Development of Biomimetic Microfluidic Adhesive Substrates for Cell Separation by Chia-Hua Lee CMASSACHUSETTS B.S., National Taiwan University, Taiwan (2003) M.S., National Taiwan University, Taiwan (2005) INSTt1UTE OF TECHNOLOGY OF.TECNOLOG MAY 14211 M.S., Massachusetts Institute of Technology, USA (2008) LiBRARES Submitted to the Department of Materials Science and Engineering in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy at the MASSACHUSETTS INSTITUTE OF TECHNOLOGY February 2014 © 2014 Massachusetts Institute of Technology. All rights reserved Signature of Author Department of Materials Science and Engineering January 10 th014 Certified by Rohit Karnik Associate Professor of Mechanical Engineering Thesis Co-advisor Krystyn J. Van Vliet V Associat Pr fessor of Materials Science and Engineering Thesis Co-advisor Accepted by Chair, Depart ommittee on Graduate Student Development of Biomimetic Microfluidic Adhesive Substrates for Cell Separation by Chia-Hua Lee Submitted to the Department of Materials Science and Engineering on January 10 th 2014 in partial fulfillment of the requirements for the Degree of Doctor of Philosophy in Materials Science and Engineering ABSTRACT Cell separation is important in medical, biological research, clinical therapy, diagnostics and many other areas. The conventional methods of cell sorting have limited applications due to sophisticated equipment settings, high costs, or time-intensive and labor-intensive processing steps. Inspired from natural cell sorting system-cell rolling, a novel microfluidic device design was proposed for point-of-care and point-of-use applications. It relies on interaction of cells with biomimetic adhesive substrates comprising multiple inclined, asymmetric bands of weak adhesive molecules. Such device design allows continuous sorting of cells without irreversible capture of cells. To realize such device, comprehensive studies of how cells settle onto the substrate, how cells capture by the substrate, the effect of substrate parameters on separation potential, and selection of adhesion molecules are needed to optimize device performance. In this thesis, first, how the cells settle and how they are captured by the receptors were studied using HL60 cells as a model leukocyte cell line and P-selectin as a model receptor. Settling distance of HL60 cells under different shear stresses inside microfluidic channels was identified from the study of convection velocity of cells at different position along the channel. The results show that settling distance increases with increasing shear stress. Cell capture was then quantified by characterizing how far settled HL60 cells travelled before they were captured by P-selectin molecules, defined as the attachment distance. Cumulative probabilities of attachment distance of cells at different shear stresses revealed that increasing shear stress results in exponential increase of the attachment distance of cells by receptor molecules. An empirical model was developed to predict capture probability by an inclined receptor band and the prediction value was verified by experimental data from a device. 3 Second, a patterning method involving microcontact printing was developed to create biomimetic adhesive substrates comprising multiple inclined receptor bands of P-selectin molecules. The patterned substrates were then used to study how transport of HL60 cells can be controlled by the substrate parameters including pattern inclination angle with respect to shear flow direction, shear stress magnitude, and P-selectin incubation concentration. The effects of substrate parameters were quantified in terms of the edge tracking length, lateral displacement, and the rolling velocity. The edge inclination angle was identified as the strongest modulator of edge tracking length on a single band for captured cells. To study optimization of the device design, experimental data of cell settling, cell attachment, and edge tracking length were integrated into a model to predict device performance including device capture efficiency and total lateral displacement. General guidelines for microfluidic device design were established based on the results from the model: smaller band width, edge angle of 15-20', and lower shear stress. Finally, to develop new specific receptor-ligand systems, M13 pVIII and pIll phage libraries were used for selecting peptides with affinity to CD4 proteins. Screened phage from pVIII library was immobilized on the gold surface and capture efficiency of CD4+ cells were characterized. The interaction between selectin phage and CD4+ cells were demonstrated to be CD4-dependent. Moreover, the selected phage from pIII library and the corresponding synthetic peptides were demonstrated to exhibit specificity to CD4 proteins. In summary, this thesis focuses on development of biomimetic adhesive substrates for microfluidic devices involving transient interactions between the cells and the receptorpatterned substrates. How cells flow and get captured by patterned biomimetic substrates inside the microfluidic channels, how substrates parameters affect cell rolling trajectories and device performance, and how to identify new receptor-ligand systems were discussed in this thesis. This study has led to realization of a microfluidic device for separating neutrophils from blood. This microfluidic system provides continuous sorting without irreversible capture of cells, and is believed to be an effective method that can potentially be used in many point-of-care applications. Keywords: microfluidics, cell separation, cell rolling, selectin, biopanning, M13 Thesis Committee: Prof. Rohit Karnik, Associate Professor of Mechanical Engineering, MIT. (Thesis Co-advisor) Prof. Krystyn J. Van Vliet, Associate Professor of Materials Science and Engineering, MIT. (Thesis Co-advisor) Professor Angela M. Belcher, Professor of Materials Science and Engineering, MIT. Professor Darrell J. Irvine, Professor of Materials Science and Engineering, MIT. Prof. Jeffrey M. Karp, Associate Professor of Medicine, Harvard Medical School, Brigham and Women's Hospital. 4 Acknowledgements My time at MIT has been an incredible journey. I owe thanks to many people for their guidance, support, encouragement, patience, and friendship. I would like to express my gratitude to my PhD thesis advisors (Prof. Rohit Karnik and Prof. Krystyn Van Vliet) and my committee members (Prof. Angela Belcher, Prof. Jeffrey Karp and Professor Darrell Irvine) for all their support and guidance, which without, this thesis could not have been achieved. Prof. Kamik, especially, has been an exceptional role model and mentor to me. He has been always there to listen and give advice with his patience. His unflinching encouragement and support have helped me overcome many crisis situations and finish this dissertation. I am also deeply grateful to Prof. Van Vliet for the discussions that have helped me sort out a lot of technical details of my work in the past few years. She has left her mark on me with her professionalism and encouragement of my work. Moreover, my thesis work would not have been possible without the support from Prof. Belcher. It has been an extraordinary experience for me to work in her lab, too. Her insightful comments and constructive criticisms have helped me in all the time of my research work. Prof. Karp has offered fresh perspectives on this work, which have motivated its completion. Prof. Irvine has offered many advices over the course of the thesis and for that I am grateful as well. I am lucky to have interactions with three amazing groups of people (the Kamik group, the VV group and the Belcher group) that I have ever had the opportunity to work with. In particular, Suman in the Kamik group trained me in nearly every critical area and offered much help and guidance to me. Chong also provided invaluable assistance for my research, too. Jongho, Marco, Jong-Min, Sean, Tarun, Mike, Sunandin, Sung-Yong, HuiYing, Minsoung, Mohamed, who joined the Kamik group well after me, have been valuable friends and colleagues throughout. I also greatly appreciate that the VV group members always give me their full support. From Belcher group, I especially owe much to Nimrod, Rana, and Gaelen, who have all actively contributed to my work with suggestions and feedback. I also thank other Belcher members who have offered much help to me. There have been innumerable others at MIT who have affected my life here and I would like to express my gratitude to them as well: Prof. Karen Gleason, Prof. Sang-Gook Kim, and Prof. Samuel Allen, Richa, Yin, Jane, Yvonne, Liang-Yi, Yi-Chun, Po-Yen, Alina, Ting-Yen, Chia-Hou, Alice, Sabrina, Hao-Wei, David, Yu-Chung, Wei-Shan, Lei, Xiaoting, Hyeongho, Benita, Geetha, and Meri. Moreover, I would like to thank HisnYing, Ivy, Kevin, Ying-Chieh, Jiun-Tai, and Min-Chen for their friendship for years. Last, but not the least, I would like to specially thank my family: my grandparents, my parents, Chung-Yi, Wan-Yu, and Yueh-Hsun for their love and support. 5 This page is intentionally left blank. 6 1 Introduction.....................................................................19 1.1 Background and Motivation........................................................................... 19 1.1.1 Surface Marker-Based Cell Separation ............................................. 20 1.1.2 Low Affinity Based Separation ........................................................ 23 1.1.2.1 C ell Rolling .......................................................................... 24 1.1.2.2 Cell Rolling-Based Separation.............................................28 2 1.2 Scope of the Thesis W ork ............................................................................. 31 1.3 Chapter Reference ........................................................................................ 32 Free-Flow Travel and Capture of HL60 Cells inside Microfluidic Channels............................................................................37 2.1 Introduction .................................................................................................... 37 2.2 Part A. Cell Settling inside Microfluidic Channels...................................... 40 Fabrication of Substrate and Microfluidic Channel ......................... 40 2.2.1.1 M aterials............................................................................... 40 2.2.1 2.2.1.2 Fabrication of Substrate Incorporated PDMS channel.........41 2.2.2 Convection Velocity Experiment ...................................................... 41 2.2.2.1 C ell Culture .......................................................................... 41 2.2.2.2 Experim ent Setup ................................................................. 41 2.2.2.3 Results and Data Analysis....................................................42 2.3 Part B. Attachment Distance of the Cells in the Microfluidic Channel........46 2.3.1 Fabrication of Substrate and Microfluidic Channel ......................... 46 2.3.1.1 M aterials............................................................................... 46 2.3.1.2 Fabrication of Substrate Incorporated PDMS channel.........47 2.3.2 2.3.1.3 Substrate Characterization ................................................... 47 Cell Capture Experiment ................................................................. 48 2.3.2.1 Cell Culture and Experiment Setup.......................................48 2.3.2.2 D ata A nalysis ........................................................................ 2.3.3 48 Flow Cytometry Analysis.................................................................54 2.4 C onclusion .................................................................................................... 55 2.5 Chapter Reference ........................................................................................ 56 7 3 Lateral Displacement of HL60 Cells Rolling on Inclined P-Selectin Adhesive Bands.....................................................................59 3.1 Introduction ................................................................................................... 59 3.2 Fabrication of P-selectin-Patterned Substrates............................................. 61 3.2.1 M aterials.......................................................................................... 61 3.2.2 Fabrication of PDMS Stamp ............................................................. 61 3.2.3 Fabrication of Patterned Substrates ................................................. 62 3.2.4 Substrate Characterization............................................................... 63 3.3 Cell Rolling on Asymmetric Patterns in a Flow Chamber............................64 3.3.1 C ell C ulture...................................................................................... 64 3.3.2 Experim ent Setup ............................................................................ 64 3.3.3 Results and Data Analysis ............................................................... 65 3.3.3.1 Effect of Edge Angle on the Rolling Behavior of HL60 Cells 68 3.3.3.2 Effect of Shear Stress on Rolling Behavior of HL60 Cells ..... 70 3.3.3.3 Effect of P-selectin Incubation Concentration on Rolling Behavior of HL60 Cells ........................................................ 72 3.3.3.4 Detachment of Cells Rolling Along an Edge Can be Described by a Poisson Process. ........................................................... 3.4 73 Optimization of Device Design .................................................................... 80 3.4.1 Effect of Band Width (w) on Device Performance...........................84 3.4.2 Effect of Edge Inclination Angle on Device Performance ............... 3.4.3 Effect of Shear Stress on Device Performance.................................87 86 3.5 C onclusion ...................................................................................................... 91 3.6 Chapter 92 ........................................................................................ 4 Identification of Adhesion Molecules with Weak Affinity via Phage Display from M13 pVIII Library .............................................................. 97 4.1 Introduction ................................................................................................... 97 4.2 Biopanning of pVIII Library ........................................................................ 99 4.2.1 Phage display library of random peptides ........................................ 99 4.2.2 Biopanning Procedure ........................................................................ 8 100 4.2.3 Phage Titering.....................................................................................102 4.2.3.1 Lysogeny Broth (LB) Medium Preparation............102 4.2.3.2 Tetracyline (Tet) Stock Solution Preparation ........................ 102 4.2.3.3 IPTG/Xgal Stock Solution Preparation..................................102 4.2.3.4 LB/IPTG/Xgal Plates Preparation..........................................102 4.2.3.5 Agarose Top...........................................................................102 4.2.3.6 Titration Procedure.................................................................103 4.2.4 Amplification of M13 bactriophage ................................................... 104 4.2.4.1 PEG/NaCl Stock Solution Preparation...................................104 4.2.4.2 Amplification between Rounds of Panning ........................... 