymal stem cells to osteogenic, adipogenic, chondrogenic, and myogenic lineages; (4) functional proliferation of cardiomyocytes and islets/beta cells in adults; (5) cellular plasticity and dedifferentiation of lineage-restricted somatic cells; and (6) developmental signaling pathways. In addition, we are doing systemic biochemical and cellular studies, including detailed investigations of structure-activity relationships, affinity chromatography for target identification, genome-wide expression analysis with microarrays, and cDNA and/or RNA interference complementation screens to map signaling pathways to characterize the molecular mechanism of these identified small molecules and genes. Recent examples of small molecules of interest include neuropathiazol, which can direct differentiation of primary rat adult neural stem cells selectively toward neurons; pluripotin, which can sustain self-renewal of murine embryonic stem cells in a chemically defined medium; and a purine analog that functions as a synergistic Wnt pathway agonist and can induce Xenopus axis duplication in combination with Wnt8. These studies may ultimately facilitate the therapeutic application of stem cells and the development of small-molecule drugs to stimulate tissue and organ regeneration in vivo. PUBLICATIONS Warashina, M., Min, K.H., Kuwabara, T., Huynh, A., Gage, F.H., Schultz, P.G., Ding, S. A synthetic small molecule that induces neuronal differentiation of adult hippocampal neural progenitor cells. Angew. Chem. Int. Ed. 45:591, 2006. Zhao, Y., Clark J., Ding, S. Genomic studies in stem cell systems. Curr. Opin. Mol. Ther. 7:43, 2005. Chemical Etiology of the Structure of Nucleic Acids A. Eschenmoser, R. Krishnamurthy, G. Kumar, F. De Riccardis, R. Kondreddi, Y. Osornio, M. Guerrero D uring the past year we worked on the following projects. O L I G O M E R S B A S E D O N T R I A Z I N E - TA G G E D OLIGODIPEPTIDE AND OLIGODIPEPTOID BACKBONES We continued our studies on the self- and crosspairing properties of triazine-tagged oligodipeptides consisting of alternating aspartic acid (Asp) and glutamic acid (Glu) residues (Fig. 1). Although the oligo-(AspGlu)dipeptides tagged with 2,4-diaminotriazine had strong THE SCRIPPS RESEARCH INSTITUTE 73 F i g . 1 . Formulas of triazine-tagged oligomers. IDA indicates iminodiacetic acid. cross-pairing with complementary RNA and DNA oligonucleotide sequences, the corresponding oligo(AspGlu)-dipeptides tagged with 2,4-dioxotriazine, much to our surprise, showed only weak pairing with the natural oligonucleotides. The intrasystem self-pairing of 2,4-diaminotriazine–, 2,4-dioxotriazine–, and 2amino-4-oxo-triazine–tagged oligo-(AspGlu)-dipeptides was equally weak. The base-pairing properties of triazine-tagged oligo(AspAsp)-dipeptides paralleled the trends observed in the oligo-(AspGlu)-dipeptide series, but, as expected, were consistently weaker in base-pairing strength. A variation of the oligo-(AspAsp)-dipeptide shown in Figure 1B is the achiral oligodipeptoid derived from iminodiacetic acid units (Fig. 1C). Again, oligodipeptoids containing 2,4-diaminotriazines cross-paired with RNA and DNA oligonucleotides, but no discernible pairing occurred with the 2,4-dioxotriazine–tagged oligodipeptoids. Our studies indicate that the family of triazine-based recognition elements lacks the balance in pairing strength characteristic of the purine-pyrimidine combination in the natural series, presumably because of the imbalance in protophilicity of dioxotriazines (pKa about 6) vs diaminotriazines (pKa about 3.9), in an aqueous environment. Such imbalance in pairing potential leads to the conclusion that triazines, irrespective of their generational simplicity, would have been functionally incapable of fulfilling the role of recognition elements in a primordial genetic system. This realization has led us pursue the following project. 74 CHEMISTRY 2006 O L I G O M E R S B A S E D O N 5 - A M I N O P Y R I M I D I N E – TA G G E D OLIGODIPEPTIDE BACKBONES Oligomerization of hydrogen cyanide, a potentially prebiotic reaction generally assumed to have acted as the primordial source of the canonical nucleobases adenine and guanine, produces in addition pyrimidines, not the canonical ones, but mostly 5-aminopyrimidines, which do not play a role in contemporary biology (Fig. 2). THE SCRIPPS RESEARCH INSTITUTE Mittapalli, G.K., Osornio, Y.M., Guererro, M.A., Reddy, K.R., Krishnamurthy, R., Eschenmoser, A. Mapping the landscape of potentially primordial informational oligomers: oligodipeptides and oligodipeptoids tagged with 2,4-disubstituted-5amino-pyrimidines as recognition elements. Angew. Chem. Int. Ed., in press. Wagner, T., Han, B., Koch, G., Krishnamurthy, R., Eschenmoser, A. Tautomerism in 5,8-diaza-7,9-dicarbaguanine (“alloguanine”). Helv. Chim. Acta 88:1960, 2005. Organic, Materials, and Analytical Chemistry M.G. Finn, J. Kuzelka, D. Prasuhn, S. Presolski, V. Rodionov, Y.-H. Lim, B. Venkataiah n addition to synthetic chemistry research on viruses, our program encompasses organic, organometallic, and materials chemistry. Special emphasis is placed on methods of chemical synthesis, the discovery of functional molecules, and catalysis. I M E C H A N I S M S A N D A P P L I C AT I O N S O F C L I C K CHEMISTRY F i g . 2 . Top, Formulas of 5-aminopyrimidine heterocycles. Bottom, Also shown, as a representative example, is the 5-amino-2,4-dioxo– tagged oligomer containing alternating residues of aspartic and glutamic acid. Chemical reasoning makes a study of the base-pairing properties of the members of this family highly desirable; they not only can potentially act as substitutes in the 2 canonical Watson-Crick base pairs but also offer a unique opportunity to tag polypeptide chains bearing recurring carboxyl groups by using simple (regioselective) amide formation. We have synthesized 5-aminopyrimidine–tagged oligo-(AspGlu)-dipeptides (up to hexadecamers) by using all 4 members of the family and have explored base-paring properties of the tagged dipeptides. Preliminary results indicated crosspairing between all of these recognition elements with the corresponding complementary RNA and DNA oligonucleotides, although the 5-aminopyrimidine heterocycles have stark differences in base-pairing strength. Also, cross-pairing occurs between the 2,4-diaminotriazine–tagged oligo-(AspGlu)-dipeptides and 5-amino2,4-dioxopyrimidine–tagged oligo-(AspGlu)-dipeptides. PUBLICATIONS Eschenmoser, A. Searching for nucleic acid alternatives. Chimia 59:836, 2005. Mittapalli, G.K., Reddy, K.R., Xiong, H., Munoz, O., Han, B., De Riccardis, F., Krishnamurthy, R., Eschenmoser, A. Mapping the landscape of potentially primordial informational oligomers: oligodipeptides and oligodipeptoids tagged with triazines as recognition elements. Angew. Chem. Int. Ed., in press. The copper-catalyzed azide-alkyne cycloaddition reaction, discovered in 2002 by V.V. Fokin and K.B. Sharpless, Department of Chemistry, has been adopted by chemists all over the world for organic synthesis, drug development, and materials science. We have continued our mechanistic studies of the reaction and our efforts to apply the reaction to the synthesis of biologically active compounds, materials, and bioconjugates. A protocol for using the reaction in the polyvalent decoration of scaffolds has been optimized (Fig. 1). In F i g . 1 . Bioconjugation of alkynes to polyvalent azides via the copper complex of bathophenanthroline ligand 1. the most demanding situations, with sensitive proteins at micromolar concentrations, the use of sulfonated bathophenanthroline (compound 1 in Fig. 1) is vital. We continue to develop new catalysts to remove the last barrier to convenient application of the method, the need to perform the reaction in an inert atmosphere when protein instability prevents the simultaneous use of a reducing agent. In addition, a variation of the CHEMISTRY 2006 standard copper-catalyzed process has been uncovered in which aromatic azides react with alkynes to give the 1,5-triazole isomer rather than the customary 1,4-triazole. Last, mechanistic studies have revealed changes in the rate-limiting steps when certain copper-binding ligands and substrates are used. SYNTHESIS AND USE OF FORMAMIDINE COMPOUNDS We synthesized amidines, including formamidines and formamidine ureas, and tested them for binding to the acetylcholine-binding proteins of Lymnaea stagnalis and Aplysia californica, soluble homologs of the nicotinic acetylcholine receptor. Compounds 2, 3, and 4 (Fig. 2) have moderate to high affinities for the target THE SCRIPPS RESEARCH INSTITUTE 75 Díaz, D.D., Rajagopal, K., Strable, E., Schneider, J., Finn, M.G. “Click” chemistry in a supramolecular environment: stabilization of organogels by copper(I)-catalyzed azide-alkyne [3 + 2] cycloaddition. J. Am. Chem. Soc. 128:6056, 2006. Díaz, D.D., Ripka, A.S., Finn, M.G. 1-(tert-Butyl-imino-methyl)-1,3-dimethyl-urea hydrochloride. Org. Synth. 82:59, 2005. Johnson, J.A., Lewis, D.R., Díaz, D.D., Finn, M.G., Koberstein, J.T., Turro, N.J. Synthesis of degradable model networks via ATRP and click chemistry. J. Am. Chem. Soc. 128:6564, 2006. Meng, J., Fokin, V.V., Finn, M.G. Kinetic resolution by copper-catalyzed azidealkyne cycloaddition. Tetrahedron Lett. 46:4543, 2005. Punna, S., Kaltgrad, E., Finn, M.G. “Clickable” agarose for affinity chromatography. Bioconjug. Chem. 16:1536, 2005. Punna, S., Meunier, S., Sen Gupta, S., Venkataiah, B., Truong, P., McGavern, D., Finn, M.G. Polyvalent inhibition of the LFA-ICAM interaction. J. Am. Chem. Soc., in press. Rae, C.S., Khor, I.W., Wang, Q., Destito, G., Gonzalez, M.J., Singh, P.R., Thomas, D.M., Estrada, M.N., Powell, E., Finn, M.G., Manchester, M. Systemic trafficking of plant virus nanoparticles in mice via the oral route. Virology 343:224, 2005. Sen Gupta, S., Kuzelka, J., Singh, P., Lewis, W.G., Manchester, M., Finn, M.G. Accelerated bioorthogonal conjugation: a practical method for the ligation of diverse functional molecules to a polyvalent virus scaffold. Bioconjug. Chem. 16:1572, 2005. F i g . 2 . Amidine derivatives that bind to nicotinic receptor proteins. proteins, representing a new class of receptor ligands. Whereas amidines such as compound 4 are relatively stable, formamidine ureas such as compounds 2 and 3 are deactivated during a period of approximately 1 hour by hydrolysis when not bound. Using fluorescence spectroscopy and x-ray crystallography, we showed that the bound molecules reside in the canonical hydrophobic pocket. Electrophysiologic measurements indicated that compound 4 is a nicotinic receptor agonist, consistent with its observed binding behavior. We are extending this research to molecules specific for subtypes of the nicotinic receptor family. These studies are conducted in collaboration with P. Taylor, University of California, San Diego, and A. Markou, Molecular and Integrative Neurosciences Department. Sen Gupta, S., Raja, K.S., Kaltgrad, E., Strable, E., Finn, M.G. Virus-glycopolymer conjugates by copper(I) catalysis of atom transfer radical polymerization and azide-alkyne cycloaddition. Chem. Commun. (Camb.) 4315, 2005, Issue 34. Whiting, M., Muldoon, J., Lin, Y.-C., Silverman, S.M., Lindstrom, W., Olson, A.J., Kolb, H.C., Finn, M.G., Sharpless, K.B., Elder, J.H., Fokin, V.V. Inhibitors of HIV-1 protease via in situ click chemistry. Angew. Chem. Int. Ed. 45:1435, 2006. Wu, P., Malkoch, M., Hunt, J.N., Vestberg, R., Kaltgrad, E., Finn, M.G., Fokin, V.V., Sharpless, K.B., Hawker, C.J. Multivalent, bifunctional dendrimers prepared by click chemistry. Chem. Commun. (Camb.) 5775, 2005, Issue 46. Design of Functional Synthetic Systems M.R. Ghadiri, G. Ashkenasy, N. Ashkenasy, J. Beierle, A. Chavochi, N. Gianneschi, W.S. Horne, Z.-Z. Huang, P. Imming, L. Leman, A. Loutchnikov, A. Montero, L. Motiei, D. Nicoletti, Y. Norikane, J. Picuri, N. Rahe, D. Radu, S. Rahimipour, J. Shin, R. Yamasaki, Y.S. Yoo PUBLICATIONS Díaz, D.D., Converso, A., Sharpless, K.B., Finn, M.G. 2,6-Dichloro-9-thiabicyclo[3.3.1]nonane: multigram display of azides and cyanides components on a versatile scaffold. Molecules 11:212, 2006. Díaz, D.D., Finn, M.G. Facile synthesis of N,N′-bis[formamidine]ureas and symmetrical N,N′-dsubstituted formamidines. Lett. Org. Chem. 2:621, 2005. Díaz, D.D., Finn, M.G., Mishima, M. Substituent effects on the gas-phase basicity of formamidine ureas. Eur. J. Org. Chem. 235, 2006, Issue 1. Díaz, D.D., Lewis, W.G., Finn, M.G. Acid-mediated amine exchange of N,Ndimethylformamidines: preparation of electron-rich formamidines. Synlett 2214, 2005, Issue 14. Díaz, D.D., Lewis, W.G., Finn, M.G. Activation of urea as a leaving group in substitution reactions of formamidine ureas. Chem. Lett. 34:78, 2005. e are engaged in a multidisciplinary research effort to uncover new chemical and biochemical approaches for the design of functional molecular, supramolecular, and complex self-organized systems. Our endeavors span disciplines ranging from synthetic organic, bioorganic, and physical organic chemistry to nanotechnology, biophysics, enzymology, and molecular biology. Current research includes the design of synthetic peptide catalysts, antimicrobial self-assembling peptide nanotubes, semisynthetic allosteric enzymes, self-replicating molecular systems and emergent net- W 76 CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE works, single-molecule stochastic DNA sensing, molecular computation, and prebiotic chemistry. ANTIMICROBIAL PEPTIDE NANOTUBES We showed that appropriately designed cyclic peptide subunits can self-assemble through hydrogen bond–directed ring stacking into open-ended hollow tubular structures that have marked antibacterial and antiviral activities in vitro. The effectiveness of this novel supramolecular class of bioactive species as selective antibacterial agents was highlighted by the high efficacy of one of these antimicrobials against lethal methicillin-resistant Staphylococcus aureus infections in mice. Currently, we are exploring rational design of cyclic glycopeptides and selections from combinatorial libraries to discover novel antiviral and anticancer supramolecular compounds (Fig. 1). F i g . 2 . Schematic representation of an intrasterically inactivated inhibitor-DNA-enzyme construct (left) and the DNA hybridization–triggered enzyme activation (right). The construct can be used to sense low concentrations of cDNA because of its built-in capacity for signal amplification via rapid turnover of substrate. membrane protein α-hemolysin as a rapid and highly sensitive sensor element for stochastic analysis of the molecules lodged or trapped inside the protein pore; the analysis relies on detecting the perturbations in the conductance levels produced in the ion channel in the native protein. Using this technique, we developed an approach by which a single-stranded DNA molecule can be trapped in a specific configuration inside an αhemolysin channel (Fig. 3), manipulated, and studied F i g . 1 . Antiviral agents based on self-assembling cyclic peptide nanotubes. Cyclic D,L-α-peptides act on endosomal membranes to prevent the development of low pH in endocytic vesicles, arrest the escape of virions from the endosome, and abrogate adenovirus infection. DESIGN OF SIGNAL SELF-AMPLIFYING DNA SENSORS We constructed a novel sequence-specific DNA detection system based on rationally designed semisynthetic enzymes. The system is composed of covalently associated inhibitor-DNA-enzyme modules that function via DNA hybridization–triggered allosteric enzyme activation and signal amplification through substrate turnover (Fig. 2). The functional capacity of the system is highlighted by the sequence-specific detection of approximately 10 fmol of DNA in less than 3 minutes under physiologic conditions. Our studies suggest that rationally designed intrasterically regulated enzymes may be a promising new class of reagents for highly sensitive, rapid, 1-step detection of label-free DNA sequences that does not depend on polymerase chain reactions. with high sensitivity at the single-molecule level. Moreover, a single adenine nucleotide at a specific location on a strand of polydeoxycytidine can be detected by its characteristic effect in reducing the ion conductance in α-hemolysin. We are extending this approach to the design of rapid single-molecule DNA sensing and sequencing. S T O C H A S T I C A N A LY S I S O F S I N G L E - M O L E C U L E D N A SYNTHETIC NETWORKS R O TA X A N E S Living cells use complex networks of evolutionarily selected biomolecular interactions and chemical transformations to process multiple extracellular input signals We are interested in the study of matter at the level of single molecules. For these studies we use the trans- F i g . 3 . Functional supramolecular chemistry at the single-mole- cule level. Single strands of DNA can be captured inside an αhemolysin transmembrane pore protein to form single-species pseudorotaxanes composed of α-hemolysin and DNA. This process can be used to identify a single adenine nucleotide at a specific location on a strand of DNA on the basis of the characteristic reductions in the α-hemolysin ion conductance. CHEMISTRY 2006 rapidly and simultaneously. We are interested in understanding and experimentally modeling the organizational and functional properties of biological networks. We have developed a general strategy for the design and construction of self-organized synthetic peptide networks based on the sequence-selective autocatalytic and cross-catalytic template-directed coiled coil peptide fragment condensation reactions in aqueous solutions. The synthetic networks have some of the basic architectural and dynamic features of the living networks, reorganize in response to changes in environmental conditions and inputs (Fig. 4), and perform THE SCRIPPS RESEARCH INSTITUTE 77 marginally prebiotic. We showed that carbonyl sulfide, a simple gas present in the emissions from present-day volcanoes, is a condensing agent that brings about the formation of peptides from amino acids under mild conditions in aqueous solution (Fig. 5). We have studied the F i g . 5 . Peptide formation under plausibly prebiotic reaction con- ditions. Carbonyl sulfide, a volcanic gas, is the most simple and effective amino acid–condensing agent for the formation of peptides in aqueous solutions. F i g . 4 . Adaptive reorganization in a synthetic peptide network. The graph structure or wiring of a synthetic peptide network responds dramatically to changes in the environmental stimuli (pH or salt content). basic Boolean logic functions such as OR, NOR, and NOTIF logic. We suggest that the ability to rationally construct predictable chemical circuitry might be useful in advancing the modeling and better understanding of some of the basic dynamic information-processing characteristics of the more complex cellular networks. PREBIOTIC CHEMISTRY In almost all discussions of prebiotic chemistry, it is assumed that amino acids, nucleotides, and possibly other monomers were first formed on Earth or brought to it in comets and meteorites and that the monomers subsequently condensed nonenzymatically to form oligomeric products. Unfortunately, attempts to create plausibly prebiotic polymerization reactions have met with limited success. Direct heating of solid mixtures leads to nonspecific products, and the condensing agents that have been studied, with the possible exception of inorganic polyphosphates, are relatively inefficient and/or carbonyl sulfide–mediated condensations of α-amino acids under aerobic and anaerobic conditions in the absence of any added reagents and in the presence of metal ions, oxidizing agents, or alkylating agents. Depending on the reaction conditions and additives used, exposure of α-amino acids to carbonyl sulfide generates peptides in yields of up to 80% in minutes to hours at room temperature. PUBLICATIONS Askkenasy, N., Sánchez-Quesada, J., Bayley, H., Ghadiri, M.R. Recognizing a single base in an individual DNA strand: a step toward DNA sequencing in nanopores. Angew. Chem. Int. Ed. 44:1401, 2005. Horne, S.W., Ashkenasy, N., Ghadiri, M.R. Modulating charge transfer through cyclic D,L-α-peptide self-assembly. Chemistry 11:1137, 2005. Horne, S.W., Wiethoff, C.M., Cui, C., Wilcoxen, K.M., Amorin, M., Ghadiri, M.R., Nemerow, G.R. Antiviral cyclic D,L-α-peptides: targeting a general biochemical pathway in viral infections. Bioorg. Med. Chem. 13:5145, 2005. Yadav, M.K., Redman, J.E., Leman, L.J., Alvarez-Gutiérrez, J.M., Zhang, Y., Stout, C.D., Ghadiri, M.R. Structure-based engineering of internal cavities in coiled-coil peptides. Biochemistry 44:9723, 2005. 78 CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE A Merging of Chemistry and Biology K.D. Janda, J. Ashley, C. Berndt, G. Boldt, A. Brogan, C. Chung, S. De Lamo Marin, T. Dickerson, L. Eubanks, M. Hixon, A. Ino, G. Kaufmann, J. Kennedy, Y. Kim, J. Liu, Y. Liu, C. Lowery, H. Ma, S. Mahajan, L. McAllister, G. McElhaney, K. McKenzie, J. Mee, M. Meijler, J. Park, S. Steiniger, J. Treweek, A. Willis, Y. Xu, B. Zhou, H. Zhou uring the past year, we explored various applications of organic chemistry at the interface of chemistry and biology. Representative examples of our results were obtained in 3 research programs: catalysis of retinal isomerization by the nicotine metabolite nornicotine, “superactivation” of botulinum neurotoxin by small molecules, and the development of a cocaine esterase–bacteriophage construct with suitable kinetics for the degradation of cocaine in humans. D A LT E R E D R E T I N O I D H O M E O S TA S I S C ATA LY Z E D B Y A N I C O T I N E M E TA B O L I T E In recent years, we have been studying the role of long-lived drugs of abuse and their metabolites in drugrelated diseases. Much of this effort has centered on the Maillard reaction, a process by which amine-containing molecules irreversibly react with proteins through the intermediacy of glucose, the dominant serum monosaccharide. The mechanism of the Maillard reaction parallels that of amine organocatalysts; iminium and/or enamine intermediates are necessary for rate enhancement. In the past year, we expanded our studies beyond the Maillard reaction to other biological processes in which iminium ion intermediates are critical. Retinoids (vitamin A) play 2 major roles in higher animals: light absorption in vision and gene regulation in growth and development. Specifically, these processes are regulated by the conformation of the double bonds in the polyunsaturated hydrocarbon chain. For example, in the visual cycle, 11-Z-retinal is converted to all-Eretinal by a photon of light, ultimately leading to the perception of vision. Much of the biosynthetic pathways leading up to this reaction are controlled by the formation of iminium ions between the retinal terminal aldehyde group and a lysine side chain from an appropriate enzyme. We hypothesized that nornicotine, a metabolite of nicotine, could also perform this type of chemistry (Fig. 1) and thus alter the concentrations of retinal intermediates. This reaction would provide an intriguing F i g . 1 . Mechanism of nornicotine-catalyzed Z-to-E isomerization. mechanism for the pathologic changes in key smoking-related diseases, because the accumulation of compounds such as all-E-retinal feeds the N-retinylidene-N-retinylethanolamine biosynthetic pathway, forming an undigestible byproduct of the visual cycle and a fluorescent chromophore characteristic of the pathologic changes in age-related macular degeneration, a leading cause of blindness. Smoking is accepted as the primary environmental factor contributing to agerelated macular degeneration, and thus elucidating the molecular mechanism of this contribution is clinidand retinal compounds, we conclusively showed that nornicotine can indeed catalyze the Z-to-E isomerization of unsaturated compounds at rates that could have biological significance in the context of disease. S U P E R A C T I VAT I O N O F B O T U L I N U M N E U R O T O X I N S E R O T Y P E A L I G H T - C H A I N M E TA L L O P R O T E A S E The 7 neurotoxins (A–G) of the bacterium Clostridium botulinum are the most lethal poisons known. Exposure to these toxins leads to progressive flaccid paralysis resulting from cleavage of proteins critical for proper release of neurotransmitters from peripheral nerve cells. Despite their potent toxicity, botulinum neurotoxins are widely used in medicine, as well as cosmetically for treating facial wrinkles. Conditions including multiple sclerosis, stroke, cerebral palsy, migraine, and backache can all be treated with the neurotoxins. Yet, repeated exposure to the toxins can result in the development of a marked immune response to them, thereby compromising their efficacy. Tolerance develops most rapidly when patients are treated frequently with high doses of the toxins. We speculated that the coadministration of a botulinum neurotoxin with a molecule that can “activate” the catalytic activ- CHEMISTRY 2006 ity of the toxin would lead to lower doses, thus reducing the unintended immune response. In recent investigations of light-chain metalloprotease inhibitors of botulinum neurotoxin A, we discovered that the molecule arginine hydroxamic acid is a modest inhibitor. Using this compound as a guide, we prepared a small collection of compounds containing a zinc-binding motif (2-acylthiophene) combined with an arginine-side-chain mimetic (acylguanidine). To our surprise, although no inhibition occurred, one compound (compound 1 in Fig. 2) consistently produced a 2-fold THE SCRIPPS RESEARCH INSTITUTE 79 the importance of botulinum neurotoxins continues to expand, methods such as this may ultimately provide a method for minimizing dosage of the toxins and thereby increase the clinical efficacy of the molecules. DEGRADING COCAINE WITH VIRUSES Cocaine is a powerful stimulant and among the most reinforcing of all drugs. Consequently, abuse of cocaine continues to be a major problem. Cocaine acts as an indirect dopamine agonist by blocking the dopamine transporter in the pleasure-reward center of the brain. This obstruction leads to an excess of dopamine in the synapses, amplifying the sensation of pleasure. Despite intensive efforts, no effective pharmacotherapy for cocaine abuse exists. The inherent difficulties in antagonizing a blocker have led to the development of proteinbased therapeutics designed to treat cocaine abuse. In an approach termed immunopharmacotherapy, we have devoted extensive efforts to the use of antibodies to cocaine that can sequester cocaine, retarding its ability to enter the CNS. We have also developed a parallel strategy that involves use of catalytic antibodies specific for the hydrolysis of the benzoyl ester of cocaine to give the nonpsychoactive products benzoate and methyl ecgonine (Fig. 3). F i g . 2 . Chemical structures of molecules that can superactivate botulinum neurotoxin serotype A. The specific motifs used in the design of these compounds are highlighted. enhancement of activity. Further structure-activity relationship studies revealed that specific features of this compound were critical for activation, such as the thiophene sulfur atom and the acylguanidine group. When these initial screening efforts were completed, compound 2 (Fig. 2) was the most potent activator. Because of the clinical promise of an activator of botulinum neurotoxin, we further examined the mechanism of this phenomenon. Extensive kinetic characterization indicated that these compounds operate primarily by reducing the Michaelis constant (Km), not by altering the turnover number (k cat). In this context, compound 2 is the most potent small-molecule activator of a protease reported to date, with up to 14-fold rate enhancement at limiting concentrations of substrate. Indeed, as little as 2-fold enzyme activation has previously been reported as a state of superactivation. In total, the activation profile and structure-activity relationship for activation suggests the presence of a specific “activation domain” on the enzyme. Because F i g . 