4.2.5 DNA extraction for sequence identification.......................................105 4.2.6 Biopanning Results.............................................................................107 4.3 Immobilization of phage on Au-coated glass slide ......................................... 4.4 Substrate Characterization...............................................................................110 4.5 Cell Capture Experiments ............................................................................... 4.6 104 109 112 4 .5.1 C ell Culture.........................................................................................112 4.5.2 Cell Staining ....................................................................................... 112 4.5.3 Cell Capture Experiment in Flow Chamber ....................................... 113 Cell Capture Efficiency...................................................................................114 114 4.6.1 Phage-engineered Surface .................................................................. 4.6.2 CD4-blocked Phage-engineered Surface............................................116 4.7 Peptide Sequences Characterization of pVIII Library .................................... 117 4 .8 C onclu sion ....................................................................................................... 120 4.9 Chapter Reference ........................................................................................... 120 5 Identification of Adhesion Molecules with Weak Affinity via Phage Display from M 13 pII Library ............................................................... 5.1 Introduction ..................................................................................................... 5.2 Biopanning of pIll Library..............................................................................125 5.3 5.2.1 Biopanning Procedure ........................................................................ 5.2.2 Biopanning Results.............................................................................127 Enzyme-linked Immunosorbent Assay (ELISA) ............................................ 9 123 123 125 130 5.3.1 Surface Density of CD4 on Well Plate as the Target of Biopanning .130 5.3.1.1 Materials.................................................................................130 5.3.2 5.3.1.2 M ethod ................................................................................... 131 5.3.1.3 ELISA Analysis ..................................................................... 131 Specificity of the Selected Phage to CD4 Protein .............................. 132 5.3.2.1 M aterials.................................................................................132 5.3.2.2 M ethods..................................................................................132 5.3.2.3 ELISA Analysis ..................................................................... 5.4 Specificity of the Synthetic Peptide to CD4 protein ....................................... 137 5.4.1 Synthetic peptide ................................................................................ 137 5.4.2 Conjugation of Peptide to the Alexa Fluor 488 Dye .......................... 137 5.4.3 Purification of Dye-conjugated Peptides............................................138 5.4.4 Staining the CD4-functionalized Beads with the Dye-conjugated P eptid es .............................................................................................. 5.4.5 13 9 Characterization of Binding Specificity between Peptide and the Functionalized Beads ......................................................................... 6 134 5.5 Conclusion.......................................................................................................143 5.6 Reference.........................................................................................................143 Conclusion and Future W ork.............................................................145 10 140 List of Figures Figure 1-1 Multistep leukocyte adhesion cascade in response to inflammation. Selectins initiate capture and rolling of the leukocytes. Integrins are involved in adhesion and cause the cells to arrest and transmigrate into the underlying tissues .................... 24 Figure 1-2 Selectins and their major ligands. Three selectin family members (P-, E-, and L-selectin) and their corresponding ligands are shown in this figure. These three .... 25 Figure 1-3 Force-dependent lifetimes of single bonds of PSGL-1/ P-selectin (blue) and PSGL-1/ L-selectin (purple) under different shear stress. The catch-to-slip bond transition under increasing forces has been observed: the lifetime increases (catch bond) first until it reaches a maximal value and then decreases (slip bond)..........................26 Figure 1-4 Scanning electron microscope (SEM) images of neutrophils. (a) Localization of PSGL-1 on microvilli. A human neutrophil was incubated with a mixture of the anti-PSGL-1 antibodies, followed by a rabbit anti-mouse antibody, and then with protein A-gold of 5 nm diameter. The arrow shows the gold nanoparticles (b) low magnification of tethered neutrophils. The neutrophil cell has multiple projections named microvilli on its surface. (c) high magnification of tethered neutrophils. The neutrophil was rolling on P-selectin under shear stress 2 dyn/cm 2 . The arrow shows a 28 tether. Scare bars are all referred to 1 m. .................................................................. Figure 1-5 Effect of a receptor pattern on the motion of a rolling cell. Tracks of HL60 cells rolling on patterned P-selectin edge show that patterning can direct the trajectories of rolling cells along the edges (only long tracks are shown on the right)....................29 Figure 1-6 Schematic diagram of a device for separation of cells. Cells rolling along band edges are laterally displaced into the adjacent buffer stream, resulting in separation. Pink lines indicate adhesion molecule-functionalized regions. Red and blue circles are interacting cells and non-interacting cells, respectively...............................................30 Figure 2-1 Schematic diagrams of cells travelling in a microfluidic channel. (a) The illustration of a cell entering the microfluidic channel and travelling in both x and y directions. The settling distance, xs, is defined as how far a cell will travel before it settles close enough to the bottom surface. (b) Illustration of cells passing the Pselectin-coated area. A cell may be captured from the flowing stream at an attachment 39 distance, /a, after it enters the P-selectin-coated area. ................................................... Figure 2-2(a) Convection velocity (u,,) of the cells at the different horizontal position. Convection velocity divided by shear stress decreases and reaches a minimum value as the cells travel along the channel. Data are presented as the mean and the standard deviation of the convection velocity obtained from 20 cells under each experiment condition. Different markers and colors represent different shear stresses. (b) 11 of the cells at different horizontal positions (c) Variation of the convection velocity of the settled cells with the shear stress. Inset summaries the mean and the standard deviation of the convection velocity of the cells at position x = 13 cm. The relationship between u, and T is well described by a linear fit (dashed line). ................................. 46 Figure 2-3 Fluorescent images of the substrate. The bright area corresponds to FITCB SA -coated area................................................................................................................47 Figure 2-4 HL60 cells being captured onto the P-selectin-coated area at the shear stress of 0.25 dyn/cm 2. Some cells were observed to flow out of the field of view........49 Figure 2-5 The attachment distances of HL60 cells at the shear stress of 0.25 dyn/cm 2. P-selectin edge is located at 0 gin. The data obtained by analyzing 65 images acquired at 1 fps using a customized Matlab code........................................................49 Figure 2-6 Attachment distance of HL60 cells at different shear stresses of 0.25-1.0 dyn/cm 2. Representative results are shown for only one experiment for each shear stress. (a) Distribution of attachment distance at different shear stresses (0.25-1.0 dyn/cm 2 ). (b) Cumulative probability (CP) of attachment distance at different shear stresses (0.25-1.0 dyn/cm 2) .......................................................................................................... 51 Figure 3-1 Illustration of a typical cell rolling trajectory along the receptor pattern inclined an angle (a) to the fluid flow direction. The cell binds within the receptor line, and rolls in the direction of shear flow toward the pattern edge. The cell then tracks the edge to define an edge tracking length 4, resulting in a net lateral displacement d, before detaching to continue along the direction of fluid flow before possible reattachment and rolling along a new receptor line. Cell rolling velocity v, along within the receptorfunctionalized line in the x-direction of fluid flow can be distinguished from the velocity Ve along the line edge, where ve,yis lateral velocity (the vertical component perpendicular to the streamlines and parallel to the lateral displacement, d)........................61 Figure 3-2 Schematic diagram for patterning of P-selectin on a gold substrate involving microcontact printing. Step 1: Selective deposition of PEG molecules on the gold surface. Step 2: Filling in of the uncoated surface with P-selectin. .................. 62 Figure 3-3 Characterization of P-selectin patterned substrates. AFM images of 10 pm wide P-selectin lines separated by 15 gm wide PEG bands (after step 2), displaying63 Figure 3-4 Rectangular flow chamber for cell rolling on multiple asymmetric pattern study. The flow chamber was attached to the P-selectin-patterned substrate via a vacuum manifold to assemble the device. The thickness of the gasket determines the height of the chamber, here, height = 0.005 inch)...........................................................65 Figure 3-5 The interaction of HL60 cells with P-selectin/PEG patterned substrate in the flow chamber. The substrate was prepared using large line-patterned stamp (LS). The edge inclination angle was 100 and the shear stress was 0.5 dyn/cm 2 . The HL60 cells only interact with the P-selectin patterned region, not PEG patterned region, which confirms the anti-fouling property of PEG........................................66 12 Figure 3-6 Tracks of HL60 cells rolling on P-selectin patterns. The tracks (blue lines) on P-selectin lines (pink) were obtained by analyzing 300 images acquired at 1 fps using a customized Matlab code. The edge inclination angle was 100 and the shear stress was 0.5 dyn/cm 2 . Inset shows a track corresponding to a cell that first rolled inside the Pselectin line (green) in the direction of fluid flow and then tracked along the edge (black). ................... 68 ....... ............................ ......... Figure 3-7 Effect of edge inclination angle a on rolling behavior of HL60 cells at a fluid shear stress magnitude of 0.5 dyn/cm 2 . Variation of (a) edge tracking length, 1e; (b) lateral displacement, d; (c) rolling velocities v, and ve within the P-selectin lines and on the edge, respectively; and (d) lateral velocity, ve,,y (component of the edge rolling velocity in the direction of lateral displacement). Error bars represent one standard deviation, with n = 3 replicate experiments for each condition................................70 Figure 3-8 Effect of shear stress T on rolling behavior of HL60 cells at an edge inclination angle of 50. Variation of (a) edge tracking length, le; (b) lateral displacement, d; (c) rolling velocities v, and ve within the P-selectin lines and on the edge, respectively; and (d) lateral velocity, ve,,y. The effect of shear stress on rolling behavior of HL60 cells is not statistically significant as observed in (a), (b), (c) and (d). Error bars represent one standard deviation, where n = 3 replicate experiments for each condition...................71 Figure 3-9 Effect of P-selectin incubation concentration on rolling behavior of HL60 cells. Variation of (a) edge tracking length, le; (b) lateral displacement, d; (c) rolling velocities v, and ve within the P-selectin lines and on the edge, respectively; and (d) lateral velocity, vey at edge inclination angles a = 100 and 20' and shear stress of 0.5 dyn/cm2 . Error bars represent one standard deviation, where n = 3 replicate experiments .......... 73 for each condition................... ................................ Figure 3-10 Detachment of cells rolling along an edge is well described by a Poisson distribution. (a) Cumulative distribution function of edge tracking lengths le (filled triangles) was fitted to a Poisson distribution described by Eq. 3-1 (solid lines). Insets show the frequency distribution of the experimentally measured edge tracking lengths, along with that predicted by the Poisson distribution fit to the CDF (solid lines). Colors indicate different inclination angles a of the receptor pattern. Representative results are shown for only one experiment for each a. (b) Variation of the average value of k with edge inclination angle is well described by a linear fit on a semi-log plot (solid line). (c) Variation with the edge inclination angle of the average value of the lateral displacement (solid line) obtained from the empirical fit shown in (b) along with the experimental results (open circles). Error bars in (b) and (c) represent one standard deviation. Shear stress is 0.5 dyn/cm 2 . . . ............................ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76 Figure 3-11 Prediction of cell trajectories on a receptor-patterned substrate (a) Probability distribution of net lateral displacement of HL-60 cells after rolling on three consecutive bands of P-selectin patterns as obtained from Monte Carlo simulations (shaded area) and experimental observations. a = 200 (shear stress 0.5 dyn/cm 2 ). (b) Prediction of the downstream distribution of HL60 cells rolling on patterned P-selectin in 79 a separation device. .............................................................. 13 Figure 3-12 Optimization device design. (a) Schematic diagram of cell settling, capture, rolling on the patterns. (b) Schematic diagram of a device. Device capture efficiency, total lateral displacement, and required chamber length can be modeled based on different device design parameters.......................................................................... 81 Figure 3-13 (a) Schematic diagram showing cell hopping onto downstream band. (b) Variation of effective chamber length on total lateral displacement when hopping events were (p, = 1.0) and were not (p, = pf = 0.03) considered for the pattern design of a = 150, w = 10 tm, g = 10 pIm, and r= 0.25 dyn/cm2 .... . . . . . . . ..... 84 Figure 3-14 The relationship between device capture efficiency (Pd) and effective chamber length (xe) and the relationship between total lateral displacement (d,) and effective chamber length (xe) when w varies from 10 ptm to 50 pIm with a = 15*, g = 10 stm, and r = 0.25 dyn/cm2 ........ . .. ... ....... ........ ...... .. ..... .... . . . .85 Figure 3-15 The relationship between device capture efficiency (Pd) and effective chamber length (xe) and the relationship between total lateral displacement (d) and effective chamber length (xe) when a varies from 5 0 to 250 with w = g =10 pim, and r . . . . . . . . . . . . . . . . .. . . . . . . .. . . . . . . . . . . . . . . . . . 87 = 0.25 dyn/cm 2 . . . . . Figure 3-16 The relationship between device capture efficiency (Pd) and effective chamber length (xe) and the relationship between total lateral displacement (d) and effective chamber length (xe) when i-varies from 0.25 dyn/cm 2 to 1.0 dyn/cm 2 with a = 15 0 and w = g =10 pm.................................................89 Figure 3-17 Device optimization parameters including edge inclination angle a, band width w, gap width g, and shear stress T, for a miniaturized device of a chamber length of L, with device capture efficiency Pd, and total lateral displacement d,. x, is settling distance and xe is effective chamber length. ........... 91 Figure 4-1 Schematic diagram of M13 bacteriophage and the summary of coat protein.................................................... .......... ...................... ...................... ... 98 Figure 4-2 Schematic diagram for identifying peptide motifs of affinity to CD4 protein by biopanning using pVIII library. The pVIII library was exposed to the CD4coated well plate. Unbound phage was then washed off from the target and the bound phage was eluted from the surface by acid solution. The eluted phage was amplified to serve as the sub-library for the next round of the panning process. The peptide sequences of the eluted and amplified phage was identified by titering and DNA sequencing. The panning process was repeated until the consensus peptides were found. ....................... 101 Figure 4-3 Titration of phage on LB/IPTG/Xgal plates (dilution range is from 101 to 104). The phage was titered on LB/IPTF/Xgal plates and the blue plaque was then randomly selected for sequencing..................................... ..................................... 104 Figure 4-4 Schematic diagram of DNA extraction for sequence identification. DNAs of the phage was extracted from the bacteria. The bacteria were lysed and the DNAs were isolated from the lysed bacteria and then eluted using QlAprep Spin Miniprep Kit....106 14 Figure 4-5 Schematic diagram for immobilizing phage on a gold substrate. Step 1: Functionalization of DSP onto the gold surface. Step 2: Immobilization of phage on the reactive gold surface...............................................................110 Figure 4-6 Surface characterization of phage-engineered substrates. AFM height images of phage immobilized on the gold-coated substrate. All the images are 1 Om X 1Opm....................................................................................-----------------.......................112 Figure 4-7 Fluorescent microscope images of the cells on the RK1-engineered gold substrate (A) before the flow was started (B) after the flow was started for 60 s. The white dots are stained K562 cells and black dots are non-stained SUPTI cells.............114 Figure 4-8 Capture efficiencies of RK1-RK12-immobilized substrates at shear stress of 0.1 dyn/cm 2. Error bars represent one standard deviation, where n = 4 replicate experiments for each condition......................................................115 Figure 4-9 Capture efficiencies of RK1-immobilized substrates. The effect of different shear stress on the capture efficiencies of the cells on the RK1-immobilized substrates. Error bars represent one standard deviation, where n = 4 replicate experiments for each condition................................................116 Figure 4-10 Microscope images of the SUPT1 cells captured on the CD4-blocked RK1-engineered gold substrate (a) before the flow was started (b) after the flow was started for 60 sec..............................................................................117 Figure 5-1 (a) CD4 protein physically adsorbed on regular polystyrene surface. (b) His-tagged CD4 protein immobilized on Ni (II)-NTA-coated surface with specific orientation.....................................................-------------------------------------------------------------..124 Figure 5-2 Schematic diagram for identifying peptide motifs of affinity to CD4 by biopanning using pIII library. The pIII library was exposed to the CD4-coated well plate. Unbound phage was then washed off from the target and the bound phage was.. 126 Figure 5-3 Schematic diagram of ELISA procedure. CD4-His protein was immobilized on Ni (II) coated well plate. The phage was then exposed to the CD4-coated well plate. HRP-conjugated anti-M13 antibody binds specifically to the target (pVIII protein of the phage). Several washing steps were repeated between each ELISA step to remove unbound materials. Substrate (TMB) was added and the colored end product from enzyme-substrate reaction is produced. The color signal correlates to the amount of ... 134 the phage present in the well plate. ..................................................................... Figure 5-4 ELISA assay output. (a) Qualitative results. Representative results are shown for only one experiment at each condition. Wells in column 1, 3, and 5 were coated with CD4 and then BSA. Wells in column 2, 4, and 6 were coated with only BSA as control. Phage with different input concentration were added. Phage AMB1 were incubated in column 1 and 2. Phage AMB2 were incubated in column 3 and 4. WT phage was incubated in column 5 and 6; (b) Quantitative results. The relationship between the absorbance of the colored end product and the input concentration of the phage. Error 15 bars represent one standard deviation, with n = 3 replicate experiments for each condition.......................--...........................-- - -. -----............................................... 136 Figure 5-5 Reaction of the Alexa Fluor@ 488 C5 Maleimide with a thiol-peptide. .137 Figure 5-6 Collected drained samples. The first five fractions were collected before the dye-conjugated peptide solution drained through and thus contained mostly buffer (colorless). The orange color came from dye-conjugated peptide. The non-conjugated dye shows yellow color ....... - . .... ... -........ ............................................... 138 Figure 5-7 Schematic diagram for staining the CD4-coated beads with dyeconjugated peptide. The Ni(II) beads were incubated with CD4-His and AF 647 conjugated BSA, separately. The beads were then mixed and incubated with BSA to block non-specific interaction. The mixed CD4 and AF 647-B SA-coated beads were then incubated with AF 488-conjugated peptide. Several washing steps were repeated between each step to remove unbound materials before the samples were imaged......................140 Figure 5-8 Fluorescence microscope analysis. (a-b) CD4-immobilized beads. Images of (a) bright field and (b) FITC channel. CD4-immobilized beads do not show autofluorescence. (c-d) BSA-coated beads that were incubated with AF 488 dyeconjugated peptides. Images of (c) bright field and (d) FITC channel. AF 488 dyeconjugated peptides do not show non-specific binding to BSA-coated beads. (e-f) The mixture of the CD4-immobilized beads and the AF 647 dye-BSA-coated beads stained with AF 488 dye-conjugated peptides. Images of (e) bright field; (f) Cy5 channel; and (g) FITC channel; (h) is the Cy5/FITC channel merged image. AF 488 dye shows bright, green fluorescence and AF 647 shows bright, red fluorescence. Beads number 1, 2, 6, and 11 were coated with BSA and beads number 3, 4, 5, 7, 8, 9, and 10 ........................ ..............---------------............................ ..................... 142 Figure 6-1 Experimental design for characterizing hopping behavior of cells. Cells flow over parallel receptor bands with different gap widths aligned perpendicular to flow direction. The minimum gap width without making cell cross over the patterns can be identified................. ................ ............. ....... ..... .. 147 16 List of Tables Table 1-1 Kinetics and affinity of selectin-ligand and antibody-antigen interactions .................................................... 23 ..................................... Table 2-1 Comparison of the average values and standard deviation of A and <Ia> 2 obtained from curve fitting at different shear stresses of 0.25-1.0 dyn/cm . n = 3 replicate experiments. Data were obtained by fitting to two empirical equations with 51 experimentally measured attachment distance ............................................................. Table 3-1 Comparison of the experimental average of 1e, Poisson average value of k, and the Poisson mean value ' from the empirical fit. n = 3 replicate experiments.... 76 Table 3-2 Prediction of required effective chamber lengths (xe). The range of xe which target at Pd - 90%, dt : 850 ptm and both criteria (Pd > 90% and d, ? 850 Pim)2 when w varies from 10 pm to 50 pin with a = 150, g = 10 ptm, and T = 0.25 dyn/cm . ............................................... ........ .......................------------ 86 Table 3-3 Prediction of required effective chamber lengths (xe). The range of xe which target at Pd ! 90%, d, ? 850 ptm and both criteria (Pd > 90% and d, f 850 pim) 2 ... . ..... . . ... .... 87 when a varies from 5 0 to 254 with w = g =10 pim, and r =0.25 dyn/cm Table 3-4 Prediction of required effective chamber lengths (xe). The range of xe which target at Pd !90%, dt ? 850 gim and both criteria (Pd > 90% and d, ? 850 stm) when r varies from 0.25 dyn/cm 2 to 1.0 dyn/cm 2 with a = 151 and w = g =10 pm.....89 Table 4-1 Input and output concentration of phage and the recovery of each round of screening. The concentration is defined as the number of the phage per well. For each round of screening, four wells coated with CD4 were used...................................107 Table 4-2 CD4 protein binding sequences from biopanning of pVIII library.........107 Table 4-3 Selected peptide sequences and their properties. (negatively charged amino acids in red; positively charged amino acids in blue; polar amino acid without charged in yellow; hydrophobic amino acid in gray; aromatic amino acids in green; Tyrosine in 109 ...---------------------------............................ ............................ orange)................................ Table 4-4 Peptide sequences from biopanning of pVIII library. (96 samples were o........................117 ----------------------.................. selected).................. Table 5-1 Binding affinity (dissociation constant (KD) of oligo-histidine peptides to -- - -- -.... 125 Ni(II)....................................................................--- 17 Table 5-2 Input and output concentration of phage and the recovery of each round of screening. The concentration is defined as the number of the phage per well. For each round of screening, four wells coated with CD4 were used............................................127 Table 5-3 CD4 protein binding sequences from biopanning of pIII library............128 Table 5-4 Selected peptide sequences and their properties. (negatively charged amino acids in red; positively charged amino acids in blue; polar amino acid without charged in yellow; hydrophobic amino acid in gray; aromatic amino acid in green; Histidine in orange).............................................................................................................................130 18 1 1.1 Introduction Background and Motivation Separation and analysis of heterogeneous cell populations are important for diagnostic and therapeutic applications, and for elucidating the biology of rare cell types. Numerous cell-sorting techniques have been developed, with sorting characteristics based on physical properties of the cells, such as size, density, and deformability [1-4]. However, these physical separation methods may not be sufficiently specific enough to distinguish similar cell types. Surface maker-based cell separation/analysis techniques allow cells to be distinguished via biochemical differences (protein expression), and can confer higher specificity. Some standard techniques include fluorescence-activated cell sorting (FACS), magnetic-activated cell sorting (MACS), and cell affinity chromatography (CAC). Among these, FACS or flow cytometry has been the gold standard of cell separation for years. However, sophisticated equipment, and time-intensive and labor-intensive sample preparation processes limit the use of most methods. For example, white blood cell counts and leukocyte differential are routinely used in clinical diagnostics [5]. Blood counts are usually performed using automated counters to identify major blood cell types and separation of subgroups of immune cells are done through FACS [5]; however, flow cytometry is typically not available in the situation with resource-limited settings. Also, transportation of clinical samples from patients to labs and multiple sample processing steps are time-consuming and labor-intensive. Microfluidic devices offer a promising platform for cell separation in which all of the processes can be compacted in one single step. Small amount of samples required and portability of microfluidic devices facilitate the application in point-of-care testing. However, most of surface-marker-based microfluidic devices use antibodies to capture cells with specific antigens and therefore separate cells. The antibodies with high affinity to their antigen limit the usability of these microfluidic devices since retrieval of the captured cells is difficult. Therefore, there exists a need for developing techniques of rapid cell separation based on specific surface markers but weak adhesion which will allow cells to be retrieved and will be useful for many applications. A number of molecules in nature exhibit weak, but relatively specific adhesive interactions. For example, selectins mediate leukocyte rolling 19 which is a process in inflammatory or immune responses [6-9]. Selectin molecules-based separation has been studied before; however, it was limited to a batch process [10]. Karnik et al have demonstrated that patterning of selectin molecules inside the microfluidic device create the possibility of continuous sorting of cells by transient, weak interaction between cell and patterned selectin molecules [11]. Comprehensive studies of cell settling, cell attachment, pattern geometry, flow condition, and selection of adhesion molecules are needed in order to design and realize these devices. 1.1.1 Surface Marker-Based Cell Separation Surface markers are typical proteins expressed on the surface of cells. Cluster of differentiation (CD) molecules are a group of surface markers mostly expressed on leukocytes and associated with immune function or properties. The cells can be defined based on which CD molecules expressed on their surface. Using multiple CD markers allows the cells to be defined more specifically. For most of existing surface markerbased cell sorting techniques, the surface cluster of differentiation proteins (antigens) are identified via binding of conjugate proteins (antibodies). An important advantage of surface marker-based cell separation is that it can be used for sorting the cell populations of the same size or density. For example, CD4 and CD8 proteins are well known surface markers for helper T lymphocytes (T cells) and suppressor/cytotoxic T cells, respectively[12]. The diagnosis of HIV disease relies on the efficient separation of human CD4+ T lymphocytes (T cells) from whole blood. The most general way to distinguish CD4+ T cells and CD8+ T cells is to identify them by antibodies that can specifically bind to CD4 or CD8 proteins expressed on the T cells. The antibodies can be conjugated to fluorescent molecules or magnetic beads, or can be immobilized onto columns or microchips. The methodologies or operation procedures will differ, depending on the materials to which the antibodies are conjugated or immobilized. The most common surface marker-based technologies are: fluorescence-activated cell sorting (FACS), magnetic-activated cell sorting (MACS), cell affinity chromatography (CAC), and antibody-based microfluidic devices (ABM) [1-3, 13], which are outlined below. 