3 . Hydrolysis products resulting from cleavage of cocaine esters. Both the uncatalyzed reaction (path a) and the cocaine esterase–catalyzed hydrolysis (path b) pathways are shown. Although the potential of this method has been demonstrated in rodent models of cocaine overdose and reinforcement, the kinetic constants of these antibodies must be improved before the method will be practical as a clinical treatment. Furthermore, these approaches are only effective in the periphery, whereas a pharmacotherapy that could act in both the CNS and the periphery is desirable. Bacteriophages are viruses that infect bacteria yet lack intrinsic tropism for eukaryotic cells. Because of 80 CHEMISTRY 2006 the genetic flexibility of bacteriophages, a wide range of proteins and peptides can be expressed on the phage coat in an approach termed phage display. Furthermore, phage molecules can penetrate virtually all tissues, including the CNS. We recently reported that cocaine-binding antibodies displayed on the surface of bacteriophages can be administered intranasally and that the treated animals are protected from the locomotor stimulation associated with exposure to cocaine. However, because of the requisite 1:1 stoichiometry of any traditional antibody pharmacotherapy, obtaining a meaningful concentration of the therapeutic agent in vivo is difficult. We envisioned that this limitation could be overcome by displaying a catalyst on the phage surface that can degrade cocaine, yielding a therapeutically practical approach for treating cocaine abuse. For these studies, we used cocaine esterase, a globular bacterial enzyme that is the most efficient protein catalyst for cocaine hydrolysis reported to date. We displayed this enzyme on the phage coat and then used high-performance liquid chromatography to determine the kinetic parameters. We found that the catalytic efficiency of cocaine esterase–phage constructs was reduced relative to the efficiency of the native enzyme yet exceeded the postulated therapeutically relevant threshold. No reported catalytic antibody capable of cocaine hydrolysis achieves this value, and indeed, only recently described “designer” mutants of the enzyme butyrylcholinesterase are comparable to our cocaine esterase–phage constructs. These results indicate that phage display of clinically relevant enzymes can be achieved without compromising the catalytic efficacy of the desired enzyme. We envision that this new technology will stimulate further development of other protein-based treatments for CNS-related disorders and will lead to powerful tools to combat drug abuse. PUBLICATIONS Boldt, G.E., Dickerson, T.J., Janda, K.D. Emerging chemical and biological approaches for the preparation of discovery libraries. Drug Discov. Today 11:143, 2006. Boldt, G.E., Eubanks, L.M., Janda, K.D. Identification of a botulinum neurotoxin A protease inhibitor displaying efficacy in a cellular model. Chem. Commun. (Camb.) 3063, 2006, Issue 29. Boldt, G.E., Kennedy, J.P., Hixon, M.S., McAllister, L.A., Barbieri, J.T., Tzipori, S., Janda, K.D. Synthesis, characterization and development of a high-throughput methodology for the discovery of botulinum neurotoxin A inhibitors. J. Comb. Chem. 8:513, 2006. Boldt, G.E., Kennedy, J.P., Janda, K.D. Identification of a potent botulinum neurotoxin A protease inhibitor using in situ lead identification chemistry. Org. Lett. 8:1729, 2006. THE SCRIPPS RESEARCH INSTITUTE Brogan, A.P., Dickerson, T.J., Boldt, G.E., Janda, K.D. Altered retinoid homeostasis catalyzed by a nicotine metabolite: implications in macular degeneration and normal development. Proc. Natl. Acad. Sci. U. S. A. 102:10433, 2005. Carrera, M.R.A., Trigo, J.M., Wirsching, P., Roberts, A.J., Janda, K.D. Evaluation of the anticocaine monoclonal antibody GNC92H2 as an immunotherapy for cocaine overdose. Pharmacol. Biochem. Behav. 81:709, 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. Dickerson, T.J., Janda, K.D. Recent advances for the treatment of cocaine abuse: central nervous system immunopharmacotherapy. AAPS J. 7:E579, 2005. Eubanks, L.M., Dickerson, T.J., Janda, K.D. Vitamin B2-mediated cellular photoinhibition of botulinum neurotoxin A. FEBS Lett. 579:5361, 2005. Kaufmann, G.F., Sartorio, R., Lee, S.H., Mee, J.M., Altobell, L.J. III, Kujawa, D.P., Jeffries, E., Clapham, B., Meijler, M.M., Janda, K.D. Antibody interference with N-acyl homoserine lactone-mediated bacterial quorum sensing. J. Am. Chem. Soc. 128:2802, 2006. Kim, Y., Lillo, A., Moss, J.A., Janda, K.D. A contiguous stretch of methionine residues mediates the energy-dependent internalization mechanism of a cell-penetrating peptide. Mol. Pharm. 2:528, 2005. Lee, B.S., Mahajan, S., Janda, K.D. Asymmetric dihydroxylation catalyzed by ionic polymer-supported osmium tetroxide. Tetrahedron Lett. 46:4491, 2005. Lee, B.S., Mahajan, S., Janda, K.D. Molecular iodine-catalyzed imine activation for three-component nucleophilic addition reactions. Synlett 1325, 2005, Issue 8. Lillo, A.M., McKenzie, K.M., Janda, K.D. Phage-displayed antibody libraries. In: Cell Biology: A Laboratory Handbook, 3rd ed. Celis, J., et al. (Eds.). Academic Press, San Diego, 2006, p. 491. Ma, H., Zhou, B., Kim, Y., Janda, K.D. A cyclic peptide-polymer probe for the detection of Clostridium botulinum neurotoxin serotype A. Toxicon 47:401, 2006. Matsushita, M., Meijler, M.M., Wirsching, P., Lerner, R.A., Janda, K.D. A blue fluorescent antibody-cofactor sensor for mercury. Org. Lett. 7:4943, 2005. McAllister, L.A., Hixon, M.S., Kennedy, J.P., Dickerson, T.J., Janda, K.D. Superactivation of the botulinum neurotoxin serotype A light chain metalloprotease: a new wrinkle in botulinum neurotoxin. J. Am. Chem. Soc. 128:4176, 2006. McKenzie, K.M., Meijler, M.M., Lowery, C.A., Boldt, G.E., Janda, K.D. A furanosyl-carbonate autoinducer in cell-to-cell communication of V. harveyi. Chem. Commun. (Camb.) 4863, 2005, Issue 38. Moss, J.A, Stokols, S., Hixon, M.S., Ashley, F.T., Chang, J.Y., Janda, K.D. Solidphase synthesis and kinetic characterization of fluorogenic enzyme-degradable hydrogel cross-linkers. Biomacromolecules 7:1011, 2006. Qi, L., Yamamoto, N., Meijler, M.M., Altobell, L.J. III, Koob, G.F., Wirsching, P., Janda, K.D. ∆9-Tetrahydrocannabinol immunochemical studies: haptens, monoclonal antibodies, and a convenient synthesis of radiolabeled ∆9-tetrahydrocannabinol. J. Med. Chem. 48:7389, 2005. Rogers, C.J., Dickerson, T.J., Janda, K.D. Kinetic isotope and thermodynamic analysis of the nornicotine-catalyzed aqueous aldol reaction. Tetrahedron 62:352, 2006. Rogers, C.J., Dickerson, T.J., Wentworth, P., Jr., Janda, K.D. A high-swelling reagent scaffold suitable for use in aqueous and organic solvents. Tetrahedron 61:12140, 2005. Rogers, C.J., Mee, J.M., Kaufmann, G.F., Dickerson, T.J., Janda, K.D. Toward cocaine esterase therapeutics J. Am. Chem. Soc. 127:10016, 2005. Shimomura, O., Lee, B.S., Meth, S., Suzuki, H., Mahajan, S., Nomura, R., Janda, K.D. Synthesis and application of polytetrahydrofuran-grafted polystyrene (PS-PTHF) resin supports for organic synthesis. Tetrahedron 61:12160, 2005. CHEMISTRY 2006 Shute, T.S., Matsushita, M., Dickerson, T.J., La Clair, J.J., Janda, K.D., Burkart, M.D. A site-specific bifunctional protein labeling system for affinity and fluorescent analysis. Bioconjug. Chem. 16:1352, 2005. Toker, J.D., Tremblay, M.R., Yli-Kauhaluoma, J., Wentworth, A.D., Zhou, B., Wentworth, P., Jr., Janda, K.D. Exploring the scope of the 29G12 antibody catalyzed 1,3-dipolar cycloaddition reaction. J. Org. Chem. 70:7810, 2005. Wu, W., Luo, Y., Sun, C., Liu, Y., Kuo, P., Varga, J., Xiang, R., Reisfeld, R., Janda, K.D., Edgington, T.S., Liu, C. Targeting cell-impermeable prodrug activation to tumor microenvironment eradicates multiple drug-resistant neoplasms. Cancer Res. 66:970, 2006. Xu, Y., Shi, J., Yamamoto, N., Moss, J.A., Vogt, P.K., Janda, K.D. A credit-card library approach for disrupting protein-protein interactions. Bioorg. Med. Chem. 14:2660, 2006. Xu, Y., Yamamoto, N., Ruiz, D.I., Kubitz, D.S., Janda, K.D. Squaric monoamide monoester as a new class of reactive immunization hapten for catalytic antibodies Bioorg. Med. Chem. Lett. 15:4304, 2005. Yamashita, M., Lee, S.-H., Koch, G., Zimmermann, J., Clapham, B., Janda, K.D. Solid-phase synthesis of oxazolones and other heterocycles via Wang resin-bound diazocarbonyls. Tetrahedron Lett. 46:5495, 2005. Yao, Y., Martinez-Yamout, M., 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. Zhang, L., Long, H., Boldt, G.E., Janda, K.D., Schatz, G.C., Lewis, F.D. α- and β-Stilbenosides as base-pair surrogates in DNA hairpins. Org. Biomol. Chem. 4:314, 2006. 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. Strategies to Ameliorate Protein Misfolding Diseases J.W. Kelly, J. Bieschke, D.A. Bosco, E. Culyba, M.T.A. Dendle, W. D’Haeze, T.R. Foss, D.M. Fowler, Y. Fu, J. Gao, M.-Y. Gao, S.M. Johnson, T. Mu, E.T. Powers, A.R. Sawkar, L. Segatori, S. Siegel, J.Y. Suk, R.L. Wiseman, I. Yonemoto, Z. Yu ur goal is to better understand the molecular mechanisms that lead to protein misfolding diseases, including Alzheimer ’s, Parkinson’s, and Gaucher diseases, so that we can design new strategies to ameliorate such maladies. To accomplish this goal, we use organismal and cell biological disease models and spectroscopic and biophysical approaches in combination with chemical synthesis and molecular biology techniques. Successful collaborations with W.E. Balch, Department of Cell Biology, P. Wentworth, Jr., Department of Chemistry, and A. Dillin, the Salk Institute for Biological Studies, La Jolla, California, are critical to achieve our goals. O TRANSTHYRETIN AMYLOIDOGENESIS Transthyretin is a 55-kD homotetrameric protein that transports holo-retinol binding protein and L-thy- THE SCRIPPS RESEARCH INSTITUTE 81 roxine in the blood and cerebrospinal fluid. More than 99.5% of the transthyretin binding sites for L-thyroxine remain unoccupied in blood, and only about 50% of transthyretin tetramers in plasma are bound to a single holo-retinol binding protein. As a consequence of a mutation or denaturation stress associated with aging and/or oxidative stress, dissociation of the native transthyretin tetramer, the rate-limiting step for amyloidogenesis, followed by changes in the tertiary structure of the monomer make the monomeric subunits competent to misassemble into aggregates, including amyloid fibrils. The deposition of transthyretin amyloid is linked with a number of human diseases, including senile systemic amyloidosis, familial amyloid polyneuropathy (FAP), familial amyloid cardiomyopathy, and CNS-selective amyloidosis. A suitable strategy to slow or prevent the formation of aggregates is to inhibit the rate-limiting dissociation of the transthyretin tetramer by making the native state more stable than the dissociative transition state. We have designed, synthesized, and characterized several classes of structurally distinct small molecules that bind to and stabilize the transthyretin tetramer. One of these molecules is being tested in phase 2/3 clinical trials for treatment of FAP. Compounds discovered in highthroughput screening tests, such as genistein, a natural compound present in soy products, also inhibit formation of fibrils of wild-type amyloid, as well as amyloidogenesis by the transthyretin variants V30M and V122I, 2 of the most common disease-associated variants. Furthermore, a clinical study in healthy human subjects indicated that kinetic stabilization of transthyretin mediated by orally administered diflunisal, a nonsteroidal anti-inflammatory agent also discovered by screening, should ameliorate transthyretin amyloidosis. The effect of diflunisal on FAP is currently being evaluated in a phase 3 clinical study. Most likely the age-associated nature of neurodegenerative diseases such as the transthyretin amyloidoses can be explained by a shift from efficient to inefficient aggregate clearance, leading to increasing concentrations of the aggregates and proteotoxic effects. We showed that the reassembly of transthyretin homotetramers occurs via a monomer-dimer-trimer-tetramer pathway in which each step depends on the concentration of folded transthyretin monomers. This finding suggests that partitioning of transthyretin monomers between transthyretin tetramer reassembly and the aggregation pathway is correlated with the relative concentrations 82 CHEMISTRY 2006 of reassembly intermediates and aggregates. The concentration of the aggregates presumably increases with aging because of inefficient aggregate clearance. In another study, we found that subunits of the transthyretin variant R104H most likely act as an in vivo trans-suppressor of amyloidogenesis associated with the V30M variant. Compound heterozygotes with genes for the V30M and R104H variants did not show signs of the pathologic changes associated with FAP typical of heterozygotes with genes for the V30M variant and wild-type transthyretin; however, compound heterozygotes with genes for R104H and the aggressive mutation T59K on their second allele did have pathologic FAP effects similar to those of compound heterozygotes with genes for wild-type transthyretin and the T59K mutation. We investigated the energetics of R104H homotetramers and mixed tetramers in a fashion analogous to that used to elucidate the mechanism of T119M transthyretin interallelic trans-suppression. We found that in contrast to T119M, R104H does not suppress aggregation by a kinetic stabilization mechanism. We showed that R104H may trans-suppress transthyretin aggregation by subtle thermodynamic stabilization of the transthyretin quaternary structure. This discovery suggests that R104H could protect compound heterozygotes from transthyretin aggregation in situations in which the mutation is mildly destabilizing. This finding supports the current clinical data associated with R104H in compound heterozygotes. A B E R R A N T O X I D AT I V E M E TA B O L I T E S A F F E C T α- S Y N U C L E I N O PAT H I E S The α-synucleinopathies are characterized by cytoplasmic α-synuclein-rich aggregates within degenerating dopaminergic neurons in the substantia nigra. Clinical observations suggest a correlation between oxidative stress/inflammation and protein misfolding diseases. We wished to determine whether oxidized metabolites accelerate the aggregation of α-synuclein, research that would shed light on the correlation between oxidative stress and sporadic α-synucleinopathies. We found that overexpression of α-synuclein in a neuronal cell line is sufficient to increase the production of oxidative metabolites derived from the oxidation of cholesterol, presumably due to the production of reactive oxygen species. We showed that these oxidative metabolites are cytotoxic and that they significantly accelerate aggregation of α-synuclein in vitro. The experimental data suggest that the acceleration in aggrega- THE SCRIPPS RESEARCH INSTITUTE tion occurs predominantly via a noncovalent mechanism. Overexpression of α-synuclein may lead to the production of reactive oxygen species, which stimulates the production of oxidative cholesterol metabolites that then accelerate α-synuclein aggregation. This acceleration may enhance local oxidative stress, resulting in a vicious cycle that eventually leads or contributes to α-synucleinopathies. Although the exact role of α-synuclein in Lewy body disease and Parkinson’s disease remains to be elucidated, our findings add to an understanding of how aldehyde-based organic compounds formed as a result of aging and inflammation may contribute to neurodegenerative diseases. GELSOLIN AMYLOIDOSIS Gelsolin amyloidogenesis occurs in persons who produce D187N/Y plasma gelsolin variants. This disease is characterized by amyloid deposits composed of 5and 8-kD fragments of plasma gelsolin. The D187N/Y mutation abrogates calcium binding in domain 2, allowing aberrant furin cleavage in the Golgi apparatus during trafficking and yielding a 68-kD fragment. The fragment is then cleaved by the membrane type matrix metalloproteinase 1 (MT1-MMP), resulting in 5- and 8-kD fragments that are deposited as amyloid fibrils in the extracellular matrix. Fibroblasts from animals lacking the gene for MT1-MMP are incapable of generating 8and 5-kD fragments from the 68-kD gelsolin fragment. Biophysical studies indicated that gelsolin amyloid formation is substantially accelerated in the presence of the extracellular matrix component heparin. The extent of sulfation and the location and relative orientation of sulfate residues and the molecular weight are important factors in the heparin-mediated acceleration of gelsolin amyloidogenesis, possibly explaining the heavy deposition of gelsolin amyloid in the extracellular matrix. Most likely tissue-selective deposition of gelsolin amyloid is correlated with the localization of extracellular sulfated glycosaminoglycans, a notion supported by the colocalization of glycosaminoglycans with gelsolin amyloid in the extracellular space of gelsolin amyloidosis transgenic mice. These transgenic mice will be used to evaluate the effect of MT1-MMP inhibitors and glycosaminoglycan amyloid antagonists on gelsolin amyloidosis in vivo. CHEMICAL CHAPERONES AND GAUCHER DISEASE Mutations in glucocerebrosidase, a lysosomal hydrolase, lead to an accumulation of glucosylceramide in the lysosome, causing Gaucher disease, the most common CHEMISTRY 2006 lysosomal storage disorder. Previously, we showed that N-(n-nonyl)deoxynojirimycin increases the activity of the glucocerebrosidase variant N370S in a cell line derived from tissue from a patient with Gaucher disease. This “chemical chaperoning” effect most likely is due to the binding of N-(n-nonyl)deoxynojirimycin to the native state of N370S, allowing the glucocerebrosidase to be trafficked from the endoplasmic reticulum to the lysosomes. Interestingly, the activity of G202R glucocerebrosidase, a variant retained in the endoplasmic reticulum, is also increased in the presence of chemical chaperones, suggesting that those chemical chaperones stimulated transport to the lysosomes. Our results indicate that some chemical chaperones enhance the activity of distinct glucocerebrosidase variants to an extent thought to be sufficient to ameliorate Gaucher disease. Preliminary data suggest that certain glucocerebrosidase mutants most likely will need specifically designed chemical chaperones that target the compromised domain in order to facilitate proper trafficking and partial restoration of the function of the glucocerebrosidase. 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 fibrilization [published correction appears in Nat. Chem. Biol. 2:346, 2006]. Nat. Chem. Biol. 2:249, 2006. THE SCRIPPS RESEARCH INSTITUTE 83 Powers, E.T., Deechongkit, S., Kelly, J.W. Backbone-backbone H-bonds make context-dependent contributions to protein folding kinetics and thermodynamics: lessons from amide-to-ester mutations. Adv. Protein Chem. 72:39, 2005. Premkumar, L., Sawkar, A.R., Boldin-Adamsky, S., Toker, L., Silman, I., Kelly, J.W., Futerman, A.H., Sussman, J.L. X-ray structure of human acid-β-glucosidase covalently bound to conduritol-B-epoxide: implications for Gaucher disease. J. Biol. Chem. 280:23815, 2005. Sawkar, A.R., Adamski-Werner, S.L., Cheng, W.-C., Wong, C.-H., Beutler, E., Zimmer, K.-P., Kelly, J.W. Gaucher disease-associated glucocerebrosidases show mutation-dependent chemical chaperoning profiles. Chem. Biol. 12:1235, 2005. Sawkar, A.R., D’Haeze, W., Kelly, J.W. Therapeutic strategies to ameliorate lysosomal storage disorders: a focus on Gaucher disease. Cell. Mol. Life Sci. 63:1179, 2006. Sekijima, Y., Dendle, M.T., Wiseman, R.L., White, J.T., D’Haeze, W., Kelly, J.W. R104H may suppress transthyretin amyloidogenesis by thermodynamic stabilization, but not by the kinetic mechanism characterizing T119 trans-suppression. Amyloid 13:57, 2006. Sörgjerd, K., Ghafouri, B., Jonsson, B.-H., Kelly, J.W., Blond, S.Y., Hammarström, P. Retention of misfolded mutant transthyretin by the chaperone BiP/GRP78 mitigates amyloidogensis. J. Mol. Biol. 356:469, 2006. Suk, J.Y., Zhang, F., Balch, W.E., Linhardt, R.J., Kelly, J.W. Heparin accelerates gelsolin amyloidogenesis. Biochemistry 45:2234, 2006. Wiseman, R.L., Powers, E.T., Kelly, J.W. Partitioning conformational intermediates between competing refolding and aggregation pathways: insights into transthyretin amyloid disease. Biochemistry 44:16612, 2005. Total Synthesis, New Synthetic Technologies, and Chemical Biology Deechongkit, S., Nguyen, H., Jäger, M., Powers, E.T., Gruebele, M., Kelly, J.W. β-Sheet folding mechanisms from perturbation energetics. Curr. Opin. Struct. Biol. 16:94, 2006. K.C. Nicolaou, S. Arseniyadis, W. Brenzovich, P. Bulger, Foss, T.R., Wiseman, R.L., Kelly, J.W. The pathway by which the tetrameric protein transthyretin dissociates. Biochemistry 44:15525, 2005. P. Dagneau, R. Denton, A. Estrada, D. Edmonds, C. Fang, Fowler, D.M., Koulov, A.V., Alory-Jost, C., Marks, M.S., Balch, W.E., Kelly, J.W. Functional amyloid formation within mammalian tissue. PLoS Biol. 4:e6, 2006. V. Jeso, A. Johnson, D. Kim, A. Kislukhin, A. Lanver, K. Lee, Fu, Y., Bieschke, J., Kelly, J.W. E-Olefin dipeptide isostere incorporation into a polypeptide backbone enables hydrogen bond perturbation: probing the requirements for Alzheimer’s amyloidogenesis. J. Am. Chem. Soc. 127:15366, 2005. D. Lizos, E. Loizidou, N. Mainolfi, C. Mathison, X. Mico-Alvarez, Green, N.S., Foss, T.R., Kelly, J.W. Genistein, a natural product from soy, is a potent inhibitor of transthyretin amyloidosis. Proc. Natl. Acad. Sci. U. S. A. 102:14545, 2005. Johnson, S.M., Wiseman, R.L., Sekijima, Y., Green, N.S., Adamski-Werner, S.L., Kelly, J.W. Native state kinetic stabilization as a strategy to ameliorate protein misfolding diseases: a focus on the transthyretin amyloidoses. Acc. Chem. Res. 38:911, 2005. Kelly, J.W., Balch, W.E. The integration of cell and chemical biology in protein folding. Nat. Chem. Biol. 2:224, 2006. Nguyen, H., Jäger, M., Kelly, J.W., Gruebele, M. Engineering a β-sheet protein toward the folding speed limit. J. Phys. Chem. B Condens. Matter Mater. Surf. Interfaces Biophys. 109:15182, 2005. Page, L.J., Suk, J.Y., Huff, M.E., Lim, H.-J., Venable, J., Yates, J. III, Kelly, J.W., Balch, W.E. Metalloendoprotease cleavage triggers gelsolin amyloidogenesis. EMBO J. 24:4124, 2005. A. Burtoloso, J. Chen, K. Cole, A. Converso, J. Crawford, R. Faraoni, M. Frederick, M. Freestone, R. Gibe, S. Harrison, S. Lee, A. Lemire, A. Lenzen, A. Li, H. Li, Y. Lim, T. Lister, R. Milburn, A. Nold, A. Noncovich, R. de Noronha, A. Ortiz, C. Papageorgiou, D. Pappo, L. Pasunoori, G. Petrovic, J. Piper, D. Polet, G. Pontremoli, B. Pratt, D. Sarlah, D. Shaw, C. Stathakis, C. Solorio-Alvarado, T. Suzuki, G. Tria, C. Turner, T. Umezawa, J. Wang, H. Xu, M. Zak e focus on the total synthesis of natural products, the discovery and development of new synthetic technologies, and chemical biology. Naturally occurring substances are selected as synthetic targets because of their novel molecular architectures, important biological properties, and interesting mechanisms of action. The projects are designed to optimize the opportunities for discovery and invention in the areas of chemistry, biology, and medicine. The drug pacli- W 84 CHEMISTRY 2006 taxel (Taxol), the antitumor epothilones, the neurotoxins brevetoxins A and B, the antibiotic vancomycin, the cholesterol-lowering CP-molecules, the antibiotic everninomicin, the TNF α–associated trichodimerol, the tetrahydropyran class of natural products, apoptolidin, diazonamide A, thiostrepton, and the azaspiracids exemplify this philosophy. Current projects include studies of the antibiotics nocathiacin and abyssomicin C, the antifeedant azadirachtin, and the antitumor agents marinomycin and A lomaiviticin B (Fig. 1). THE SCRIPPS RESEARCH INSTITUTE Nicolaou, K.C. Joys of molecules, 1: campaigns in total synthesis. J. Org. Chem. 70:7007, 2005. Nicolaou, K.C. Joys of molecules, 2: endeavors in chemical biology and medicinal chemistry. J. Med. Chem. 48:5613, 2005. Nicolaou, K.C., Brenzovich, W.E., Bulger, P.G., Francis, T.M. Synthesis of isoepoxy-amphidinolide N and des-epoxy-caribenolide I structures: initial forays. Org. Biomol. Chem. 4:2119, 2006 Nicolaou, K.C., Bulger, P.G., Brenzovich, W.E. Synthesis of iso-epoxy-amphidinolide N and des-epoxy-caribenolide I structures: revised strategy and final stages. Org. Biomol. Chem. 4:2158, 2006. Nicolaou, K.C., Chen, D.Y.-K., Li, Y., Uesaka, N., Petrovic, G., Koftis, T.V., Bernal, F., Frederick, M.O., Govindasamy, M., Ling, T., Pihko, P.M., Tang, W., Vyskocil, S. Total synthesis and structural elucidation of azaspiracid-1: synthesisbased analysis of originally proposed structures and indication of their non-identity to the natural product. J. Am. Chem. Soc. 128:2258, 2006. Nicolaou, K.C., Denton, R.M., Lenzen, A., Li, A., Edmonds, D.J., Milburn, R.R., Harrison, S.T. Stereocontrolled synthesis of model core systems of lomaiviticins A and B. Angew. Chem. Int. Ed. 45:2076, 2006. Nicolaou, K.C., Frederick, M.O., Loizidou, E.Z., Petrovic, G., Cole, K.P., Koftis, T.V., Yamada, Y.M.A. Second-generation total synthesis of azaspiracids-1, -2, and 3. Chem. Asian J. 1:245, 2006. Nicolaou, K.C., Frederick, M.O., Petrovic, G., Cole, K.P., Loizidou, E. Total synthesis and confirmation of the revised structures of azaspiracid-2 and azaspiracid3. Angew. Chem. Int. Ed. 45:2609, 2006. Nicolaou, K.C., Harrison, S.T. Total synthesis of abyssomicin C and atrop-abyssomicin C. Angew. Chem. Int. Ed. 45:3256, 2006. Nicolaou, K.C., Kim, D.W., Schlawe, D., Lizos, D.E., de Noronha, R.G., Longbottom, D.A. Total synthesis of halipeptins A and D and analogues. Angew. Chem. Int. Ed. 44:4925, 2005. Nicolaou, K.C., Koftis, T.V., Vyskocil, S., Petrovic, G., Tang, W., Frederick, M.O., Chen, D.Y.-K., Li, Y., Ling, T., Yamada, Y.M.A. Total synthesis and structural elucidation of azaspiracid-1: final assignment and total synthesis of the correct structure of azaspiracid-1. J. Am. Chem. Soc. 128:2859, 2006. Nicolaou, K.C., Lim, Y.H., Papageorgiou, C.D., Piper, J.L. Total synthesis of (+)rugulosin and (+)-2,2′-epi-cytoskyrin A through cascade reactions. Angew. Chem. Int. Ed. 44:7917, 2005. Nicolaou, K.C., Lizos, D.E., Kim, D.W., Schlawe, D., de Noronha, R.G., Longbottom, D.A., Rodriquez, M., Bucci, M., Cirino, G. Total synthesis and biological evaluation of halipeptins A and D and analogues. J. Am. Chem. Soc. 128:4460, 2006. Nicolaou, K.C., Mathison, C.J.N. Synthesis of imides, N-acyl vinylogous carbamates and ureas, and nitriles with Dess-Martin periodinane. Angew. Chem. Int. Ed. 44:5992, 2005. Nicolaou, K.C., Papageorgiou, C.D., Piper, J.L., Chadha, R.K. The cytoskyrin cascade: a facile entry into cytoskyrin A, deoxyrubroskyrin, rugulin, skyrin and flavoskyrin model systems. Angew. Chem. Int. Ed. 44:5846, 2005. F i g . 1 . Selected target molecules. In addition, we are developing synthetic technologies and strategies for chemical synthesis and chemical biology studies. Our overall aims are to advance the art and science of chemical synthesis and to develop enabling technologies for biology and medicine while maximizing educational opportunities and training of young men and women in chemistry. PUBLICATIONS Ito, E., Frederick, M.O., Koftis, T.V., Tang, W., Petrovic, G., Ling, T., Nicolaou, K.C. Structure toxicity relationships of synthetic azaspiracid-1 and analogs in mice. Harmful Algae. 