20 Fluorescence-Activated Cell Sorting (FACS). FACS is the gold standard cell subpopulation separation technique [14, 15]: Typically, specific cell surface markers of the cells labeled by antibodies. Those antibodies are labeled with fluorescent molecules and the cells are then identified by fluorescent intensity corresponding to the number of antigens on each cell. By collecting the information such as fluorescent intensity of each cell, the machine can identify and then separate cells of different antigen surface density. FACS has several technical advantages, such as single-cell-level sensitivity, high throughput (~10,000 cells/sec) of separation/counting, and ability to track four to five different cell surface markers at the same time [15]. However, the capital investment and maintenance of equipment, and the multiple sample preparation steps, hinder the applications in point-of-care tests or in resource-limited settings environment. Furthermore, cell viability after ejection from the machine, as well as the potential need to remove antibody labels before human use of such sorted cells, remain major issues, that limit the use of FACS in clinical or translational applications [14, 15]. Magnetic-Activated Cell Sorting (MACS). Similar to FACS, in MACS, the surface markers of the cells are recognized by the antibodies. These antibodies are coated onto the surface of magnetic nanoparticles [16]. The cells can be sorted by applying a strong magnetic field that collects bead-bound cells of higher antigen density or affinity. Compared with FACS, by which the cells are sorted one by one, MACS can separate the cells in serial (continuous mode) or in parallel (batch mode), resulting in higher throughput (more than 1011 cells per hour [2, 13]). Another advantage is that cells can be sorted positively or negatively depending on whether targeted or nontargeted cells are labeled. However, there are several drawbacks for this method, such as the timeconsuming, sensitive process of labeling cells onto the antibodies-coated magnetic beads, complications of removing magnetic labels, and difficulty of sorting more than two cell populations at the same time. Cellular Affinity Chromatography (CAC). Affinity chromatography has been widely used for separating biological molecules, such as peptides, proteins and enzymes [17-22]. Affinity chromatography can also be applied in cell separation by using different column 21 design [23-26]. Cells can be sorted by being passed via fluid flow through a column packed with antibody-coated beads or other solid supports. Cells with affinity to the immobilized antibody will be to be selectively retained; other cells will be passed through unretarded. One advantage of CAC is that the sample preparation is simple. For example, there are no complicated processes of labeling cells with fluorescent dye or magnetic beads [1]. However, it is often difficult to release the bound cells from the beads; elution is typically achieved by applying high shear flow or adding competitive agents [27]. Adhesion-based Microfluidics Devices for Cell Sorting (ABM). Similar to CAC, sorting of the cells by adhesion-based microfluidics devices is achieved by binding cells of interest onto the devices where the specific molecules are immobilized. The adhesive molecules (usually antibodies) can recognize the surface markers present on the surface of the cells. The separation can be completed in a single processing step. The untargeted cells are allowed to flow through without interacting with the adhesive molecules, while the targeted cells are captured inside the chip. The sensitivity of this method is enhanced because of the larger surface area to volume ratio in a microscale chip than in macroscale setup. Moreover, the residence time inside the microchips is shorter than that of CAC. The low-cost microchips provide advantages of flexibility for point-of-care diagnostics. However, the difficulty in retrieving the captured cells makes analysis and enumeration sorted cells difficult. Most major surface marker-based cell separation techniques involve using antibodies that target corresponding antigens expressed on the cell surfaces. However, removal of the labels without changing cellular functions and phenotypes is one of the major issues that hinder the use of these methods. Additionally, these methods are then limited to isolation of cells with distinct antigens and for which purified antibodies are readily available. There exists a growing need for technologies that do not require modification or labeling of the sorted cells. Receptors with high specificity but weak affinity resulting in transient interaction with conjugate molecules on the cells may be used to fulfill the requirement. 22 1.1.2 Low Affinity Based Separation It has been widely studied that typical antibody-antigen interactions show high affinity where on-rate (k,,) is in the rage of 10'~108 M-'s-1 , off-rate (korr) is in the rage of 102 _10-4 s-1 and dissociation constant (Kd) is in the range of 0.3~1.5 pM [28-30]. The high affinity property results in firm adhesion between antibody and antigen, which limits the usability of these microfluidic devices since retrieval of the captured cells is difficult. In order to develop microfluidic devices that allow cells to be retrieved, adhesion molecules with weak but specific interaction is needed. In nature, a number of molecules exhibit unique kinetics and affinity that allows the cells to be specifically identified, but weakly captured. For example, selectins and their ligands mediate inflammatory or immune responses [6-9] and allow leukocytes to be captured from the blood stream and roll on the vascular endothelium. There are three types of selectins: (1) P-selectin (CD62P), (2) Eselectin (CD62E), and (3) L-selectin (CD62L) [6, 31-33]. Table 1-1 lists kinetics and affinities of selectin-ligand and antibody-antigen interactions [34-36]. Compared with typical antibody-antigen interaction, the moderate ko. and high koff of the selectin-ligand interactions suggest that the adhesive bond can be formed and be broken quickly and therefore the cells of ligand molecules can roll on the receptor molecules. Using this weak but specific adhesion opens possibility of separating cells without labeling and allowing cells to be retrieved. Table 1-1 Kinetics and affinity of selectin-ligand and antibody-antigen interactions Receptor and Ligand kon(M 1 s1) koff(s-) Ka(M 1 Kd(PM) ) 6 Reference P-selectin /PSGL1 (0.93 - 4.4)x10 6 1.4 L-selectin /PSGL1 2x10 5 10 2x10 4 50 [35] E-selectin /ESL1 7.4x10 4 4.6 1.6x10 4 62 [36] Antibody/Antigen 10 ~ 108 10-2 10-4 23 0.66-3.1x10 107 _ 1012 1.5 0.3 10-6 101 [34, 35] [28-30] 1.1.2.1 Cell Rolling Cell rolling is mainly observed as one of the steps in recruiting leukocytes from circulating blood into the sites of inflammation, as shown in Figure 1-1 [37]. Rolling is initiated first, followed by integrin-dependent adhesion and transmigration to the site of inflammation. Cell rolling involves transient interactions between the leukocyte and the vascular endothelium (blood vessel lining) under fluid shear in a physiological process. The interaction is mediated by glycoprotein receptors known as selectins and their ligands. Rolling mechanism also plays an important role in homing of hematopoietic stem cells and metastasis of tumor cells [6-9]. Capture Rolng salyl Lewis X Integrins ITuimin Selectins ICAM-1 VCAM- PECAM VAP- Figure 1-1 Multistep leukocyte adhesion cascade in response to inflammation. Selectins initiate capture and rolling of the leukocytes. Integrins are involved in adhesion and cause the cells to arrest and transmigrate into the underlying tissues [37]. Selectin Family. Selectins including P-selectin, E-selectin and L-selectin and their ligands mediate leukocyte rolling interaction [6, 31-33]. P-selectin molecules are expressed by endothelial cells and platelets [38]. E-selectin molecules are expressed on endothelial cells and L-selectin molecules are expressed on leukocytes. Figure 1-2 lists selectins and their major ligands [6]. The major motifs of selectin molecules are: Ca+2_ dependent lectin domain, epidermal growth factor (EGF) like domain, short consensus repeat domain (SCR), transmembrane motif, and a short intracellular cytoplasmic tail as shown in Figure 1-2. Each selectin molecule mediates adhesion of rolling cells through the interaction of its lectin domain with sialyl Lewis x (sLex) ligand in the presence of extracellular Ca . Several posttranslational modifications (glycosylation and sulfation) 24 of the sLex motif have been identified for being able to preferentially bind to specific type of selectin [31]. For example, attachment of sLeX to the O-linked glycan of the Pselectin glycoprotein ligand-1 (PSGL-1) and the sulfation of N-terminal tyrosine in PSGL-1 are required for P-selectin binding [39]. Aeftated Plaftiet L -- P-selecin TELOkct ewPSGL-1 Actb*a&W ondotheftl eN LeuM"cye PSGL-1 P-selectin CD4 E-selactin - Un~ F %ESL-1 I WicllLu At .1aiW GIyCAM-1 PNAd 4m4 - MoYte L-eelectln CD34 L.Podocalyxin Laeukocy" L-eFeadn ==4PSGL-1 Lectin domain OEGF domain COnSMnSuS rmpt E GNAc M GNAC @ Gal A Fuc * SIS SSO fNr-9ycan I O-ycan Figure 1-2 Selectins and their major ligands. Three selectin family members (P-, E-, and L-selectin) and their corresponding ligands are shown in this figure. These three 25 selectin molecules share similar structure: N-terminal Ca+2-dependent lectin domain, epidermal growth factor like domain, short consensus repeat domain (SCR), transmembrane domain, and a short intracellular cytoplasmic tail (last two parts are not highlighted here). CD, cluster designation; EGF, epidermal growth factor; ESL-1, Eselectin ligand-1; GlyCAM-1, glycosylated cell adhesion molecule-1; GalNAc, Nacetylglucosamine; Gal, galactose; Fuc, fucose; Sia, sialic acid. [61 The force exerted from the flow on the cells may affect the lifetimes of selectin-ligand bonds by changing their off-rates. Bond lifetime of a receptor-ligand bond may increase (catch bonds) or decrease (slip bonds) [6, 40-45]. Lifetime is equal to the reciprocal of off-rate, l1/kff. The catch-to-slip bond transition under increasing forces has been observed experimentally for PSGL-l/ P-selectin and PSGL-l/ L-selectin as shown in Figure 1-3 [6, 40, 41, 46]. The external force has a biphasic effect on selectin-ligand interactions: the lifetime increases (catch bond) first until it reaches a maximal value and then decreases (slip bond). The catch bond behavior was observed over wider range of external force for L-selectin (up to 50 pN per bond) compared with PSGL-l/ P-selectin (up to 20 pN per bond) [40, 41]. In contrast, only slip bonds were observed for selectin/antibody interaction under external force [40, 41]. 05 0.40.4 - E P-selectln/PSGL-1 *# 03- 0.2 L-selectin/PSGL-1 0.05 - 0 0.4 0.8 1.2 1. 2 Wall shear stress (dyn cm- 2 ) Figure 1-3 Force-dependent lifetimes of single bonds of PSGL-1/ P-selectin (blue) and PSGL-1/ L-selectin (purple) under different shear stress. The catch-to-slip bond transition under increasing forces has been observed: the lifetime increases (catch bond) first until it reaches a maximal value and then decreases (slip bond). [61 26 Contributions to Rolling Motion. Cell rolling is a complex phenomenon involving multiple mechanical forces ranging from nanoscale to microscale [46]. For example, kinetics of receptor and ligand interaction [47], clustering of receptors [48, 49], elongation of microvilli [47, 48, 50-52], deformation of the cells [53], and the fluid forces acting on the cells [54, 55] can all perturb cell adhesion durations. Cellular properties that modulate cell rolling. It has been demonstrated that selectin(or selectin ligand-) coated microspheres can roll on the substrate immobilized with selectin ligand (or selectin). As those microspheres do not exhibit other biological features of cell rolling (microvilli extension, cell deformability and ligand cluster which will be discussed later), this suggests that molecular properties of selectin and its ligand are sufficient enough for supporting rolling [56-58]. We note that, compared to such beads, cells are deformable. Deformability of the cells enlarges the contact area between the bottom of the rolling cells and the endothelial cells, which results in a higher number of selectin-ligand bonds [59]. Additionally, the surfaces of cells are highly irregular. As compared to polymeric beads, for example, neutrophils (the most abundant leukocytes in the peripheral blood of human) have multiple microvilli and it has been observed that PSGL-1 and L-selectin are localized on the tip of microvilli on neutrophils as shown in (Figure 1-4(a)) [60, 61]. The bond cluster on the microvilli enhances tethering by increasing the contact with the receptors on the endothelial cells [62]. Tethers extend under shear flow by stretching microvilli (Figure 1-4(b) and (c)) [48] and the force on selectin-ligand bonds can be reduced by stretching tethered mivrovilli [63]. Most of the tethers retract after the selectin-ligand bond breaks [50]. 27 Figure 1-4 Scanning electron microscope (SEM) images of neutrophils. (a) Localization of PSGL-1 on microvilli. A human neutrophil was incubated with a mixture of the anti-PSGL-1 antibodies, followed by a rabbit anti-mouse antibody, and then with protein A-gold of 5 nm diameter. The arrow shows the gold nanoparticles (b) low magnification of tethered neutrophils. The neutrophil cell has multiple projections named microvilli on its surface. (c) high magnification of tethered neutrophils. The neutrophil was rolling on P-selectin under shear stress 2 dyn/cm 2 . The arrow shows a tether. Scare bars are all referred to 1 pim. (Images are adapted from [48], and [60].) 1.1.2.2 Cell Rolling-Based Separation Several groups have attempted to mimic physiological process of leukocyte rolling to develop cell separation techniques [64-70]. In contrast to conventional high affinity- antibody-based methods, separation based on immobilized receptors involved in cell rolling obviates labeling and label removal steps, enabling facile elution with minimized alteration of viability or function post-sorting [64-66]. For example, Chang et al. have demonstrated E-selectin-coated micropillar arrays for partially fractionating two cell types (HL60 and U937) based on the difference in transient interactions with the receptors [64]. Choi at al. also demonstrated that HL60 cells can be separation from K562 cells by using three dimensional P-selectin-coated inclined microstructure which promotes rolling of HL60 cells in the lateral direction [68]. Karnik et al. have demonstrated the transient interaction of HL60 cells and an inclined P-selectin band as shown in Figure 1-5 [11]. The P-selectin molecules were coated on the substate where the 28 edge of P-selectin area was inclined. The rolling cells can be displaced orthogonally to the direction of the flow, which opens new possibilities for separation of cells by diverting the rolling direction of cells. (a) (b)j Figure 1-5 Effect of a receptor pattern on the motion of a rolling cell. Tracks of HL60 cells rolling on patterned P-selectin edge show that patterning can direct the trajectories of rolling cells along the edges (only long tracks are shown on the right). [11] Figure 1-6 shows the concept of a device that adapts this observation, based on controlling the trajectories of the rolling cells along the edges of multiple receptor bands [70]. On the bottom of the device, multiple inclined, asymmetric adhesive bands are coated. The interacting cells from the continuously flowing sample can be captured on the adhesive bands and then roll on them. The asymmetry of the band orientation with respect to the flow direction alters the trajectory of the interacting cells, such that they track along the edge of the band, get displaced into the buffer stream and get sorted out. The non-interacting continue with the flow trajectory and stay in the sample stream. Such devices design with biomimetic adhesive substrates enables cell separation in a simple, continuous-flow, label-free manner. To realize the devices, a comprehensive study of interaction of cell and substrate is required. Cell settling and attachment onto substrates, the effect of substrate parameters on separation potential, and selection of adhesion molecules for the substrates will be discussed in this thesis later in order to maximize separation potential of cell rolling microfluidic devices. 29 Buffer Cells #Sortedeeel (interacting cells) Aoo Cells Adhesive substrate %ee **:Other Cells (non interacting cells) Figure 1-6 Schematic diagram of a device for separation of cells. Cells rolling along band edges are laterally displaced into the adjacent buffer stream, resulting in separation. Pink lines indicate adhesion molecule-functionalized regions. Red and blue circles are interacting cells and non-interacting cells, respectively. Since cell rolling phenomenon is exhibited in vivo or in vitro by several types of cells including leukocytes, hematopoietic stem cells, and some cancer cells [6-9], those types of cells have potential to be separated by steering them on biomimetic adhesive bands using the device as described in Figure 1-6. Such devices can be suitable for point-of care applications, especially in non-industrialized settings, which require device simplicity and minimized cell processing steps. Furthermore, continuous flow sorting can promote separation efficiency and selectivity. Another advantage is that observation of cellsurface interactions on immobilized receptors offers the opportunity for label-free enumeration of cells [71]. Separation methods for therapeutics or subsequent analysis of cells need to preserve the cell phenotype and such devices allow invasive separation that can promise for these applications. Cell rolling separation devices with multiple adhesive bands also offer potential for direct analysis of the cell phenotype or receptor expression. In addition, co-immobilization of other cell type-specific molecules along with selectins can significantly influence cell rolling [72, 73], which opens the possibility of further tuning the specificity of separation and analysis based on the expression of a pertinent receptor on the cell. On the other hand, although directing rolling trajectories of cells by patterning of weak adhesion molecules onto the microfluidic device is promising for label-free cell sorting, this technology is limited to the molecules known for enabling cell rolling. To apply this technique to general surface markers, new receptors other than the existing molecules need to be identified. Biopanning is an affinity selection technique 30 used for identifying peptides that bind to a target of affinity within micromolar range [74], which potentially can be used to identify new receptor with weak adhesion to the cells. The possibility of using biopanning to select receptors for cell separation will be studied in this thesis. 1.2 Scope of the Thesis Work To successfully develop rolling adhesion microfluidic devices that can deflect cells by inclined receptor bands as shown in Figure 1-6, a biomimetic adhesive substrate is the key. Therefore, the goal to this thesis work is to develop biomimetic adhesive substrates for cell separation in microfluidic devices. My three main aims are: - Aim 1: To understand transport and attachment of cells to biomimetic adhesive substratesin microfluidic channels. Chapter II discusses how the cells settle and how they are captured by receptor functionalized surface under fluid shear flow. In Part A, to quantify cell settling, settling distance under different shear stresses is characterized from the study of convection velocity of cells at different position along the channel. In part B, to quantify cell capture, cumulative probabilities of cell attachment distance and average attachment distance of cells at different shear stresses is analyzed. An empirical model is developed to predict capture probability by an inclined receptor band. - Aim 2: To investigate effects of biomimetic adhesive substrate parameters on cell separationpotential In Chapter III, a patterning method involving microcontact printing is developed to study how cell transport can be controlled on inclined receptor bands. The effects of edge inclination angle with respect to shear flow direction (a = 50, 100, 150 and 20'), shear stress magnitude (r = 0.5, 1, 1.5 and 2.0 dyn/cm 2 ), and P-selectin incubation concentration (15, 20, and 25 ptg/mL) are quantified in terms of the edge tracking length, lateral displacement, and rolling velocity. A methodology is developed to predict device performance to enabled successful design of separation device. 31 - Aim 3: to extend this approach to other and novel cell surface markers Beyond the PSGL-1 on leukocytes, this adhesive band patterning approach may be extensible to tether cell surface targets, such as CD4 proteins, that is indicative of disease state. M13 pVIII (Chapter IV) and pIll (Chapter V) phage libraries are used for selecting peptides with affinity to CD4 proteins. In Chapter IV, selected phage is immobilized on the gold surface and capture efficiency of CD4+ cells is characterized. The interaction between selectin phage and CD4+ cells is demonstrated to be CD4-dependent. In Chapter V, specificity of selected phage clone and synthetic peptides to CD4 proteins is confirmed. 1.3 Chapter Reference [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] M. Radisic, R. K. Iyer, and S. K. Murthy, Micro- and Nanotechnology in Cell Separation.Int J Nanomedicine, 2006. 1(1): p. 3. A. A. Bhagat, H. Bow, H. W. Hou, S. J. Tan, J. Han, and C. T. Lim, Microfluidics for Cell Separation. Med Biol Eng Comput, 2010. 48(10): p. 999. M. J. Tomlinson, S. Tomlinson, X. B. Yang, and J. Kirkham, Cell Separation: Terminology and Practical Considerations. J Tissue Eng, 2013. 4: p. 2041731412472690. D. R. Gossett, W. M. Weaver, A. J. Mach, S. C. Hur, H. T. Tse, W. Lee, H. Amini, and D. Di Carlo, Label-Free Cell Separation and Sorting in Microfluidic Systems. Anal Bioanal Chem, 2010. 397(8): p. 3249. R. A. McPherson, M. R. Pincus, and J. B. Henry, Henry's ClinicalDiagnosisand Management by Laboratory Methods. 21st ed, 2007, Philadelphia: Saunders Elsevier. p. p. R. P. McEver and C. Zhu, Rolling Cell Adhesion. Annu Rev Cell Dev Biol, 2010. 26: p. 363. P. J. Quesenberry and P. S. Becker, Stem Cell Homing: Rolling, Crawling, and Nesting. Proc Natl Acad Sci U S A, 1998. 95(26): p. 15155. R. Giavazzi, M. Foppolo, R. Dossi, and A. Remuzzi, Rolling and Adhesion of Human Tumor Cells on Vascular Endothelium under Physiological Flow Conditions. J Clin Invest, 1993. 92(6): p. 3038. T. Lapidot, A. Dar, and 0. Kollet, How Do Stem Cells Find Their Way Home? Blood, 2005. 106(6): p. 1901. A. D. Hughes, J. Mattison, L. T. Western, J. D. Powderly, B. T. Greene, and M. R. King, Microtube Device for Selectin-Mediated Capture of Viable Circulating Tumor Cellsfrom Blood. Clin Chem, 2012. 58(5): p. 846. 32 [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] R. Karnik, S. Hong, H. Zhang, Y. Mei, D. G. Anderson, J. M. Karp, and R. Langer, Nanomechanical Control of Cell Rolling in Two Dimensions through Surface PatterningofReceptors. Nano Lett, 2008. 8(4): p. 1153. T. A. Springer, Adhesion Receptors of the Immune System. Nature, 1990. 346(6283): p. 425. A. Thiel, A. Scheffold, and A. Radbruch, Immunomagnetic Cell Sorting--Pushing the Limits. Immunotechnology, 1998. 4(2): p. 89. W. A. Bonner, H. R. Hulett, R. G. Sweet, and L. A. Herzenberg, Fluorescence Activated Cell Sorting. Rev Sci Instrum, 1972. 43(3): p. 404. L. Bonetta, Flow Cytometry Smaller and Better. Nature Methods, 2005. 2(10): p. 785. S. Miltenyi, W. Muller, W. Weichel, and A. Radbruch, High GradientMagnetic Cell Separationwith Macs. Cytometry, 1990. 11(2): p. 231. P. Cuatrecasas, M. Wilchek, and C. B. Anfinsen, Selective Enzyme Purificationby Affinity Chromatography.Proc Nati Acad Sci U S A, 1968. 61(2): p. 636. A. H. Ross, D. Baltimore, and H. N. Eisen, Phosphotyrosine-ContainingProteins Isolated by Affinity Chromatography with Antibodies to a Synthetic Hapten. Nature, 1981. 294(5842): p. 654. M. C. Smith, T. C. Furman, T. D. Ingolia, and C. Pidgeon, Chelating PeptideImmobilized Metal Ion Affinity Chromatography. A New Concept in Affinity ChromatographyforRecombinantProteins. J Biol Chem, 1988. 263(15): p. 7211. H. Gadgil, S. A. Oak, and H. W. Jarrett, Affinity Purification of DNA-Binding Proteins.J Biochem Biophys Methods, 2001. 49(1-3): p. 607. C. Tozzi, L. Anfossi, and G. Giraudi, Affinity ChromatographyTechniques Based on the Immobilisation of Peptides Exhibiting Specific Binding Activity. J Chromatogr B Analyt Technol Biomed Life Sci, 2003. 797(1-2): p. 289. S. A. Camperi, N. B. Lannucci, G. J. Albanesi, M. Oggero Eberhardt, M. Etcheverrigaray, A. Messeguer, F. Albericio, and 0. Cascone, Monoclonal Antibody Purificationby Affinity Chromatographywith Ligands Derivedfrom the Screening of Peptide Combinatory Libraries. Biotechnol Lett, 2003. 25(18): p. 1545. K. Berg, C. A. Ogburn, K. Paucker, K. E. Mogensen, and K. Cantell, Affinity Chromatography of Human Leukocyte and Diploid Cell Interferons on Sepharose-BoundAntibodies. J Immunol, 1975. 114(2 Pt 1): p. 640. M. M. Baran, D. M. Allen, S. R. Russell, M. E. Scheetz, 2nd, and J. F. Monthony, Cell Sorting Using a Universally Applicable Affinity Chromatography Matrix: Solid-Phase Anti-Fluorescein Isothiocyanate Antibody. J Immunol Methods, 1982. 53(3): p. 321. C. M. Hertz, D. J. Graves, D. A. Lauffenburger, and F. T. Serota, Use of Cell Affinity Chromatography for Separation of Lymphocyte Subpopulations. Biotechnol Bioeng, 1985. 27(5): p. 603. A. Kumar and A. Srivastava, Cell Separation Using Cryogel-Based Affinity Chromatography.Nat Protoc, 2010. 5(11): p. 1737. E. Mandrusov, A. Houng, E. Klein, and E. F. Leonard, Membrane-Based Cell Affinity Chromatographyto Retrieve Viable Cells. Biotechnol Prog, 1995. 11(2): p. 208. 33 [28] [29] [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] S. Lin, S.-Y. Lee, C.-C. Lin, and C.-K. Lee, Determination of Binding Constant and Stoichiometry for Antibody-Antigen Interaction with Surface Plasmon Resonance. Current Proteomics, 2006. 3(12): p. 271. C. A. Borrebaeck, A. C. Malmborg, C. Furebring, A. Michaelsson, S. Ward, L. Danielsson, and M. Ohlin, Kinetic Analysis of Recombinant Antibody-Antigen Interactions: Relation between Structural Domains and Antigen Binding. Biotechnology (N Y), 1992. 10(6): p. 697. S. Hearty, P. Leonard, and R. O'Kennedy, MeasuringAntibody-Antigen Binding Kinetics Using Surface Plasmon Resonance. Methods Mol Biol, 2012. 907: p. 411. S. M. Lucila and U. H. von Andrian, Immunological Adhesion and Homing Molecules, in Els200 1, John Wiley & Sons, Ltd. L. A. Lasky, Selectin-Carbohydrate Interactions and the Initiation of the Inflammatory Response. Annual Review of Biochemistry, 1995. 64: p. 113. R. P. McEver, Selectins: Lectins That Initiate Cell Adhesion under Flow. Curr Opin Cell Biol, 2002. 14(5): p. 581. P. Mehta, R. D. Cummings, and R. P. McEver, Affinity and Kinetic Analysis ofPSelectin Binding to P-Selectin Glycoprotein Ligand-1. J Biol Chem, 1998. 273(49): p. 32506. A. G. Klopocki, T. Yago, P. Mehta, J. Yang, T. Wu, A. Leppanen, N. V. Bovin, R. D. Cummings, C. Zhu, and R. P. McEver, Replacing a Lectin Domain Residue in L-Selectin Enhances Binding to P-Selectin Glycoprotein Ligand-1 but Not to 6Sulfo-Sialyl Lewis X J Biol Chem, 2008. 283(17): p. 11493. M. K. Wild, M. C. Huang, U. Schulze-Horsel, P. A. van der Merwe, and D. Vestweber, Affinity, Kinetics, and Thermodynamics of E-Selectin Binding to ESelectin Ligand-1. J Biol Chem, 2001. 276(34): p. 31602. K. Noda, S. Nakao, S. Ishida, and T. Ishibashi, Leukocyte Adhesion Molecules in Diabetic Retinopathy. J Ophthalmol, 2012. 2012: p. 279037. G. A. Zimmerman, Two by Two: The Pairings of P-Selectin and P-Selectin Glycoprotein Ligand 1. Proc Natl Acad Sci U S A, 2001. 98(18): p. 10023. P. P. Wilkins, K. L. Moore, R. P. McEver, and R. D. Cummings, Tyrosine Sulfation of P-Selectin Glycoprotein Ligand-1 Is Required for High Affinity Binding to P-Selectin. J Biol Chem, 1995. 270(39): p. 22677. K. K. Sarangapani, T. Yago, A. G. Klopocki, M. B. Lawrence, C. B. Fieger, S. D. Rosen, R. P. McEver, and C. Zhu, Low Force DeceleratesL-Selectin Dissociation from P-Selectin Glycoprotein Ligand-1 and Endoglycan. J Biol Chem, 2004. 279(3): p. 2291. B. T. Marshall, M. Long, J. W. Piper, T. Yago, R. P. McEver, and C. Zhu, Direct Observation of Catch Bonds Involving Cell-Adhesion Molecules. Nature, 2003. 423(6936): p. 190. R. Alon, S. Chen, K. D. Puri, E. B. Finger, and T. A. Springer, The Kinetics ofLSelectin Tethers and the Mechanics of Selectin-Mediated Rolling. J Cell Biol, 1997. 138(5): p. 1169. M. Dembo, D. C. Torney, K. Saxman, and D. Hammer, The Reaction-Limited Kinetics of Membrane-to-Surface Adhesion and Detachment. Proc R Soc Lond B Biol Sci, 1988. 234(1274): p. 55. 34 [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] [57] [58] [59] [60] W. Thomas, Catch Bonds in Adhesion. Annu Rev Biomed Eng, 2008. 10: p. 39. R. Alon, S. Chen, R. Fuhlbrigge, K. D. Puri, and T. A. Springer, The Kinetics and Shear Threshold of Transient and Rolling Interactions of L-Selectin with Its Ligand on Leukocytes. Proc Natl Acad Sci U S A, 1998. 95(20): p. 11631. P. Sundd, M. K. Pospieszalska, L. S. Cheung, K. Konstantopoulos, and K. Ley, Biomechanics ofLeukocyte Rolling. Biorheology, 2011. 48(1): p. 1. P. Pawar, S. Jadhav, C. D. Eggleton, and K. Konstantopoulos, Roles of Cell and Microvillus Deformation and Receptor-Ligand Binding Kinetics in Cell Rolling. Am J Physiol Heart Circ Physiol, 2008. 295(4): p. H1439. E. Y. Park, M. J. Smith, E. S. Stropp, K. R. Snapp, J. A. DiVietro, W. F. Walker, D. W. Schmidtke, S. L. Diamond, and M. B. Lawrence, Comparison of Psgl-1 Microbead and Neutrophil Rolling: Microvillus Elongation Stabilizes P-Selectin Bond Clusters. Biophys J, 2002. 82(4): p. 1835. G. Schumacher, U. Bakowsky, C. Gege, R. R. Schmidt, U. Rothe, and G. Bendas, Lessons Learnedfrom Clustering of FluorinatedGlycolipids on Selectin Ligand Function in Cell Rolling. Biochemistry, 2006. 45(9): p. 2894. V. Ramachandran, M. Williams, T. Yago, D. W. Schmidtke, and R. P. McEver, Dynamic Alterations of Membrane Tethers Stabilize Leukocyte Rolling on PSelectin. Proc Natl Acad Sci U S A, 2004. 