5:586, 2006. Nicolaou, K.C., Pihko, P.M., Bernal, F., Frederick, M.O., Qian, W., Uesaka, N., Diedrichs, N., Hinrichs, J., Koftis, T.V., Loizidou, E., Petrovic, G., Rodriquez, M., Sarlah, D., Zou, N. Total synthesis and structural elucidation of azaspiracid-1: construction of key building blocks for originally proposed structure. J. Am. Chem. Soc. 128:2244, 2006. Nicolaou, K.C., Pratt, B.A., Arseniyadis, S., Wartmann, M., O’Brate, A., Giannakakou, P. Molecular design and chemical synthesis of a highly potent epothilone. Chemmedchem 1:41, 2006. Nicolaou, K.C., Safina, B.S., Zak, M., Lee, S.H., Nevalainen, M., Bella, M., Estrada, A.A., Funke, C., Zécri, F., Bulat, S. Total synthesis of thiostrepton: retrosynthetic analysis and construction of key building blocks. J. Am. Chem. Soc. 127:11159, 2005. Nicolaou, K.C., Schlawe, D., Kim, D.W., Longbottom, D.A., de Noronha, R.G., Lizos, D.E., Manan, R.R., Faulkner, D.J. Total synthesis of halipeptins: isolation of halipeptin D and synthesis of oxazoline halipeptin analogues. Chemistry 11:6197, 2005. CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE Nicolaou, K.C., Xu, H. Total synthesis of floresolide B and ∆6,7-Z-floresolide B. Chem. Commun. (Camb.) 600, 2006, Issue 6. DUAL OPIOID AGONISTS–CHOLECYSTOKININ Nicolaou, K.C., Zak, M., Rahimipour, S., Estrada, A.A., Lee, S.H., O’Brate, A., Giannankakou, P., Ghadiri, M.R. Discovery of a biologically active thiostrepton fragment. J. Am. Chem. Soc. 127:15042, 2005. N E U R O PAT H I C PA I N Nicolaou, K.C., Zak, M., Safina, B.S., Estrada, A.A., Lee, S.H., Nevalainen, M. Total synthesis of thiostrepton: assembly of key building blocks and completion of the synthesis. J. Am. Chem. Soc. 127:11176, 2005. Translational Chemistry and Medicine E. Roberts, Z.Y. Chen, O. Ghoneim, C. Martinez, S. Sinha, C. Zoni iscovery and development of new medicines require the integration of several scientific disciplines. For example, medicinal chemistry relies on iterative in vitro and in vivo biological testing of molecules. These biological investigations, in turn, rely on the design and synthesis of new molecules with therapeutic potential to delineate and validate pathways for therapeutic intervention. The primary focus of our research is to identify potential new medicines for the treatment of diseases that currently have inadequate or no current therapy. D G A L A N I N L I G A N D S F O R T H E T R E AT M E N T O F NEUROLOGIC DISEASES Epilepsy is a disease in which a hyperexcited state of the CNS is caused by an imbalance between inhibitory and excitatory neurotransmission. Current epilepsy therapy focuses on modulating the classical neurotransmitters glutamate and γ-aminobutyric acid. The neuropeptide galanin antagonizes excitatory glutaminergic neurotransmission in the hippocampus, suggesting that galanin may have a role in seizure activity. Galanin and its receptors may be useful in developing novel antidepressant pharmacotherapies. In a recent study in humans with depression, intravenously administered galanin had a rapid and strong antidepressant effect. To identify new nonpeptidic ligands for galanin receptors, we are using a small set of known molecules that can potently displace the peptide galanin from its binding site on the R3 receptor subtype. Selectivity and potency can then be established by appending appropriate accessory binding motifs selective for the desired protein. This research is being done in collaboration with T. Bartfai, Molecular and Integrative Neurosciences Department, and A.M. Mazarati, University of California, Los Angeles. 85 A N TA G O N I S T S F O R T R E AT M E N T O F C H R O N I C A N D Nociception, or the perception of pain, and its modulation depend on the interaction of many endogenous neurotransmitters in the spinal cord. The interaction of endogenous peptides such as cholecystokinin with exogenously administered opioids markedly alters activity in acute and chronic pain states. Cholecystokinin antagonizes the analgesic effects of morphine, whereas cholecystokinin antagonists enhance them. The interplay between cholecystokinin antagonists and opiates may lead to the development of novel medications that are more effective and safer than currently used opioids alone. Molecules with both opioid agonist and cholecystokinin antagonist properties would be useful in conditions in which the effectiveness of opioids is reduced, as in the development of tolerance to opiate pain relievers in chronic pain associated with cancer. The molecules might also be useful in neuropathic pain conditions in which opioids are ineffective. Thus, because of the prevention (or reversal) of tolerance, the possibility of physical dependence on opioids might be diminished or inhibited. The advantages of developing a single compound with dual opioid agonist–cholecystokinin antagonist activity rather than a combination of an opioid agonist taken with a separate cholecystokinin antagonist are clear. Development of a single compound involves only a single set of parameters, such as toxicology, pharmacokinetics, and formulation, rather than 2 independent and often unrelated sets of data. In collaboration with F. Porreca and J. Lai, University of Arizona, Tucson, we are using a limited set of molecular templates that have affinity across a wide range of type 1 G protein–coupled receptors. The 3 cloned opiate receptors (µ, δ, and κ) and the cholecystokinin 1/(A) and 2/(B) receptors are all members of this subclass of G protein–coupled receptors. Ligands are known for both sets of receptors that contain the diphenylmethyl moiety as a privileged or biased template. Other critical elements for activity may be appended to chemically “silent” sites (Fig. 1). NEUROPHARMACOLOGIC APPROACHES FOR P R E V E N T I O N A N D T R E AT M E N T O F A U T I S M Autism is a bioneurologic developmental disability that generally appears before the age of 3 years. This disability affects the normal development of the brain in the areas of social interaction, communication skills, 86 CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE F i g . 3 . A, Known mixed vasopressin V1a/V2 receptor antagoF i g . 1 . Proposed opioid agonist–cholecystokinin antagonist hybrids. and cognitive function. The prevalence of autistic diseases has reached pandemic proportions. In the United States, autism occurs in an estimated 1 in 166 births, and roughly 1.5 million persons have some form of autism. This rate is increasing significantly and now surpasses that of all types of cancer combined. No drug is consistently effective in treating the signs and symptoms of autism. Vasopressin and oxytocin are 2 nonapeptides (Fig. 2) secreted by the hypothalamus. nists. B, Known vasopressin V1a receptor antagonists. F i g . 4 . Known oxytocin receptor agonists. and A. Roberts, Molecular and Integrative Neurosciences Department. PUBLICATIONS Roberts, E., Sancon, J.P., Sweeney, J.B. A new class of ammonium ylid for [2,3]sigmatropic rearrangement reactions: end-endo-spiro ylids. Org. Lett. 7:2075, 2005. F i g . 2 . Structures of vasopressin and oxytocin. They differ in only 2 amino acids, and both exert their effects at protein receptors that belong to the G protein–coupled receptor superfamily, namely vasopressin V1a, V1b, and V2 receptors and oxytocin receptors. When oxytocin and vasopressin exert their agonist effects at the oxytocin and vasopressin V1a receptors, respectively, in rodents, marked effects occur in the CNS. These effects include behaviors associated with autism, such as social behaviors (e.g., bonding, aggression); cognition (e.g., memory and active-passive avoidance); and repetitive, patterned movements (e.g., grooming or social interaction). Antagonists reverse or have no effect on the observed behaviors. We are designing and developing small-molecule oxytocin agonists that can penetrate the CNS and previously unknown vasopressin V1a receptor agonists to establish the potential roles for these molecules in the treatment of autism (Figs. 3 and 4). This research is being done in collaboration with T. Bartfai, G.F. Koob, Workman, J.A., Garrido, N.P., Sancon, J., Roberts, E., Wessel, H.P., Sweeney, J.B. Asymmetiric [2,3]-rearrangement of glycine-derived allyl ammonium ylids. J. Am. Chem. Soc. 127:1066, 2005. Chemical, Biological, and Biophysical Approaches to Understanding Evolution F.E. Romesberg, D.A. Bachovchin, J.K. Chin, R.T. Cirz, M.E. Cremeens, C. Gil-Lamaignere, N. Gingles, D.A. Harris, A.A. Henry, G.T. Hwang, A.M. Leconte, E.T. Lis, S. Matsuda, E.L. Oakman, B.A. O’Neill, T.C. Roberts, L.B. Sagle, P.A. Smith, M.C. Thielges, P. Weinkam, W. Yu, J. Zimmermann he molecules of biology are unique because they have been evolved for function. We take a unique and multidisciplinary approach to understanding and manipulating these processes. T CHEMISTRY 2006 INCREASING THE CHEMICAL AND GENETIC POTENTIAL OF DNA Biological information storage is based on the natural genetic alphabet, composed of the 2 base pairs guanine-cytosine and adenine-thymine. We are interested in increasing the information potential of DNA by expanding the genetic alphabet with a third base pair composed of unnatural nucleobases. Using hydrophobicity, polarity, shape complementarity, and hydrogen bonding, we are developing novel unnatural base pairs, including several that are replicable in vitro. Nature developed the natural genetic code, not only by optimizing DNA and RNA but also by evolving the polymerases that synthesize these nucleic acids. We developed an activity-based selection system (Fig. 1) to THE SCRIPPS RESEARCH INSTITUTE 87 malian cells will revolutionize our understanding of cancer and aging and identify drug targets whose inhibition might actually inhibit these processes. In bacteria, induced mutation can lead to antibiotic resistance. We have fully characterized the mechanism of induced mutation and antibiotic resistance in Escherichia coli (Fig. 2), and we are characterizing F i g . 2 . Induced mutation in E coli is controlled by the transcrip- tional regulator LexA. Duplex DNA is shown in black; each gray box indicates a double-strand break. The breaks can be repaired through pathway A, B, or C. Derepression of the error-prone polymerases Pol IV and Pol V leads to mutations (pathway D). these pathways in other bacterial pathogens. We are also designing a drug that inhibits bacterial mutation and thus evolution. F i g . 1 . Activity-based phage display selection system for evolv- EVOLUTION OF PROTEIN DYNAMICS ing polymerases with novel activity. Infection of phage (B) with the polymerase library (A) leads to production of phage particles that The products of evolution are molecules with unique vibrational dynamics. The study of vibrational dynamics in proteins and nucleic acids has been limited by spectral complexity, but selective deuteration of a protein or a nucleic acid results in a carbon-deuterium oscillator that absorbs light in an otherwise transparent region of the infrared spectrum. The synthesis of selectively deuterated proteins has provided us with a residue-specific probe of flexibility, function, and folding. We are also using multidimensional femtosecond spectroscopy to characterize how protein motion is evolved during the somatic evolution of antibodies. We discovered that the immune system can manipulating protein dynamics, a finding that suggests a role for these dynamics in molecular recognition. display 0–1 copies of the polymerase and 3–5 copies of the acidic peptide. Phage particles are combined with DNA primer–template (C) and incubated with the desired nucleoside triphosphates. Active mutants are isolated (D) and characterized. evolve polymerases for any desired function. Using this system, we have already evolved polymerases with a variety of novel functions, including the synthesis of DNA containing one of the unnatural base pairs. We are optimizing these polymerases and evolving new ones. DNA DAMAGE RESPONSE Evolution requires mutation, but mutations also make cells susceptible to aging and cancer. It is now understood that at times of sufficient stress, cells induce error-prone replication to facilitate their own evolution. We used genome-wide high-throughput methods to identify genes involved in both error-free and error-prone responses to DNA stress in budding yeast. Characterization of the proteins required for mutation in mam- PUBLICATIONS Chin, J.K., Bashkirov, V.I., Heyer, W.D., Romesberg, F.E. Esc4/Rtt107 and the control of recombination during replication. DNA Repair (Amst.) 5:618, 2006. Cirz, R.T., Gingles, N., Romesberg, F.E. Side effects may include evolution. Nat. Med. 8:890, 2006. 88 CHEMISTRY 2006 Cirz, R.T., O’Neill, B.M., Hammond, J.A., Head, S.R., Romesberg, F.E. Defining the Pseudomonas aeruginosa SOS response and its role in the global response to the antibiotic ciprofloxacin. J. Bacteriol. 188:7101, 2006. Cirz, R.T., Romesberg, F.E. Induction and inhibition of ciprofloxacin resistance-conferring mutations in hypermutator bacteria. Antimicrob. Agents Chemother. 50:220, 2006. Cremeens, M., Fujisaki, H., Zhang, Y., Zimmermann, J., Sagle, L.B., Matsuda, S., Dawson, P.E., Straub, J.E., Romesberg, F.E. Efforts toward developing direct probes of protein dynamics. J. Am. Chem. Soc. 128:6028, 2006. 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. Henry, A.A., Romesberg, F.E. Evolution of DNA polymerases with novel activities. Curr. Opin. Biotechnol. 16:370, 2005. Hwang, G.T., Romesberg, F.E. Substituent effects on the pairing and polymerase recognition of simple unnatural base pairs. Nucleic Acids Res. 34:2037, 2006. Kim, Y., Leconte, A.M., Hari, Y., Romesberg, F.E. Stability and polymerase recognition of pyridine nucleobase analogues: role of minor-groove H-bond acceptors. Angew. Chem. Int. Ed., in press. Leconte, A.M., Chen, L., Romesberg, F.E. Polymerase evolution: efforts toward expansion of the genetic code. J. Am. Chem. Soc. 127:12470, 2005. Leconte, A.M., Matsuda, S., Hwang, G., Romesberg, F.E. Efforts towards expansion of the genetic alphabet: pyridone and methyl pyridone nucleobases. Angew. Chem. Int. Ed. 45:4326, 2006. Leconte, A.M., Matsuda, S., Romesberg, F.E. An efficiently extended class of unnatural base pairs. J. Am. Chem. Soc. 128:6780, 2006. Leconte, A.M., Romesberg, F.E. Amplify this! DNA and RNA get a third base pair. Nat. Methods 3:667, 2006. Lis, E.T., Romesberg, F.E. Role of Doa1 in the Saccharomyces cerevisiae DNA damage response. Mol. Cell. Biol. 26:4122, 2006. Matsuda, S., Henry, A.A., Romesberg, F.E. Optimization of unnatural base pair packing for polymerase recognition. J. Am. Chem. Soc. 128:6369, 2006. Sagle, L.B., Zimmermann, J., Dawson, P.E., Romesberg, F.E. Direct and high-resolution characterization of cytochrome c equilibrium folding. J. Am. Chem. Soc., in press. Sagle, L.B., Zimmermann, J., Matsuda, S., Dawson, P.E., Romesberg, F.E. Redoxcoupled dynamics and folding in cytochrome c. J. Am. Chem. Soc. 128:7909, 2006. Zimmermann, J., Oakman, E.L., Thorpe, I.F., Shi, X., Abbyad, P., Brooks, C.L. III, Boxer, S.G., Romesberg, F.E. Antibody evolution constrains conformational hetereogeneity by tailoring protein dynamics. Proc. Natl. Acad. Sci. U. S. A. 103:13722, 2006. Biological Chemistry P.G. Schultz, A. Boitano, E. Brustad, M. Bushey, S. Chen, J. Guo, J. Grbic, D. Groff, J. Grünewald, W.Y. Hur, M. Jahnz, T. Kuo, J. Lee, J.