101(37): p. 13519. M. K. Pospieszalska and K. Ley, Dynamics of Microvillus Extension and Tether Formationin Rolling Leukocytes. Cell Mol Bioeng, 2009. 2(2): p. 207. J. Y. Shao, H. P. Ting-Beall, and R. M. Hochmuth, Static and Dynamic Lengths ofNeutrophil Microvilli. Proc Natl Acad Sci U S A, 1998. 95(12): p. 6797. C. Dong and X. X. Lei, Biomechanics of Cell Rolling: Shear Flow, Cell-Surface Adhesion, and Cell Deformability.Journal of Biomechanics, 2000. 33(1): p. 35. A. J. Goldman, R. G. Cox, and H. A. Brenner, Slow Viscous Motion of a Sphere Parallel to a Plane Wall. I. Motion through a Quiescent Fluid. Chemical Engineering Science, 1967. 22: p. 637. A. J. Glodman, R. G. Cox, and H. A. Brenner, Slow Viscous Motion of a Sphere Parallelto a Plane Wall. Ii. Couette Flow. Chemical Engineering Science, 1967. 22: p. 653. A. W. Greenberg, D. K. Brunk, and D. A. Hammer, Cell-Free Rolling Mediated by L-Selectin and Sialyl Lewis(X) Reveals the Shear Threshold Effect. Biophys J, 2000. 79(5): p. 2391. A. 0. Eniola, P. J. Willcox, and D. A. Hammer, Interplay between Rolling and Firm Adhesion Elucidatedwith a Cell-Free System Engineered with Two Distinct Receptor-LigandPairs.Biophys J, 2003. 85(4): p. 2720. D. K. Brunk and D. A. Hammer, Quantifying Rolling Adhesion with a Cell-Free Assay: E-Selectin and Its CarbohydrateLigands. Biophys J, 1997. 72(6): p. 2820. X. Lei, M. B. Lawrence, and C. Dong, Influence of Cell Deformation on Leukocyte Rolling Adhesion in Shear Flow. J Biomech Eng, 1999. 121(6): p. 636. K. L. Moore, K. D. Patel, R. E. Bruehl, F. Li, D. A. Johnson, H. S. Lichenstein, R. D. Cummings, D. F. Bainton, and R. P. McEver, P-Selectin Glycoprotein Ligand1 Mediates Rolling of Human Neutrophils on P-Selectin. J Cell Biol, 1995. 128(4): p. 661. 35 [61] [62] [63] [64] [65] [66] [67] [68] [69] [70] [71] [72] [73] [74] D. F. Bainton, Distinct Granule Populations in Human Neutrophils and Lysosomal Organelles Identified by Immuno-Electron Microscopy. J Immunol Methods, 1999. 232(1-2): p. 153. U. H. von Andrian, S. R. Hasslen, R. D. Nelson, S. L. Erlandsen, and E. C. Butcher, A Central Role for Microvillous Receptor Presentation in Leukocyte Adhesion under Flow. Cell, 1995. 82(6): p. 989. T. Yago, A. Leppanen, H. Y. Qiu, W. D. Marcus, M. U. Nollert, C. Zhu, R. D. Cummings, and R. P. McEver, Distinct Molecular and Cellular Contributions to Stabilizing Selectin-MediatedRolling under Flow. Journal of Cell Biology, 2002. 158(4): p. 787. W. C. Chang, L. P. Lee, and D. Liepmann, Biomimetic Technique for AdhesionBased Collection and Separation of Cells in a Microfluidic Channel. Lab on a Chip, 2005. 5(1): p. 64. A. W. Greenberg and D. A. Hammer, Cell Separation Mediated by Differential Rolling Adhesion. Biotechnology and Bioengineering, 2001. 73(2): p. 111. D. D. Nalayanda, M. Kalukanimuttam, and D. W. Schmidtke, Micropatterned Surfaces for Controlling Cell Adhesion and Rolling under Flow. Biomed Microdevices, 2007. 9(2): p. 207. C. Edington, H. Murata, R. Koepsel, J. Andersen, S. Eom, T. Kanade, A. C. Balazs, G. Kolmakov, C. Kline, D. McKeel, Z. Liron, and A. J. Russell, Tailoring the Trajectory of Cell Rolling with Cytotactic Surfaces. Langmuir, 2011. 27(24): p. 15345. S. Choi, J. M. Karp, and R. Karnik, Cell Sorting by Deterministic Cell Rolling. Lab Chip, 2012. 12(8): p. 1427. C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Studying Cell Rolling Trajectorieson Asymmetric Receptor Patterns.J Vis Exp, 2011(48). C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Examining the Lateral Displacement of H160 Cells Rolling on Asymmetric P-Selectin Patterns. Langmuir, 2011. 27(1): p. 240. X. H. Cheng, D. Irimia, M. Dixon, K. Sekine, U. Demirci, L. Zamir, R. G. Tompkins, W. Rodriguez, and M. Toner, A Microfluidic Device for Practical Label-Free Cd4+TCell Counting of Hiv-Infected Subjects. Lab on a Chip, 2007. 7(2): p. 170. S. Q. Chen, R. Alon, R. C. Fuhlbrigge, and T. A. Springer, Rolling and Transient Tethering of Leukocytes on Antibodies Reveal Specializations of Selectins. Proceedings of the National Academy of Sciences of the United States of America, 1997. 94(7): p. 3172. N. Charles, J. L. Liesveld, and M. R. King, Investigating the Feasibility of Stem Cell Enrichment Mediated by Immobilized Selectins. Biotechnology Progress, 2007. 23(6): p. 1463. B. K. Kay, J. Kasanov, and M. Yamabhai, Screening Phage-Displayed CombinatorialPeptide Libraries.Methods, 2001. 24(3): p. 240. 36 2 Free-Flow Travel and Capture of HL60 Cells inside Microfluidic Channels 2.1 Introduction Chapter Two aims to study how cells travel in microfluidic channels before they are drawn out of the flowing stream by affinity-mediated cell capture. How the cells are captured on the substrate is important because these parameters influence the design and performance of microfluidic devices for cell separation. Adhesive receptor molecules exhibiting weak interaction with cells can recruit cells out from a continuously flowing stream resulting in cell rolling and tracking along the patterns [1-11]. Continuous label-free devices for the separation or analysis of cells based on transient interactions with low affinity receptor-coated bottom surfaces within the device have attracted a lot of attention recently [9-14]. When a cell enters the microfluidic channel where the flow is in the low-Reynold-number regime (the inertial effects are negligible), it travels in both x and y directions with the flow: (a) in y (vertical) direction, the cell settles to the bottom of the channel because of the gravity while experiencing frictional force and buoyant force in the opposition direction; (b) in x (horizontal) direction, the cell travels with the flowing stream by the convection force before it is captured by the adhesive receptors coated on the bottom of the device [15, 16]. The design of the length of the channel should be long enough to ensure the majority of the cells can settle under the influence of gravity while it travels by convection until it starts to interact with the receptor molecules at the bottom surface of the channel. Accordingly, it is important to know how far the cell travels before it reaches the bottom surface. This distance is defined as the settling distance, xs, as shown in Figure 2-1(a). Rolling may be initiated after the cell is captured onto the substrate. Cell capture in bio-functionalized microchannels under shear flow has been studied [1520]. For example, Tempelman et al have studied the effect of shear rate on the adhesion of anti-dinitrophenol (DNP)- IgE sensitized cells on to the DNP-coated area in the flow 37 chamber: the cells were allowed to settle before they entered the DNP-coated area and it was observed that the density of adherent cells decreases with distance down the chamber; however, only the percentage of adhesion of the cells within the field of view was qualitatively analyzed [18]. On the other hand, Cheung et al have analyzed the spatial distributions of captured circulating tumor cells (CTCs) along the anti-EpCAMfunctionalized microchannels and results show that at the shear stress of 0.207 dyn/cm 2, the most probable location for the cells to be captured is about x/L=0.28, where x is the locations from the inlet and L is the total length of the channel (=32 cm). However, in their system, the whole microchannel was coated with antibodies and the cells were not settled at x/L =0 [17]. In Chapter One, a separation device with multiple inclined receptor bands has been proposed (Figure 1-6). The interacting cell can be captured onto the patterns and roll on them. The asymmetry of the patterns with respect to the flow alters the trajectory of the interacting cells. The interacting cells can track along the edge of the pattern and get displaced into the buffer stream and are thus separated from the non-interacting cells. Since multiple patterns are designed on the substrate, it is important to understand the capture process on each inclined pattern from the free flowing stream. For example, what is the attachment distance, /a, which is defined as how far a settled cell will travel from the beginning of a receptor-coated area until it is captured and starts to roll on the substrate (see Figure 2-1(b)). This parameter relates to the design of the width of pattern and further plays an important role in the performance of the device. On the other hand, the distribution of the attachment distance is unknown and it is not fully understood how the attachment distance changes with shear stress magnitudes. Therefore, systematic study of free-flow travel and the recruitment process of the cells from the flow onto the substrate inside the microfluidic channels is prerequisite for the design and the development of microfluidic devices involving transient interactions between the cells and the receptor-coated substrate for analysis or separation of the cells. The problem is also of interest in cell biology research, for example, seeding of cells in microchannels for tissue culture [21]. 38 (a) inlet, (b) x,: settling distance Se ed cell [R] Receptor 5 cm Q tAttachment Di ance, 1. * Attached cells vomw Flowing cells Figure 2-1 Schematic diagrams of cells travelling in a microfluidic channel. (a) The illustration of a cell entering the microfluidic channel and travelling in both x and y directions. The settling distance, x,, is defined as how far a cell will travel before it settles close enough to the bottom surface. (b) Illustration of cells passing the Pselectin-coated area. A cell may be captured from the flowing stream at an attachment distance, I., after it enters the P-selectin-coated area. In Chapter Two, the role of fluid flow on cell adhesion will be studied using simple shear flow in a parallel-plate microchannel. A long microfluidic channel envisioned in the top of Figure 2-1(b) was designed for the study of how the HL60 cells travel with the free flowing stream and be captured onto the P-selectin-coated substrate. HL60 cells and Pselectin molecules have been widely used as a model system for leukocyte rolling study 39 [22-26]. The surfaces of HL60 cells express P-selectin glycoprotein ligand-1 (PSGL-1), which binds reversibly to the receptor P-selectin to enable rolling in vivo and in vitro [22]. The long microlfuidic channel was first used to study the settling distance (x,) under different shear stresses. The changes of the convection velocity of the cells while they travel along the channel were analyzed to quantify how far the cells travel before settling under the gravity. In the second part, the P-selectin molecules were coated at the end region of the channel (see Figure 2-1(b)) to ensure the cells have enough time to settle before they start to interact with the receptor molecules at the bottom surface. The attachment distances (Ia) of the cells under different shear stresses were quantified and an empirical relation between the characteristic attachment distance and the shear stress was established based on the experimental results. 2.2 Part A. Cell Settling inside Microfluidic Channels Part A aims to quantitatively analyze free-flow travel of the cells inside microfluidic devices and identify the settling distance (x,) under different shear stresses. A long microchannel (- 15 cm) was designed for this study. The convection velocities of the cells at different location along the channel were analyzed. The convection velocity was expected to decrease as the cells settle to the surface under the gravity and x, is identified as how far the cells travel before settling. 2.2.1 2.2.1.1 Fabrication of Substrate and Microfluidic Channel Materials Petri dishes were purchased from BD FalconTM. Polydimethylsiloxane (PDMS; Sylgard@184) was purchased from Dow Coming. Bovine serum albumin (BSA, Rockland Immunochemicals, Inc.) was diluted in 150 mM NaCl Dulbecco's phosphate buffered saline (DPBS, Mediatech Inc.). All materials employed in this study were used without further purification unless specified otherwise. 40 2.2.1.2 Fabrication of Substrate Incorporated PDMS channel The Petri dish was incubated with 3-wt% BSA in DPBS for 3 h at room temperature. The Petri dish was then rinsed with DPBS and kept wet to serve as the substrate. A silicon master mold that defines the microfluidic channel used in this study was a gift from Chong Shen in Prof. Kamik's group at MIT. The microfluidic channel was cast in PDMS from the silicon master mold. The height of this 15 cm-long channel is 95 pim and the width is 2 mm. The PDMS channel was then attached to the BSA-coated substrate via a vacuum manifold to assemble the device. 2.2.2 2.2.2.1 Convection Velocity Experiment Cell Culture HL60 myeloid cells (ATCC) were maintained in Iscove's modified Dulbecco's medium supplemented with 20% fetal bovine serum (FBS) according to the ATCC recommendation. Cells were cultured in 75 cm 2 polystyrene tissue culture flasks (BD Falcon) with the cell density maintained between 105 - 106 cells/mL. (passage number 39, 40 and 41 were used in this study). 2.2.2.2 Experiment Setup A suspension of the HL60 cells (~ 105 cells/mL) was allowed to flow into the assembled microfluidic channel as described above at room temperature of 24.5 C. A syringe pump (World Precision Instruments (WPI), SP230IW) was used to generate different flow rates of between 5 and 20 pL/min, with corresponding shear stresses of 0.25-1.0 dyn/cm 2 (-0.025-0.1 Pa). The flow was laminar (Reynold's number Re ~ 0.1-0.4), and the shear stress r was calculated using the plane Poiseuille flow equation r=6pQ/wh 2 , where p is the viscosity, Q is volumetric flow rate, w is width of the flow chamber, and h is height of the channel. An inverted microscope (Nikon TE2000-U) with a mounted camera (Andor iXon 885) was used to record the flowing HL60 cells using a 4x objective, typically at a rate of 1 frame/s for durations of 60-180 s with exposure time of 0.05-1.0 s. 41 The videos recording the cells traveling in the free flowing stream were taken at different positions away from the inlet (2, 4, 6, 8, 10, 12, and 13 cm) under different shear stresses. 2.2.2.3 Results and Data Analysis For each experiment of specific position and shear stress, 20 cells were selected randomly from the video. The position in x-coordinate (xi) of each selected cell in the free stream at time t4 was identified manually by ImageJ. The convection velocity (u,,) of a cell is defined as u = X'"(t)m X,(tn) ;m>n (Eq. 2-1) tm -tn The convection velocity of the cell in the free flowing stream is assumed to be the same as the velocity profile of steady state laminar flow for Newtonian fluid, which varies quadratically with the height from the bottom surface [27]: QY-j hW h where 4Uy h I Yiy h h (Eq. 2-2) Q: volumetric flow rate w: width of the channel h: height of channel y :height of the center of the cell from the bottom surface of the channel of the flow rate shear y7: Umax: maximum convection velocity of flow However, when the cell (radius, r) is settled to the height less than 4r (y < 4r), the convection velocity is reduced by the hydrodynamic wall effect as described by the Goldman equation [15, 16, 28]: U=~( h W 1-- 5 r 16 y 42 YY (r) 16 y (Eq. 2-3) Figure 2-2(a) summaries the convection velocities of the cells in the free flowing stream (y-axis) at different horizontal positions away from the inlet (x-axis). Wall shear stress varies from 0.25 to 1.0 dyn/cm 2 . Data are presented as the mean velocity and the standard deviation of 20 cells for each experiment condition. This convection velocity can be used to indicate whether a cell is close to the surface or not: the cell travels at maximum convection velocity near the half height of the channel; as the cell keeps settling down, the convection velocity decreases until u, reaches a minimum value. As the cell reaches the bottom surface (y ~ r), the Goldman equation can be simplified as: U,-- 11r 11 ry--T 16 16 U C (1 XT or or (Eq. 2-4) \ 16)(t Eq. 2-4 suggests that as y ~ r, the convection velocity is roughly proportional to the shear rate, y) (or shear stress r; for Newtonian fluid with viscosity of p, shear stress is defined as r = p Y). Figure 2-2(b) summarizes the values of u / II)(T)] of cells at different horizontal positions The results show that for all the different shear stresses, the values of u / I1)(-r) decreases with the travelling distance and reaches a minimum around the value of 5 [pm], which is roughly consistent with the Goldman equation (as y ~ r ~ 5p m, u / ~)] 4.95 [pm]). This suggests that the average convection velocity of the cells after settling increases linearly with the increasing shear stress within the tested range. The settling distance, xs, can be identified from Figure 2-2(a) or Figure 2-2(b). For the shear stresses of 0.25, 0.5 and 0.75 dyn/cm 2 , x, is between 4 and 6 cm. For the shear stress of 1.0 dyn/cm 2 , x, is around 10 - 12 cm. For the design of the continuous microfluidic cells separation device based on the interaction between the cells and the receptor-coated bottom surfaces, x, is be a key parameter to ensure most of the cells are able to interact with the substrate. Moreover, the average of the convection velocity of the 43 settled cells increases linearly with increasing shear stress (see Figure 2-2 (c)). The average convection velocity scales with the shear stress [28], which indicates shear stress is a controlling parameter for convective transport of the cells inside the microfluidic device. Yago et al observed similar linear relationship when the microspheres flowed inside the flow chamber [20]. 44 2000 (a) 2 T (dyn/cM 1500 ) -0-O.25 E -0-.75 S1000 500 0 12 10 8 6 4 2 Horizontal Position from the inlet, x (pm) 3 (b) 14 25 . (dyn/cM2 ) 20 +0.25 +0.5 =1.11~ 15 +0.75 10 5 0 0 (c) 12 10 8 6 4 2 Horizontal Position from the inlet, x (pm) 500 (dyn/cm 2) Uc, (pm/s) (Avg ± SD) 0.25 0.5 0.75 1.0 104± 5 205± 5 292 ± 5 391 ± 5 T 400 300 200 1 0 - .P 394r R= 0.99 100 0 0 0.25 0.75 0.5 T (dyn/cm2) 45 1 14 Figure 2-2(a) Convection velocity (u,) of the cells at the different horizontal position. Convection velocity divided by shear stress decreases and reaches a minimum value as the cells travel along the channel. Data are presented as the mean and the standard deviation of the convection velocity obtained from 20 cells under each experiment condition. Different markers and colors represent different shear stresses. (b) [u, ] /[(I I/ 16)(T /i)] of the cells at different horizontal positions (c) Variation of the convection velocity of the settled cells with the shear stress. Inset summaries the mean and the standard deviation of the convection velocity of the cells at position x = 13 cm. The relationship between uc, and T is well described by a linear fit (dashed line). 2.3 Part B. Attachment Distance of the Cells in the Microfluidic Channel Part B aims to quantitatively analyze capture process of cells onto the receptor-coated microfluidic devices under different shear stresses. HL60 cells were flowed into the 15cm long microchannel and the P-selectin molecules were coated only at the end region (13-14cm) of the bottom of the microchannel to ensure the cells have enough time to settle before starting to interact with the receptor molecules on the bottom surface. Capture process can be studied by analyzing the distribution of the attachment distance ('a), the distance between the starting point of the P-selectin area and the position where the cells were captured. 2.3.1 2.3.1.1 Fabrication of Substrate and Microfluidic Channel Materials The Petri dish was purchased from BD FalconTM. Polydimethylsiloxane (PDMS; Sylgard@184) was purchased from Dow Coming. Recombinant human P-selectin (R&D Systems Inc.) and fluorescein isothiocyanate (FITC)-conjugated BSA (Sigma-Aldrich) were diluted in 150 mM NaCl Dulbecco's phosphate buffered saline (DPBS, Mediatech Inc.). Perfusion chambers were purchased from Electron Microscopy Sciences. All materials employed in this study were used without further purification unless specified otherwise. 46 2.3.1.2 Fabrication of Substrate Incorporated PDMS channel The Petri dish was incubated with P-selectin at 5 pg/mL in DPBS using a perfusion chamber for 3 h at room temperature. The surface was then incubated with FITCconjugated BSA (1 mg/mL in DPBS) at room temperature for 15 min to identify the edge of P-selectin-coated area. The surface was then backfilled with BSA (3wt% in DPBS) at room temperature for 1 h to block nonspecific interactions. The Petri dish was then rinsed with DPBS and kept wet to serve as the substrate. The PDMS channel (the same design as being used in Part A) was then assembled with the substrate via a vacuum manifold. The P-selectin-coated area was aligned near the end of the microfluidic channel as illustrated in Figure 2-1(b). The P-selectin-coated area started from the position around 13-14 cm away from the inlet (which is larger than the settling distance (x,)) to ensure the cells had enough time to settle down. 2.3.1.3 Substrate Characterization An inverted fluorescent microscope (Nikon TE2000-U) was used to characterize the substrate. Well-defined edge of P-selectin-coated region can be identified as shown in Figure 2-3. The bright area corresponds to FITC-BSA-coated area. inlet outlet tin Figure 2-3 Fluorescent images of the substrate. The bright area corresponds to FITCBSA-coated area. 47 2.3.2 2.3.2.1 Cell Capture Experiment Cell Culture and Experiment Setup The setup of the cell capture experiment was similar to what has been described in section 2.2.2 (the convection velocity experiment). The videos were taken near the area where the P-selectin-coated area started. The HL60 cells may be captured onto the Pselectin-coated area to initiate rolling after they enter this region. The position where the cells captured from the free flowing stream onto the surface was defined as the attachment distance, /a (see Figure 2-1(b)). The wall shear stresses were varied from 0.25 to 1.0 dyn/cm 2. For each shear stress magnitude, three independent experiments were performed. 2.3.2.2 Data Analysis The image sequences were analyzed using a customized Matlab (Mathworks, Inc.) program that utilized particle-tracking freeware to detect the captured cells [29]. The total number of the cells entering the P-selectin-coated area during the certain time period within the field of view was counted manually. Figure 2-4 is an example showing one frame extracted from a video taken under shear stress of 0.25 dyn/cm 2 . After the cells entered the receptor-coated area, they can be drawn out from the free stream and be captured onto the surface. It is clearly shown that some of the cells flowing into the field of the view flowed out of the field of view. The distance where a cell was captured onto the surface from the start of the receptor coated area is defined as the attachment distance (la). The average attachment distance is useful for quantitatively understanding the capture process. Figure 2-5 shows an example of the distribution of the attachment distances of HL60 cells at shear stress of 0.25 dyn/cm 2. The cells flowed into the field of view at different time. This figure was obtained by analyzing 65 images acquired at 1 fps using a customized Matlab code. It was observed that the attachment distance has a distribution, which is consistent with previous observations [18, 48 19]. It is more insightful to understand the distribution of the attachment distance, since it could be used for studying the spatial distribution of the cells inside a microfluidic channel. ng out of of view n P+ P-sekwctn coated as x-O Figure 2-4 HL60 cells being captured onto the P-selectin-coated area at the shear stress of 0.25 dyn/cm 2 . Some cells were observed to flow out of the field of view. 80 6 .0 Poe0 a' E 4 % 0,%0* 0 *1= * 0 0 Go 2 0 500 1000 1500 2000 Attachment Distance (pm) Figure 2-5 The attachment distances of HL60 cells at the shear stress of 0.25 dyn/cm 2. P-selectin edge is located at 0 lim. The data obtained by analyzing 65 images acquired at 1 fps using a customized Matlab code. Figure 2-6(a) shows the frequency of the attachment distance at different shear stresses 2 (0.25-1.0 dyn/cm 2). For shear stresses of 0.25 and 0.5 dyn/cm , the distributions exhibit a decaying characteristic. Cumulative probability (CP) was calculated form the data and fit it with an empirical equation: 49 CP(x) = A l - exp 7 (Eq. 2-5) Here, </4> is the characteristic attachment distance and in the ideal case, parameter A will approach the value of one. A and </a> obtained by fitting the cumulative probabilities obtained from experimental data to Eq. 2-5 for different shear stresses are summarized in Table 2-1. Three independent experiments were performed for each shear stress and data are presented as mean and one standard deviation. Eq. 2-5 fits well for T = 0.25 and 0.5 dyn/cm 2 . Interestingly, the average values of A for T = 0.25 and 0.5 dyn/cm 2 have close results (0.5±0.1 and 0.5±0.1, respectively). However, for T = 0.75 and 1.0 dyn/cm 2 , the plateaus in Figure 2-6(b) were not observed due to insufficient data so it is difficult to do curve fitting using Eq. 2-5. To further investigate the empirical relation between <4,> and T, we assumed that the value of A fort = 0.75 and 1.0 dyn/cm 2 is the same as that for -= 0.25 and 0.5 dyn/cm 2 : HL60 cells may exhibit cellular heterogeneity. HL60 cells with higher PSGL-1 expression level may have small <Ia> and other cells with lower PSGL- 1 expression level may have large </a>. The fraction of the cells with small </a> is "A" and the fraction of the cells with large <I,> is "1-A." Limited by the field of view of the camera, only the cells with small <la> can be analyzed under the shear stress we are interested (0.25 and 1.0 dyn/cm 2). The empirical equation was then simplified as: CP(x) =0.5 1- exp - (Eq. 2-6) Here, 0.5 was determined by averaging the mean values of A from -u = 0.25 and 0.5 dyn/cm 2 . <4"> obtained by fitting the cumulative probabilities to Eq. 2-6 for different shear stresses are summarized in Table 2-1. The values of <a> obtained from Eq. 2-5 and 50 Eq. 2-6 for T = 0.25 and 0.5 dyn/cm 2 were compared and the maximum difference was found to be within 4% (data are not shown in the Table 2-1). (a) I 2 0.25 dyn/cm U 0 GJ 2000 1000 o 1000 2000 0* a) 15 U- 8 0.75 dyn/cm 2 1.0 dyn/cm 10 00 1000 1 2000 00 500 1000 1500 2000 Attachment Distance (pm) (b) 0.5 0.25 dyn/cm2m 0.4 .5 dyn/cM2 - 'u 0.3 0 0.2 LI 0.75 dyn/cM2 1.0 dyn/cM2 0.1 0 0 500 1000 1500 2000 Attachment Distance (pm) Figure 2-6 Attachment distance of HL60 cells at different shear stresses of 0.25-1.0 dyn/cm2. Representative results are shown for only one experiment for each shear stress. (a) Distribution of attachment distance at different shear stresses (0.25-1.0 dyn/cm 2). (b) Cumulative probability (CP) of attachment distance at different shear stresses (0.25-1.0 dyn/cm2). Table 2-1 Comparison of the average values and standard deviation of A and <J.> obtained from curve fitting at different shear stresses of 0.25-1.0 dyn/cm 2. n = 3 51 replicate experiments. Data were obtained by fitting to two empirical equations with experimentally measured attachment distance. P umba i ve CP(x)- A 1- exp )) Probability .1,.) T(dyn/cm 2) 0.25 0.5 0.75 1 0.25 0.5 0.75 1- Avg±SD 0.5±0.1 0.5±0.1 A <I? pm Probability CP(x) -0.5 T (dyn/cm 2) 0.25 0.5 0.75 1 620±100 1500±100 - 1- exp - Avg±SD <a> pm 650±80 1480±490 4840±930 15580±9000 Interestingly, the average value of </,> obtained by fitting the cumulative probability to Eq. 2-6 showed an exponential increase with increasing shear stress (Figure 2-7). This observation provides an empirical relation between the shear stress and the average characteristic attachment distance under the shear stress of 0.25-1.0 dyn/cm 2 . As a cell settles to contact the substrate, new cell-substrate adhesion may form at the contact area between the cell and receptor-coated surface. Higher flow rate results in higher shear stress and faster convection velocity. Higher convection velocity near the bottom of the substrate produces more frequent collisions between the cells and the substrate but short the contact time before the cell moves forward [8, 30]. There is no justification why an exponential relation was observed. However, the experimental results imply that the effect of shear stress on the characteristic attachment distance becomes more significant at higher shear stress, and likely originates from the nature of the molecular interactions rather than hydrodynamic effects since the velocity of settled cells remains linear with the shear stress (Figure 2-2(c)). 52 10000 - A 1000 og<Ia>= 1.9T+2.3 V R2 = 0.99 100 0 0.25 0.5 0.75 1 Shear Stress,T (dyn/cm 2) Figure 1- 1 Variation of the average value of </4> with shear stress. The average values of <1.> were obtained from the empirical fit (Eq. 2-6). The relationship between <la> and T is well described by a linear fit on a semi-log plot (dashed line). Error bars represent one standard deviation. The probability of a cell being captured by a pattern from the free flow can be obtained from the empirical equation as described above. Figure 2-8 shows the schematic diagram of a cell travelling over a single inclined pattern of width of w, with respect to the flow of angle of a. After the cell enter the pattern area, it can be captured within the area of width of x = wisina. Eq. 2-5 can be modified as: CP(x) = A I -exp - . (Eq. 2-7) For a cell travelling under a shear stress of 0.5 dyn/cm 2 over an inclined pattern of width of 15 pim, at the inclination angle of a = 150 with respect to the flow, the probability of the cells being captured by this pattern from the free flowing stream is ~ 0.02 (A = 0.5 and </a> = 1480). This value is consistent with Bose et al's study which reported the experimental probability of 0.015 - 0.038 at this specific pattern geometry and shear stress [14]. 53 Starting point l Cell% Fw Flow Figure 1- 2 Schematic diagram of a cell travelling over a single inclined pattern. A cell travels over the pattern of width w and an inclination angle a with respect to the flow. After the cell enters the pattern area, it can be captured within a distance of x = wsina. The probability of the cell being captured by this pattern from the free flowing stream is CP(x'). 2.3.3 Flow Cytometry Analysis The work of flow cytometry analysis was done in collaboration with Suman Bose (MIT). 10 % fetal bovine serum (FBS, ATCC) and 0.1% sodium azide (Sigma-Aldrich) in phosphate buffered saline (PBS, Mediatech Inc) were prepared to serve as FACS buffer. PE (phycoerythrin)- conjugated anti-PSGL-1 antibody (BD Pharmingen) was used to stain PSGL-l surface expression from HL-60 cells. Mouse IgG1, C isotype (BD Pharmingen) was used as the control. Two antibodies were diluted separately in FACS Buffer to the final concentration of 1 pg/mL. HL60 cells were transferred from the flask into two 15 mL conical tubes and were spun at 1700 rpm for 5 min. The supernatant was discarded and the pellets were suspended in diluted anti-PSGL 1-PE and isotype antibody solutions separately to a final concentration of 106 cell/mL. The suspended cells were then incubated inside the ice bath for 30 minutes. The cells were then spun down at 1700 rpm for 5 min and the pellets were then suspended in FACS Buffer. The samples were then analyzed by flow cytometry machine (LSRFortessa). 