-S. Lee, K.-B. Lee, E. Lemke, C. Liu, W. Liu, C. Lyssiotis, J. Mills, M. Mukherji, T. Nom, B. Okram, R. Perera, F. Peters, Y. Ryu, S. Schiller, M. Sever, D. Summerer, L. Supekova, E. Tippmann, M.-L. Tsao, J. Wang, T. Young, Q. Zhang, S. Zhu A lthough chemists are remarkably adept at synthesizing molecular structures, they are far less sophisticated in designing and synthesizing THE SCRIPPS RESEARCH INSTITUTE molecules with defined biological or chemical functions. Nature, on the other hand, has produced an array of molecules with remarkably complex functions, ranging from photosynthesis and signal transduction to molecular recognition and catalysis. Our aim is to combine the synthetic strategies and biological processes of Nature with the tools and principles of chemistry to create new molecules with novel chemical and biological functions. By studying the properties of the resulting molecules, we can gain new insights into the molecular mechanisms of complex biological and chemical systems. For example, we have shown that the tremendous combinatorial diversity of the immune response can be chemically reprogrammed to generate selective enzymelike catalysts. We have developed antibodies that catalyze a wide array of chemical and biological reactions, from acyl transfer to redox reactions. Characterization of the structure and mechanisms of these catalytic antibodies has led to important new insights into the mechanisms of biological catalysis. In addition, the detailed characterization of the properties and structures of germ-line and affinity-matured antibodies has revealed fundamental new aspects of the evolution of binding and catalytic function, in particular, the role of structural plasticity in the immune response. Most recently, we have focused on in vitro evolution methods that involve the development of novel chemical screens and selections for identifying metalloantibodies with proteolytic activity. Our work on catalytic antibodies redirects natural combinatorial diversity to produce new function. We are extending this combinatorial approach to many other problems, including the generation of genetic “microcalorimetes” and yeasts lacking mitochondrial genomes and the ab initio evolution of novel protein domains. We are also generating structure-based combinatorial libraries of small heterocycles that are being used in conjunction with novel cellular and organismal screens to identify important proteins involved in such cellular processes as differentiation, proliferation, and signaling. Indeed, we have identified molecules that control both adult and embryonic stem cell differentiation and self-renewal and that reprogram lineage-committed cells. We are using x-ray crystallographic and biochemical studies, together with genomics technologies, to characterize the mode of action of these compounds and to study their effects on cellular processes. We are also applying genomics tools (cell-based phenotypic screens of arrayed genomic cDNA and small CHEMISTRY 2006 interfering RNA libraries) and proteomics tools (mass spectrometric phosphoprotein profiling) to a variety of important biomedical problems in cancer biology, neurodegenerative and autoimmune diseases, and virology. In addition, we are investigating the role and regulation of noncoding RNAs. We have also developed a general biosynthetic method that can be used to site specifically incorporate unnatural amino acids into proteins in vitro and in vivo. Using this method, we effectively expanded the genetic codes of both prokaryotic and eukaryotic organisms by adding new components to the biosynthetic machinery of living cells. We have genetically encoded amino acids with novel spectroscopic and chemical properties (e.g., metal-binding, glycosylated, and fluorescent amino acids and photocross-linking and photoisomerizable amino acids) in response to unique 3- and 4-base codons. These amino acids are being used to explore protein structure and function both in vitro and in vivo and to evolve proteins with novel properties. Our results have removed a billion-year constraint imposed by the genetic code on the ability to chemically manipulate the structures of proteins. PUBLICATIONS Bose, M., Groff, D., Xie, J., Brustad, E., Schultz, P.G. The incorporation of a photoisomerizable amino acid into proteins in E. coli. J. Am. Chem. Soc. 128:388, 2006. Chen, S., Do, J., Zhang, Q., Yao, S., Yan, F., Peters, E., Scholer, H., Schultz, P.G., Ding, S. Self-renewal of embryonic stem cells by a small molecule. Proc. Natl. Acad. Sci. U. S. A., in press. Mukherji, M., Cho, C., Supekova, L., Wang, Y., Batalov, S., Bell, R., Martin, C., Sahasrabuhde, S., Orth, A.P., Chanda, S.K., Schultz, P.G. Functional analysis of human genome for cell-cycle regulators. Proc. Natl. Acad. Sci. U. S. A., in press. Summerer, D., Chen, S., Wu, N., Deiters, A., Chin, J.W., Schultz, P.G. A genetically encoded fluorescent amino acid. Proc. Natl. Acad. Sci. U. S. A. 103:9785, 2006. Turner, J.M., Graziano, J., Spraggon, G., Schultz, P.G. Structural characterization of a p-acetylphenylalanyl aminoacyl-tRNA synthetase. J. Am. Chem. Soc. 127:14976, 2005. Warashina, M., Min, K.H., Kuwabara, T., Huynh, A., Gage, F.H., Schultz, P.G., Ding, S. A synthetic small molecule that induces neuronal differentiation of adult hippocampal neural progenitor cells. Angew. Chem. Int. Ed. 45:591, 2006. Willingham, A.T., Orth, A.P., Batalov, S., Peters, E.C., Wen, B.G., Aza-Blanc, P., Hogenesch, J.B., Schultz, P.G. A strategy for probing the function of noncoding RNAs finds a repressor of NFAT. Science 309:1570, 2005. THE SCRIPPS RESEARCH INSTITUTE 89 Powerful Click Processes for Organic Synthesis, Chemical Biology, and Materials Research K.B. Sharpless, V.V. Fokin, M. Ahlquist, B. Boren, M. Cassidy, S. Chang, A. Feldman, J. Fotsing, R. Fraser, A. Grant, J. Hein, J. Kalisiak, L. Krasnova, S.-W. Kwok, Y. Liu, J. Loren, R. Manetsch, K. Nagai, S. Narayan, S. Pitram, L.K. Rasmussen, J. Raushel, S. Roeper, W. Sharpless, A. Sugawara, J. Tripp, X. Wang, J. Wassenaar, T. Weide, M. Whiting, P. Wu he driving forces in our research are the discovery and understanding of chemical reactivity, the harbingers of all new reactions. Our main goal is to develop practical transformations that facilitate synthesis of novel molecules with desired functions and allow manipulation of complex biological systems at the molecular level. T CLICK CHEMISTRY Among the many factors that determine the success of a search for compounds with desired properties, 2 stand out: the degree of diversity of the building blocks that can be used and the speed with which synthesis, screening for the desired function, and lead optimization can be performed. The greater the variety of scaffolds and functional groups that can be used in the rapid construction of candidate compounds, the more likely it is that new and useful function will be discovered. Because of the enormous number of compounds to explore (the number of small druglike compounds may be as high as 1064), the size of a given collection becomes much less important than the ability to rapidly probe the collection for a desired activity. However, many chemical methods often have restrictions such as limited scope, inaccessibility of starting materials, requirements for protecting groups, and difficult purifications. In addition, inert atmospheres and anhydrous solvents are usually required, a situation that makes these methods impractical for manipulating biological molecules in the molecules’ natural, aqueous environment. In the past several years, we have sought to develop and use only the best reactions for the synthesis of functional molecules. The reactions that fulfill the most stringent criteria of usefulness and convenience have been grouped under the name click chemistry. Most click reactions form carbon-heteroatom bonds, are tolerant of water, and are often accelerated when water 90 CHEMISTRY 2006 is used as the sole medium, even if the reagents are not soluble in water. C O P P E R - C ATA LY Z E D C Y C L O A D D I T I O N S The copper-catalyzed 1,3-dipolar cycloaddition of azides and alkynes has emerged as a premiere click reaction that enables reliable assembly of complex molecules by means of the 1,2,3-triazole heterocycle. Although both alkynes and azides are highly reactive, their reactivity profiles are quite narrow, that is, “orthogonal” to an unusually broad range of reagents, solvents, and other functional groups. These features allow clean sequential transformations of broad scope without the need for protecting groups, even if the reactions are performed under physiologic conditions. The 1,2,3-triazoles have the advantageous properties of high chemical stability (in general, being inert to severe hydrolytic, oxidizing, and reducing conditions, even at high temperatures), strong dipole moment, presence of aromatic groups, and the ability to accept hydrogen bonds. Thus, they can interact productively in several ways with biological molecules. For example, 1,2,3-triazoles can replace the amide bond in peptides, preventing proteolytic degradation of the peptides. The fundamental thermal reaction, involving terminal or internal alkynes (Fig. 1, top), has been known F i g . 1 . Thermal cycloaddition of azides and alkynes (top) requires prolonged heating and results in mixtures of both 1,4- and 1,5-regioisomers, whereas the copper-catalyzed 1,3-dipolar cycloaddition of azides and alkynes produces only 1,4-disubstituted-1,2,3-triazoles at room temperature (bottom). for more than a century and has been thoroughly investigated. Although the process is strongly thermodynamically favored, it has a relatively high kinetic barrier that makes the reaction slow at room temperature for unactivated reactants and results in the formation of regioisomers. Copper(I) catalysis dramatically accelerates the reaction, by a factor of up to 107, and regiospecifically produces only 1,4-disubstituted-1,2,3-triazoles (Fig. 1, bottom). Because of its experimental simplicity and unusually broad scope, this process has been used in a number of applications in synthesis, medicinal chemistry, molecular biology, and materials science. THE SCRIPPS RESEARCH INSTITUTE Although a number of copper(I) complexes can be used to catalyze the reaction, we found that the catalyst is often better prepared in situ by reduction of copper(II) salts, which are readily available and are easier to handle than most copper(I) salts. As the reductant, ascorbic acid (vitamin C) or sodium ascorbate is excellent. Remarkably, even copper metal can be used as a source of the catalytic species, making the experimental procedure even simpler: a small piece of copper metal (wire or turning) is all that is added to the reaction mixture, which is then shaken or stirred for 12–48 hours. This protocol is particularly convenient in parallel synthesis, because triazole products are generally isolated in high yields and can often be submitted for screening without further purification. Our studies of reactivity of sulfonyl azides in the copper-catalyzed cycloaddition of azides and alkynes resulted in the development of an experimentally simple catalytic procedure for the highly selective conversion of alkynes to N-sulfonyl azetidin-2-imines under mild conditions. This 3-component process is thought to proceed via initial reaction of in situ generated copper(I) acetylides with sulfonyl azides, resulting in transient (1-sulfonyltriazolyl) copper intermediates that upon extrusion of dinitrogen generate N-sulfonyl keteneimines. The azetidinimine products are remarkably stable in a wide range of reaction conditions, and other functional groups can be easily added (Fig. 2). This newly dis- F i g 2 . Synthesis of azetidinimines from alkynes, sulfonyl azides, and imines. covered reaction sequence rapidly produces densely functionalized azetidine derivatives from readily available terminal alkynes in just 2 or 3 simple steps and should be useful for exploring the usefulness of these 4-atom heterocycles. Although useful because of its rate and functional group tolerance, the copper-catalyzed 1,3-dipolar cycloaddition of azides and alkynes cannot produce 1,5-disubstituted 1,2,3-triazoles, and it is not effective with internal alkynes. Therefore, the recent discovery of ruthenium(II) catalysts that are active in azide-alkyne cycloaddition and result in the formation of the complementary 1,5-regioisomers of 1,2,3-tria- CHEMISTRY 2006 zoles was a welcome advance. Pentamethyl cyclopentadienyl ruthenium(II) complexes are active and easy to handle and provide triazole products in good to excellent yields (Fig. 3). In addition, these catalysts are active F i g . 3 . Ruthenium-catalyzed synthesis of fully substituted 1,2,3-triazoles. with internal alkynes, allowing easy synthesis of fully substituted triazoles. S Y N T H E S I S O F P O LY F U N C T I O N A L D E N D R I M E R S Dendrimers are highly ordered, regularly branched globular macromolecules of defined structure; dendrimers are ideal building blocks for creating bioactive macromolecules and nanomaterials. Previously, we exploited the high fidelity of the copper-catalyzed 1,3 dipolar cycloaddition of azides and alkynes in the efficient synthesis of dendrimers. We used procedures that involved little more than mixing stoichiometric quantities of reactants, stirring, and isolating dendrimer products. In collaboration with M.G. Finn, Department of Chemistry, and C.J. Hawker, University of California, Santa Barbara, we have extended this approach to synthesize chemically heterogeneous dendrimers (Fig. 4) THE SCRIPPS RESEARCH INSTITUTE 91 binding protein concanvalin A and rabbit red blood cells. The dendrimer had 240-fold greater potency than monomeric mannose, a difference that translates to a 15-fold increase in activity per unit. Additionally, our preliminary experiments indicate that this dendrimer binds to the modified surface of Escherichia coli, producing detectable fluorescent signals. These results are a significant advance in dendrimer chemistry and illustrate an evolving synergy between organic chemistry and functional materials. Studies of other applications, ranging from biology to materials science, are currently underway in our laboratories and in collaboration with M.G. Finn, P.K. Vogt, C.-H. Wong, J.H. Elder, and others at Scripps Research. PUBLICATIONS Bourne, Y., Radic, Z., Kolb, H.C., Sharpless, K.B., Taylor, P., Marchot, P. Structural insights into conformational flexibility at the peripheral site and within the active center gorge of AChE. Chem. Biol. Interact. 157-158:159, 2005. Cassidy, M.P., Raushel, J., Fokin, V.V. Practical synthesis of amides from in situ generated copper(I) acetylides and sulfonyl azides. Angew. Chem. Int. Ed. 45:3154, 2006. Hansen, T.V., Wu, P., Fokin, V.V. One-pot copper(I)-catalyzed synthesis of 3,5-disubstituted isoxazoles. J. Org. Chem. 70:7761, 2005. Loren, J.C., Krasinski, A., Fokin, V.V., Sharpless, K.B., NH-1,2,3-triazoles from azidomethyl pivalate and carbamates: base-labile N-protecting groups. Synlett 2847, 2005, Issue 18. Malkoch, M., Schleicher, K., Drockenmuller, E., Hawker, C.J., Russell, T.P., Wu, P., Fokin, V.V. Structurally diverse dendritic libraries: a highly efficient functionalization approach using click chemistry. Macromolecules 38:3663, 2005. Meng, J.-C., Fokin, V.V., Finn, M.G. Kinetic resolution by copper-catalyzed azidealkyne cycloaddition. Tetrahedron Lett. 46:4543, 2005. Petasis, N.A., Akritopoulou-Zanze, I., Fokin, V.V., Bernasconi, G., Keledjian, R., Yang, R., Uddin, J., Nagulapalli, K.C., Serhan, C.N. Design, synthesis and bioactions of novel stable mimetics of lipoxins and aspirin-triggered lipoxins. Prostaglandins Leukot. Essent. Fatty Acids 73:301, 2005. Radic, Z., Manetsch, R., Krasinski, A., Raushel, J., Yamauchi, J., Garcia, C., Kolb, H.C., Sharpless, K.B., Taylor, P. Molecular basis of interactions of cholinesterases with tight binding inhibitors. Chem. Biol. Interact. 157-158:133, 2005. Rodionov, V.O., Fokin, V.V., Finn, M.G. Mechanism of the ligand-free Cu(I)-catalyzed azide-alkyne cycloaddition reaction. Angew. Chem. Int. Ed. 44:2210, 2005. Whiting, M., Fokin, V.V. Copper-catalyzed reaction cascade: direct conversion of alkynes to N-sulfonylazetidin-2-imines. Angew. Chem. Int. Ed. 45:3157, 2006. Whiting, M., Muldoon, J., Lin, Y.-C., Silverman, S.M., Lindstrom, W., Olson, A.J., Kolb, H.C., Finn, M.G., Sharpless, K.B., Elder, J.H., Fokin, V.V. Inhibitors of HIV1 protease by using in situ click chemistry. Angew. Chem. Int. Ed. 45:1435, 2006. F i g . 4 . Synthesis of polyfunctional dendrimers via copper-catalyzed 1,3-dipolar cycloaddition of azides and alkynes. that have multiple recognition functions (such as 16 α-d-mannose units) and detection elements (2 coumarinderived fluorescent chromophores). The performance of one such bifunctional dendrimer was evaluated in a standard hemagglutination assay with the mannose- Wu, P., Hilgraf, R., Fokin, V.V. Osmium-catalyzed olefin dihydroxylation and aminohydroxylation in the second catalytic cycle. Adv. Synth. Catal. 348:1079, 2006. Wu, P., Malkoch, M., Hunt, J., Vestberg, R., Kaltgrad, E., Finn, M.G., Fokin, V.V., Sharpless, K.B., Hawker, C.J. Multivalent, bifunctional dendrimers prepared by click chemistry. Chem. Commun. (Camb.) 5775, 2005, Issue 46. Zhang, L., Chen, X., Xue, P., Sun, H.H.Y., Williams, I.D., Sharpless, K.B., Fokin, V.V., Jia, G. Ruthenium-catalyzed cycloaddition of alkynes and organic azides. J. Am. Chem. Soc. 127:15998, 2005. 92 CHEMISTRY 2006 THE SCRIPPS RESEARCH INSTITUTE Chemistry, Biology, and Inflammatory Disease P. Wentworth, Jr., J.Y. Chang, Y.P. Chen, J. Dambacher, L. Eltepu, R.K. Grover, J. Nieva, M. Puga, A. Shafton, B.D. Song, M.M.R. Peram, J. Rogel, S. Tripurenani, R. Troseth, H. Wang, A.D. Wentworth ur research is interdisciplinary and involves bioorganic, biophysical, physical organic, synthetic, and analytical chemistry coupled with biochemical techniques, cell-based assays, and animal models. These diverse approaches are combined to facilitate a better understanding of and generate new therapeutic approaches to complex disease states. Ongoing projects include studies on atherosclerosis, neurodegeneration, ischemia-reperfusion injury, macular degeneration, cancer, inflammation, and infectious diseases. O T H E A N T I B O D Y - C ATA LY Z E D WAT E R O X I D AT I O N PAT H WAY Our discovery that all antibody molecules, regardless of source or antigenic specificity, can catalyze the reaction between singlet oxygen and water to give hydrogen peroxide is causing a revision of the axiom that antibodies are the classical adapter molecule of the immune system, linking recognition and killing of foreign pathogens. Both the chemical and biological aspects of this pathway are being explored extensively, and intriguing new insights into how this pathway may play a role in immune defense and inflammatory damage are emerging. We recently showed that flavins, biologically relevant photoactive molecules, can act as efficient triggers for the antibody-catalyzed water oxidation pathway at physiologically relevant concentrations and a certain amount of visible light radiation. This observation indicates that immune defense and damage may be linked to an association of flavins, such as riboflavin or flavin mononucleotide, with immunoglobulins. The trigger would be antibody-antigen binding on cells exposed to visible light, such as the skin or retina. In collaboration with I.A. Wilson, Department of Molecular Biology, we recently studied, at atomic resolution, an antibody, IgGGAR, that has riboflavin bound in the complementarity-determining regions (Fig. 1). Of interest, the bound riboflavin is essentially photochemically inert. The x-ray structure of the IgGGAR-flavin complex offers an insight into the lack of photochemical activity; the isoalloxazine ring is effectively sand- F i g . 1 . Schematic presentation of riboflavin within the binding site of IgGGAR. Residues that form van der Waals interactions with riboflavin are indicated; those that participate in the hydrogen bonds with the riboflavin are shown in ball-and-stick representations. Hydrogen bonds are illustrated as dotted lines. wiched between several aromatic side chains. Binding of riboflavin occurs in a narrow cleft with the isoalloxazine ring stacked between parallel aromatic groups of tyrosine at position H33 (with the re face of riboflavin), phenylalanine at position H58, and tyrosine at position H100A (both associated with the si face of the flavin); the distances between the isoalloxazine ring and the respective aromatic rings from these 3 residues are about 3.2, 3.5. and 3.4 Å, respectively. Such π stacking is known to quench the excited state of riboflavin, a feature that a number of flavin-binding proteins have also evolved, presumably to protect themselves from the intrinsic photochemistry of the flavin moiety. We are searching for the active site for the antibodycatalyzed water oxidation pathway within the antibody structure. We have cloned and expressed soluble individual domains of the murine Fab 4C6. Initial attempts to efficiently express wild-type domains were unsuccessful, but by mutating hydrophobic residues involved in interdomain interactions into soluble hydrophilic residues, we prepared folded domains. All the domains we cloned and expressed have been successfully purified to homogeneity from the periplasm of Escherichia coli transformed with plasmids encoding individual domains. In addition, to assess the critical nature of tryptophan residues in the photosensitization of antibody-bound triplet oxygen to singlet oxygen, we mutated 2 tryptophan residues within domain CH1 to leucine. Interest- CHEMISTRY 2006 ingly, neither the single nor the double mutation affected the ability of ultraviolet-irradiated CH1 to generate hydrogen peroxide. C H O L E S T E R O L S E C O - S T E R O L S A N D I N F L A M M AT O R Y DISEASE We recently discovered that the 5,6-seco-sterols atheronal-A and atheronal-B (Fig. 2), compounds of a class of cholesterol ozonolysis products, are present in F i g . 2 . The cholesterol seco-sterols atheronal-A (top) and atheronal-B (bottom). human atherosclerotic plaques and plasma. In addition, we found that the atheronals are present in murine models of atherosclerosis, in a rabbit model of acute respiratory distress syndrome (studies done in collaboration with C. Cochrane, Department of Molecular and Experimental Medicine), and in brain tissue from patients with Lewy body dementia (studies done in collaboration with J. Kelly, Department of Chemistry). The atheronals have a range of biological properties that in combination would increase the number of macrophages at sites of vascular inflammation. When cultured macrophage cells were incubated with atheronal-A and atheronal-B complexed with low-density lipoprotein (LDL), marked upregulation of scavenger receptor class A, but not CD36, occurred, showing that cultured macrophages respond to complexes of atheronals and LDL in a manner highly analogous to the response to acetylated LDL. Both atheronal-A and atheronal-B induce chemotaxis of cultured macrophages in a dose-dependent manner. When complexed with LDL, atheronal-A, but not atheronal-B, induces a dose-dependent upregulation of the cell-surface adhesion molecule E-selectin on vascular endothelial cells. When complexed with THE SCRIPPS RESEARCH INSTITUTE 93 LDL, atheronal-B, but not atheronal-A, induces cultured human monocytes to differentiate into macrophage cell lineage. These in vitro data and the effects of cholesterol 5,6-seco-sterols on the formation of foam cells and the cytotoxic effects of macrophages indicate that the atheronals have biological effects that could lead in vivo to the recruitment, entrapment, dysfunction, and ultimate destruction of macrophages, the major leukocytes in inflammatory artery disease. The ultimate goals of research for genetic and environmental factors that increase the propensity of a specific protein to misfold are the understanding and treatment of disease states as diverse as atherosclerosis, light-chain deposition disease, systemic amyloidosis, Alzheimer’s disease, and Parkinson’s disease. We showed that the inflammation-derived atheronal-A and atheronal-B trigger a deformation in the secondary structure of the normally folded protein apolipoprotein B-100 into a proamyloidogenic form. In collaboration with Dr. Kelly, we extended this model and showed that these cholesterol seco-sterols also trigger the misfolding of amyloid β-peptide(1–40), leading to formation of fibrils similar to those observed in patients with Alzheimer’s disease. Interestingly, analysis of the structure-activity relationship revealed that among a panel of aldehydes, only atheronal-A, atheronal-B, and 4-hydroxynonenal trigger misfolding of amyloid β-peptide, suggesting that structural aspects of the aldehyde and not simple protein adduction were critical to this misfolding. More recently, using mutated synthetic sequences of amyloid β-peptide(1–40), we found that the accelerated aggregation of this protein only occurs when a particular lysine of the sequence is modified. We have also shown that atheronals and other lipid aldehydes accelerate the aggregation of several wild-type amyloidogenic proteins, including immunoglobulin light chains, mouse prions, and the tumor suppressor protein p53. The generality and specificity of this process suggest that inflammatory aldehydes and their posttranslational modification of amyloidogenic peptides may be the chemical link between the known associations of inflammation, oxidative damage, and various protein misfolding diseases. INTERACTION BETWEEN PROTOZOAN J-BINDING P R O T E I N 1 A N D G LY C O S Y L AT E D D N A Current treatments of parasitic infections such as leishmaniasis, African trypanosomiasis, and American trypanosomiasis are limited in terms of their effective- 94 CHEMISTRY 2006 ness, increasing drug resistance and the inherent toxic effects of the drugs. Thus, an elucidation of new parasite-specific biological targets for new therapeutic agents is needed. In this regard, the discovery that DNA from members of the order Kinetoplastida, but not from other eukaryotes, contains an unusual modified base, β-D glucosyl(hydroxymethyl)uracil, called base J (compound 1a in Fig. 3) was a breakthrough. Extracts of several kinetoplastids contain a J-binding protein (JBP) that THE SCRIPPS RESEARCH INSTITUTE that some loci around the glycan are essential for binding and some are not. This information is being translated into structure-based design of chemical libraries as inhibitors of JBP-1 binding. PUBLICATIONS Bielsch, J., Wang, Q.T., Bosco, D., Powers, E.T., Wentworth, P., Jr., Kelly, J. Inflammatory metabolite-initiated protein misfolding. Acc. Chem. Res., in press. 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 accelerate α-synuclein fibrilization [published correction appears in Nat. Chem. Biol. 2:346, 2006]. Nat. Chem. Biol. 2:249, 2006. Nieva, J., Kerwin, L., Wentworth, A.D., Lerner, R.A., Wentworth, P., Jr. Immunoglobulins can utilize riboflavin (vitamin B2) to activate the antibody-catalyzed water oxidation pathway. Immunol. Lett. 103:33, 2006. Rogers, C.J., Dickerson, T.J., Wentworth, P., Jr., Janda, K.D. A high-swelling reagent scaffold suitable for use in aqueous and organic solvents. Tetrahedron 61:12140, 2005. Takeuchi, C., Galve, R., Nieva, J., Witter, D.P., Wentworth, A.D., Troseth, R.P., Lerner, R.A., Wentworth, P., Jr. Proatherogenic effects of the cholesterol ozonolysis products, atheronal-A and atheronal-B. Biochemistry 45:7162, 2006. Toker, J.D., Tremblay, M., Yli-Kauhaluoma, J., Wentworth, A.D., Zhou, B., Wentworth, P., Jr., Janda, K.D. Exploring the scope of the 29G12 antibody catalyzed 1,3-dipolar cycloaddition reaction. J. Org. Chem. 70:7810, 2005. Witter, D., Wentworth, P., Jr. The antibody-catalyzed water-oxidation pathway from discovery to an emerging role in health and disease. Antioxid. Redox Signal., in press. F i g . 3 . Base J (1a) and base J analogs (1b–1g) synthesized and used as probes for studying JBP-1–J-DNA recognition. Inset, Image 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. Nat. Acad. Sci. U. S. A. 103:3581., 2006. of base J in doubled-stranded DNA shows how glucose sits in the major groove. specifically binds to J-containing duplex DNA. JBP-1 is essential in Leishmania. As a drug target, JBP has merit. The protein shares little homology with other proteins in the Protein Data Bank, and it has a unique ligand, J-DNA containing telomeric stretches of double-stranded DNA, that does not occur in other eukaryotes. However, a preliminary high-throughput screen, focused on disrupting binding between JBP-1 and J-DNA, with a library of compounds consisting of all the major drug pharmacophoric groups revealed no compounds of interest. In parallel, we have studied the nature of the molecular recognition that underpins JBP-1 recognition of glycosylated DNA. We synthesized a panel of modified J-containing bases (compounds 1b–1g in Fig. 3) and incorporated them into a 16-nucleotide telomeric stretch of double-stranded DNA. In collaboration with D. Millar, Department of Molecular Biology, we determined the dissociation constants of these analogs for JBP-1 and generated a ∆G of binding assessment of each hydroxyl around the glucosyl core. This analysis revealed Bioorganic and Synthetic Chemistry C.-H. Wong, C. Bennett, A. Brik, Y.-H. Chen, S. Dean, S. Ficht, M. Fujio, W. Greenberg, R. Guy, S. Hanson, Z. Hong, T.-L. Hsu, D.-R. Hwang, J.-C. Lee, P.-H. Liang, L. Liu, T. Polat, M. Sawa, M. Sugiyama, D. Thayer, S.-K. Wang, L. Whalen, C.-Y. Wu, D. Wu, M. Wuchrer, Y.-Y. Yang ur research programs involve development of new chemical and enzymatic strategies and methods for the synthesis of biologically active compounds. We use the synthesized compounds as molecular probes to explore carbohydrate-mediated biological recognition events and enzymatic reactions. O ORGANIC AND BIOORGANIC SYNTHESIS Our work in organic and bioorganic synthesis includes the development of new chemical reactions and the exploitation of native and engineered enzymes for organic synthesis. In the past year, we developed new methods for making glycopeptides and glycoproteins via native chemical ligation methods. We will use these CHEMISTRY 2006 methods to synthesize homogenous glycoproteins that are important therapeutic agents in humans. We continue to develop covalent glycoarrays for high-throughput analysis of protein-carbohydrate interactions and the use of aldolases in the synthesis of glycosyltransfer enzyme inhibitors. Using directed evolution, we developed new aldolase variants capable of making both enantiomers of sugars. DEVELOPMENT OF INHIBITORS OF ENZYMES AND RECEPTORS Our goals in the area of enzyme and receptor inhibitors are to develop new strategies to discover inhibitors and ligands with high selectivity as potential therapeutic agents. Current strategies involve the design and synthesis of structure- and mechanism-based inhibitors of enzymes associated with a variety of diseases. Targets for investigation include bacterial transglycosidase, sulfotransferases, retroviral proteases, lethal factor of Bacillus anthracis, and enzymes involved in the biosynthesis of carbohydrates essential for biological functions. We have developed new iminocyclitols and derivatives as inhibitors of glycosidases and glycosyltransferases for potential treatment of inflammatory diseases. In addition, using a new strategy based on a rapid microscale synthesis coupled with in situ high-throughput screening, we developed new tight-binding inhibitors of anthrax lethal factor, a sulfotransferase, and drug-resistant HIV proteases. We also developed new reactions based on tetrabutylammonium fluoride–mediated N- and O-alkylation and epoxide opening in aqueous solution and used the reactions to identify potent enzyme inhibitors. C A R B O H Y D R AT E C H E M I S T R Y A N D M O L E C U L A R G LY C O B I O L O G Y We continue to improve the programmable 1-pot oligosaccharide synthesis method for convenient and rapid preparation of oligosaccharides. So far, we have designed approximately 600 building blocks and measured the anomeric reactivity of each building block. Using the computer program OptiMer, developed in our laboratory, we rapidly assembled a number of oligosaccharides. We are using this method to define the specificity of interactions between carbohydrates and their receptors, with particular focus on optimization of the cancer antigen Globo H and HIV gp120 oligomannose as vaccine candidates. In collaboration with D.R. Burton, Department of Immunology, and I.A. Wilson, Department of Molecular Biology, we are evaluating a designed oligomannose-protein conjugate as an antigen to elicit antibodies for neutralizing HIV gp120 and variants. THE SCRIPPS RESEARCH INSTITUTE 95 We synthesized a series of bacterial glycolipids and analogs and found that they are active ligands for CD cell markers involved in activation of human natural killer T cells. These compounds may be useful as immunotherapeutic agents for the treatment of bacterial and viral infections, as well as cancer, and we are elucidating the structural basis for their activity. In collaboration with Dr. Wilson, we are determining the structures of the complexes formed by these glycolipids and CD1d; the results will assist in designing ligands with improved therapeutic potential. In collaboration with J.C. Paulson, Department of Molecular Biology, we developed new methods for microfabrication of saccharides on glass slides or microtiter plates for use in high-throughput analysis of sugar-protein interactions. PUBLICATIONS Brigl, M., van den Elzen, P., Chen, X., Meyers, J.H., Wu, D., Wong, C.-H., Reddington, F., Illarianov, P.A., Besra, G.S., Brenner, M.B., Gumperz, J.E. Conserved and heterogeneous lipid antigen specificities of CD1d-restricted NKT cell receptors. J. Immunol. 176:3625, 2006. Brik, A., Ficht, S., Wong, C.-H. Strategies for the preparation of homogenous glycoproteins. Curr. Opin. Chem., in press. Brik, A., Wu, C.-Y., Wong, C.-H. Microtiter plate based chemistry and in situ screening: a useful approach for enzymatic inhibitor discovery. Org. Biomol. Chem. 4:1446, 2006. Brik, A., Yang, Y.-Y., Ficht, S., Wong, C.-H. Sugar-assisted glycopeptide ligation. J. Am. Chem. Soc. 128:5626, 2006. 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, Y.-S.E., Lo, K.-H., Hsu, H.-H., Shao, Y.-M., Yang, W.-B., Lin, C.-H., Wong, C.-H. Screening for HIV protease inhibitors by protection against activitymediated cytotoxicity in Escherichia coli. J. Virol. Methods 137:82, 2006. Chuang, M.-H., Wu, M.-S., Lo, W.-L., Lin, J.-T., Wong, C.-H., Chiou, S.-H. The antioxidant protein alkylhydroperoxide reductase of Helicobacter pylori switches from a peroxide reductase to a molecular chaperone function. Proc. Natl. Acad. Sci. U. S. A. 103:2552, 2006. Fujio, M., Wu, D., Garcia-Navarro, R., Ho, D.D., Tsuji, M., Wong, C.-H. Structurebased discovery of glycolipids for CD1d-mediated NKT cell activation: tuning the adjuvant versus immunosuppression activity. J. Am. Chem. Soc. 128:9022, 2006. Hong, Z.-Y., Liu, L., Hsu, C.-C., Wong, C.-H. Three-step synthesis of sialic acids and derivatives. Angew. Chem. Int. Ed., in press. Hsu, H.-Y., Hua, K.-F., Su, Y.-C., Chu, L.-C., Su, S.-C., Chiu, H.-W., Wong, C.-H., Chen, S.-T., Shieh, C.-W., Yang, S.-S., Chen, Y.-M., Chao, L.K. Alkali-soluble polysaccharides of Rhizoclonium riparium alga induces IL-1 gene expression via protein kinase signaling pathways. J. Agric. Food Chem. 54:3558, 2006. Huang, C.-Y., Thayer, D.A., Chang, A.Y., Best, M.D., Hoffmann, J., Head, S., Wong, C.-H. Carbohydrate microarray for profiling the antibodies interacting with Globo H tumor antigen. Proc. Natl. Acad. Sci. U. S. A. 103:15, 2006. Huang, K.-T., Wu, B.-C., Lin, C.-C., Luo, S.-C., Chen, C., Wong, C.-H., Lin, C.-C. Multi-enzyme one-pot strategy for the synthesis of sialyl Lewis X-containing PSGL-1 glycopeptide. Carbohydr. Res. 341:2151, 2006. 96 CHEMISTRY 2006 Kinjo, Y., Tupin, E., Wu, D., Fujio, M., Garcia-Navarro, R., Benhnia, M.R.E.I., Zajonc, D.M., Ben-Menachem, G., Ainge, G.D., Painter, G.F., Khurana, A., Hoebe, K., Behar, S.M., Beutler, B., Wilson, I.A., Tsuji, M., Sellati, T.J., Wong, C-H., Kronenberg, M. Natural killer T cells recognize diacylglycerol antigens from pathogenic bacteria. Nat. Immunol. 7:978, 2006. Lee, J.-C., Wu, C.-Y., Apon, J.V., Siuzdak, G., Wong, C.-H. Reactivity-based onepot synthesis of the tumor-associated antigen N3 minor octasaccharide for the development of a photocleavable DIOS-MS sugar array. Angew. Chem. Int. Ed. 45:2753, 2006. Liang, F.-S., Brik, A., Lin, Y.-C., Elder, J.H., Wong, C.-H. Epoxide opening in water and screening in situ for rapid discovery of enzyme inhibitors in microtiter plates. Bioorg. Med. Chem. 14:1058, 2006. Liang, P.-H., Cheng, W.-C., Lee, Y.-L., Yu, H.-P. Wu, Y.-T., Lin, Y.-L., Wong, C.-H. Novel five-membered iminocyclitol derivatives as selective and potent glycosidase inhibitors: new structures for antivirals and osteoarthritis. Chembiochem 7:165, 2006. Liang, F.-S., Greenberg, W.A., Hammond, J.A., Hoffmann, J., Head, S.R., Wong, C.-H. Evaluation of RNA-binding specificity of aminoglycosides with DNA microarrays. Proc. Natl. Acad. Sci. U. S. A. 103:12311, 2006. Lin, K.-I., Kao, Y.-Y., Kuo, H.-K., Yang, W.-B., Chou, A., Lin, H.-H., Yu, A.L-T., Wong, C.-H. Reishi polysaccharides induce immunoglobulin production through the TLR4/TLR2-mediated induction of transcription factor Blimp-1. J. Biol. Chem. 281:24111, 2006. Lin, Y.-C., Brik, A., de Parseval, A., Tam, K., Torbett, B.E., Wong, C.-H., Elder, J.H. Altered gag polyprotein cleavage specificity of feline immunodeficiency virus/human immunodeficiency virus mutant proteases as demonstrated in a cellbased expression system. J. Virol. 80:7832, 2006. Liu, L., Bennett, C.S., Wong, C.-H. Advances in glycoprotein synthesis. Chem. Commun. (Camb.) 21, 2006, Issue 1. Liu, L., Hong, Z.-Y., Wong, C.-H. Convergent glycopeptide synthesis by traceless Staudinger ligation and enzymatic coupling. Chembiochem 7:429, 2006. Liu, H., Wong, C.-H. Characterization of a transglycosylase domain of Streptococcus pneumoniae PBP1b. Bioorg. Med. Chem. 14:7187, 2006. Qiul, H., Gabrielsen, A., Agardh, H.E., Wan, M., Wetterholm, A., Wong, C.-H., Hedin, U., Swedenborg, J., Hansson, G.K., Samuelsson, B., Paulsson-Berne, G., Haeggstrom, J.Z. Expression of 5-lipoxygenase and leukotriene A4 hydrolase in human atherosclerotic lesions correlates with symptoms of plaque instability. Proc. Natl. Acad. Sci. U. S. A. 103:8161, 2006. Sanna, M.G., Wang, S.-K., Gonzalez-Cabrera, P.J., Don, A., Marsolais, D., Matheu, M.P., Wei, S.H., Parker, I., Jo, E., Cheng, W.-C., Cahalan, M.D., Wong, C.-H., Rosen, H. Enhancement of capillary leakage and restoration of lymphocyte egress by a chiral S1P1 antagonist in vivo. Nat. Chem. Biol. 2:434, 2006. Sawa, M., Hsu, T.-L., Itoh, T., Sugiyama, M., Hanson, S.R., Vogt, P.K., Wong, C.-H. Glycoproteomic probes for fluorescent imaging of fucosylated glycans in vivo. Proc. Natl. Acad. Sci. U. S. A. 103:12371, 2006. Sawkar, A.R., Adamski-Werner, S.L., Cheng, W.-C., Wong, C.-H., Beutler, E., Zimmer, K.-P., Kelly, J.W. Gaucher disease-associated glucocerebrosidases show mutation-dependent chemical chaperoning profiles. Chem. Biol. 12:1235, 2005. Scanlan, C., Calarese, D., Lee, H.K., Blixt, O., Wong, C.-H., Wilson, I., Burton, D., Dwek, R., Rudd P. Antibody recognition of a carbohydrate epitope: a template for HIV vaccine design. Adv. Exp. Med. Biol. 564:7, 2005. Shie, J.-J., Fang, J.-M., Kuo, T.-H., Kuo, C.-J., Liang, P.-H., Huang, H.-J., Wu, Y.-T., Jan, J.-T., Cheng, Y.-S.E., Wong, C.-H. Inhibition of the severe acute respiratory syndrome 3CL protease by peptidomimetic α,β-unsaturated esters. Bioorg. Med. Chem. 13:5240, 2005. Thayer, D., Wong, C.-H. Vancomycin analogs with improved biological activity: a combined one-pot enzymatic glycosylation and chemical diversification strategy. Chem. Asian J., in press. Wei, S.H., Rosen, H., Matheu, M.P., Sanna, M.G., Wang, S.-K., Jo, E., Wong, C.-H., Parker, I., Cahalan, M.D. Sphingosine 1-phosphate type 1 receptor agonism inhibits transendothelial migration of medullary T cells to lymphatic sinuses. Nat. Immunol. 6:1228, 2005. THE SCRIPPS RESEARCH INSTITUTE Whalen, L.J., Wong, C.-H. Enzymes in organic synthesis: aldolase-mediated synthesis of iminocyclitols and novel heterocycles. Aldrichim. Acta 39:63, 2006. Wu, C.-Y., Brik, A., Wang, S.-K., Chen, Y.-H., Wong, C.-H. Tetrabutylammonium fluoride-mediated rapid alkylation reaction in microtiter plates for the discovery of enzyme inhibitors in situ. Chembiochem 6:2176, 2005. Wu, C.-Y., King, K.-Y., Kuo, C.-J., Fang, J.-M., Wu, Y.-T., Ho, M.-Y., Liao, C.-L., Shie, J.-J., Liang, P.-H., Wong, C.-H. Stable benzotriazole esters as mechanismbased inactivators of the severe acute respiratory syndrome 3CL protease. Chem. Biol. 13:261, 2006. 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. 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.