54 Heterogeneity in the cell population has been proposed to have an impact on the cell adhesion under hydrodynamic flow [18, 19]. The flow cytometry results (see Figure 2-9) suggest that the expression of PSGL-1 on HL60 cells has broad distribution, which is consistent with Gee et al's observation [31]. The broad distribution of PSGL-1 expression indicates that there is some degree of cell-to-cell difference. Such cell population heterogeneity may result in the distribution of the attachment distance and the cells with higher expression level of PSGL-1 may have shorter attachment distance and vice versa. If HL60-PSGL1-PE HL60-iso 0 103 104 10s Intensity Figure 1- 3 Flow cytometry analysis. PE conjugated anti-PSGL- 1 antibody was used to stain PSGL-1 surface expression from HL-60 cells. Mouse IgGi, K isotype antibody was used as the control. (Adapted from Suman Bose). 2.4 Conclusion In summary, the cells settle down under the influence of gravity after they enter the microfluidic channel and then travel along the channel with the free stream. The experimental results show that the average convection velocity of the cells with the flow decreases until it reaches a minimum value when the cells are close to the bottom, which is consistent with the Goldman equation. The minimum convection velocity is an indicator when the cell are settled and the position where the convection velocity reaches a minimum value or the cells settle to the bottom of the substrate is defined as the settling distance (xs). Quantification of the settling distance is useful for the design of the continuous microfluidic cell separation device based on the interaction with receptorcoated bottom surfaces. Moreover, and the shear stress has been identified as the controlling parameter of convective transport. In addition, the nature of cell recruitment 55 from the flow onto the receptor-coated area has been studied by analyzing the attachment distance (l), which is defined as the distance between the position where the cell is captured and the position where the pattern starts. Although the study was limited by the size of the field of view, the experimental results suggest that high shear stress results in long attachment distance and the effect of shear stress on the characteristic attachment distance become more significant at higher shear stress since the characteristic attachment distance increase exponentially with increasing shear stress. The empirical relationship between cumulative probability of the cells being captured and the position of the channel was established. The empirical relationship was then applied to study the probability of a cell being captured on a single inclined pattern from the free flow and the result is consistent with experimental observation. 2.5 Chapter Reference [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] G. I. Bell, Models for Specific Adhesion of Cells to Cells. Science, 1978. 200(4342): p. 618. R. Alon, D. A. Hammer, and T. A. Springer, Lifetime of the P-SelectinCarbohydrate Bond and Its Response to Tensile Force in Hydrodynamic Flow. Nature, 1995. 374(6522): p. 539. D. N. Granger and P. Kubes, The Microcirculation and Inflammation Modulation of Leukocyte-Endothelial Cell-Adhesion. Journal of Leukocyte Biology, 1994. 55(5): p. 662. L. A. Lasky, Selectin-Carbohydrate Interactions and the Initiation of the Inflammatory Response. Annual Review of Biochemistry, 1995. 64: p. 113. B. T. Marshall, M. Long, J. W. Piper, T. Yago, R. P. McEver, and C. Zhu, Direct Observation of Catch Bonds Involving Cell-Adhesion Molecules. Nature, 2003. 423(6936): p. 190. T. F. Tedder, D. A. Steeber, A. Chen, and P. Engel, The Selectins - Vascular Adhesion Molecules. Faseb Journal, 1995. 9(10): p. 866. G. A. Zimmerman, Two by Two: The Pairings of P-Selectin and P-Selectin Glycoprotein Ligand 1. Proc Natl Acad Sci U S A, 2001. 98(18): p. 10023. R. P. McEver and C. Zhu, Rolling Cell Adhesion. Annu Rev Cell Dev Biol, 2010. 26: p. 363. C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Studying Cell Rolling Trajectorieson Asymmetric Receptor Patterns.J Vis Exp, 2011(48). C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Examining the Lateral Displacement of H160 Cells Rolling on Asymmetric P-Selectin Patterns. Langmuir, 2011. 27(1): p. 240. 56 [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] R. Karnik, S. Hong, H. Zhang, Y. Mei, D. G. Anderson, J. M. Karp, and R. Langer, Nanomechanical Control of Cell Rolling in Two Dimensions through Surface PatterningofReceptors. Nano Lett, 2008. 8(4): p. 1153. S. Choi, J. M. Karp, and R. Karnik, Cell Sorting by Deterministic Cell Rolling. Lab Chip, 2012. 12(8): p. 1427. C. Edington, H. Murata, R. Koepsel, J. Andersen, S. Eom, T. Kanade, A. C. Balazs, G. Kolmakov, C. Kline, D. McKeel, Z. Liron, and A. J. Russell, Tailoring the Trajectory of Cell Rolling with Cytotactic Surfaces. Langmuir, 2011. 27(24): p. 15345. S. Bose, R. Singh, C. Shen, M. Hanewich-Hollatz, C. H. Lee, D. M. Dorfman, J. M. Karp, and R. Karnik, Affinity Flow Fractionation of Cells Via Transient Interactionswith Asymmetric Molecular Patterns. Scientific Reports, 2013(3): p. 2329. Y. Zhang and S. Neelamegham, Estimating the Efficiency of Cell Capture and Arrest in Flow Chambers: Study of Neutrophil Binding Via E-Selectin and Icam1. Biophys J, 2002. 83(4): p. 1934. Y. Zhang and S. Neelamegham, An Analysis Tool to Quantify the Efficiency of Cell Tethering and Firm-Adhesion in the Parallel-Plate Flow Chamber. J Immunol Methods, 2003. 278(1-2): p. 305. L. S. Cheung, X. Zheng, L. Wang, R. Guzman, J. A. Schroeder, R. L. Heimark, J. C. Baygents, and Y. Zohar, Kinematics of Specifically Captured Circulating Tumor Cells in Bio-Functionalized Microchannels. Microelectromechanical System 2010. 19(4): p. 752. L. A. Tempelman and D. A. Hammer, Receptor-Mediated Binding of IgeSensitized Rat Basophilic Leukemia Cells to Antigen-Coated Substrates under Hydrodynamic Flow. Biophys J, 1994. 66(4): p. 1231. D. G. Swift, R. G. Posner, and D. A. Hammer, Kinetics of Adhesion of IgeSensitized Rat Basophilic Leukemia Cells to Surface-Immobilized Antigen in Couette Flow. Biophys J, 1998. 75(5): p. 2597. T. Yago, V. I. Zarnitsyna, A. G. Klopocki, R. P. McEver, and C. Zhu, Transport Governs Flow-Enhanced Cell Tethering through L-Selectin at Threshold Shear. Biophys J, 2007. 92(1): p. 330. E. W. K. Young and D. J. Beebe, Fundamentals of Microfluidic Cell Culture in ControlledMicroenvironments. Chemical Society Reviews, 2010. 39(3): p. 1036. K. E. Norman, K. L. Moore, R. P. Mcever, and K. Ley, Leukocyte Rolling in- Vivo Is Mediated by P-Selectin Glycoprotein Ligand-1. Blood, 1995. 86(12): p. 4417. M. B. Lawrence, G. S. Kansas, E. J. Kunkel, and K. Ley, Threshold Levels of Fluid Shear Promote Leukocyte Adhesion through Selectins (Cd62l,PE). Journal of Cell Biology, 1997. 136(3): p. 717. C. Dong and X. X. Lei, Biomechanics of Cell Rolling: Shear Flow, Cell-Surface Adhesion, and Cell Deformability.Journal of Biomechanics, 2000. 33(1): p. 35. L. Wu, B. T. Xiao, X. L. Jia, Y. Zhang, S. Q. Lu, J. Chen, and M. Long, Impact of CarrierStiffness and Microtopology on Two-Dimensional Kinetics of P-Selectin and P-Selectin Glycoprotein Ligand-1 (Psgl-1) Interactions. Journal of Biological Chemistry, 2007. 282(13): p. 9846. 57 [26] [27] [28] [29] [30] [31] W. C. Chang, L. P. Lee, and D. Liepmann, Biomimetic Techniquefor AdhesionBased Collection and Separation of Cells in a Microfluidic Channel. Lab on a Chip, 2005. 5(1): p. 64. R. B. Bird, W. E. Stewart, and E. N. Lightfoot, TransportPhenomena, 1960, New York: Wiley. p. 45. A. J. Glodman, R. G. Cox, and H. A. Brenner, Slow Viscous Motion of a Sphere Parallelto a Plane Wall. Ii. Couette Flow. Chemical Engineering Science, 1967. 22: p. 653. http://physics.georgetown.edu/matlab/. K. C. Chang and D. A. Hammer, The Forward Rate of Binding of SurfaceTethered Reactants: Effect of Relative Motion between Two Surfaces. Biophys J, 1999. 76(3): p. 1280. D. J. Gee, L. K. Wright, J. Zimmermann, K. Cole, K. Soule, and M. Ubowski, Dimethylsulfoxide Exposure Modulates Hl-60 Cell Rolling Interactions. Biosci Rep, 2012. 32(4): p. 375. 58 3 Lateral Displacement of HL60 Cells Rolling on Inclined PSelectin Adhesive Bands' 3.1 Introduction Understanding the cell rolling behaviors on the receptor bands is important to develop the separation devices based on transient cell-receptor interaction. Chapter Three will discuss the development of the substrates with the receptor bands and the studies of the cells rolling over the patterned substrates. Reversible binding between glycoprotein receptors known as selectins expressed on vascular endothelial cell surface and the selectin-ligands expressed on the cell surface result in cell rolling on the vasculature under shear flow [1-8]. Karnik et al have demonstrated that a single inclined P-selectin receptor pattern can be used to direct the trajectories of cells rolling HL60 cells on the surface [9]; this finding opens the possibility of microfluidic devices for simple, continuous-flow, and label-free separation and analysis of cells [10-12]. The concept of the separation device with multiple inclined receptor bands has been shown in Chapter 1 (Figure 1-6). Such devices have the potential for rapid diagnostic applications including detection of sepsis and inflammatory conditions that alter cell rolling behavior [13-15], separation and enumeration of different leukocytes from blood using rolling ligands with appropriate specificities [16], and labelfree analysis of the expression of cell surface ligands. Since cell rolling is non-invasive and mimics the natural cell homing process, these devices are also promising for separation or analysis of subpopulations of hematopoietic or mesenchymal stem cells and enrichment of circulating rare cells [17, 18]. Figure 3-1 shows a schematic illustration of the trajectories of cells rolling on a patterned substrate. Cells that track along the edges of these patterns are laterally displaced into the adjacent buffer stream, potentially enabling their separation. Quantitative understanding Part of the content in this chapter is based on the work published in Langmuir, 2011. 27(1), 240-249. 59 of the nature of cell rolling trajectories and lateral displacements on receptor-patterned substrates is prerequisite for realization of such devices. Although Karnik el al have demonstrated that HL60 cells could track along P-selectin, the effect of flow conditions and pattern inclination angle on cell rolling trajectories was not studied [9]. Furthermore, the distribution of edge tracking lengths and lateral displacements was not analyzed due to insufficient data obtained from a single patterned edge. Therefore, how the parameters of cell rolling relate to cell rolling trajectories and lateral displacements for such asymmetric patterns is poorly understood. For example, it is unknown to what extent cell rolling is affected by shear stress magnitudes or pattern inclination angles. Further, the nature of the detachment of cells after tracking along an edge is not known, and is it not yet established whether detachment of cells from such patterns is a random process that is unaltered by interaction history with the pattern itself. Systematic study of cell rolling trajectories along such well-defined receptor bands is therefore prerequisite for addressing these issues and enabling the development of label-free devices for separation or analysis of cells such as the device envisioned in Figure 1-6. In Chapter III, cell rolling trajectories of rolling HL60 cells onto P-selectin patterned substrate were studied. HL60 cell is widely used as a model to study leukocyte rolling [19-23]. Its surfaces express a specific ligand named P-selectin glycoprotein ligand-1 (PSGL-1), which binds reversibly to the receptor P-selectin to enable rolling in vivo and in vitro [19]. A technique based on microcontact printing (pCP) to pattern alternating ptm-scale lines of adhesive P-selectin regions with passivating poly(ethylene glycol) regions on a gold substrate was developed. The edge tracking lengths and rolling velocities of HL60 cells on these patterned substrates within a flow chamber at different edge inclination angles and shear stress magnitudes, and P-selectin incubation density were quantified in this Chapter. The distribution of edge tracking lengths and observation of re-attachment of cells were incorporated into a computational simulation tool developed by Suman Bose (MIT) to predict the trajectories of cells on a patterned substrate [10]. 60 Figure 3-1 Illustration of a typical cell rolling trajectory along the receptor pattern inclined an angle (a) to the fluid flow direction. The cell binds within the receptor line, and rolls in the direction of shear flow toward the pattern edge. The cell then tracks the edge to define an edge tracking length 1e, resulting in a net lateral displacement d, before detaching to continue along the direction of fluid flow before possible reattachment and rolling along a new receptor line. Cell rolling velocity v, along within the receptorfunctionalized line in the x-direction of fluid flow can be distinguished from the velocity ve along the line edge, where ve,,y is lateral velocity (the vertical component perpendicular to the streamlines and parallel to the lateral displacement, d). 3.2 Fabrication of P-selectin-Patterned Substrates 3.2.1 Materials Polydimethylsiloxane (PDMS; Sylgardl 84) was purchased from Dow Corning. Goldcoated glass slides were purchased from EMF Corp. All slides were cleaned with piranha solution prior to use (3:1 mixture of sulfuric acid (Sigma-Aldrich) to 30% hydrogen peroxide (Sigma-Aldrich)). (l-Mercaptoundec-11-yl)tetra(ethylene glycol) (PEG alkanethiol; HS(CH 2)1 (OCH 2 CH 2)40H; Sigma-Aldrich) was diluted in absolute ethanol (Pharmco-AAPER) at a concentration of 5 mM for microcontact printing. Recombinant human P-selectin (R&D Systems Inc.) and bovine serum albumin (BSA, Rockland Immunochemicals, Inc.) were diluted in 150 mM NaCl Dulbecco's phosphate buffered saline (DPBS, Mediatech Inc.). All materials employed in this study were used without further purification unless otherwise specified. 3.2.2 Fabrication of PDMS Stamp Microcontact printing stamps that defined the receptor pattern were fabricated in PDMS using an SU-8 molding process. The small line-patterned stamp (SS) (15 ptm line width 61 and 10 pm spacing between adjacent lines) was used to characterize the patterning process. The medium line-patterned stamp (MS) (70 gm stamping regions spaced 50 pm apart) and the large line-patterned stamp (LS) (100 gm stamping regions spaced 100 jim apart) were used study cell rolling behavior. 3.2.3 Fabrication of Patterned Substrates A schematic diagram of the patterning process is shown in Figure 3-2. Step 1: Microcontact printing (pCP) was used to form a alternating self-assembled monolayer (SAMs) of PEG molecules on the gold substrate. The PDMS stamp was first inked with PEG solution in ethanol (5 mM), dried, and pressed onto the surface to be patterned for 40 s. The surface was then rinsed with ethanol and dried under a stream of N2. Step 2: After selective deposition of PEG molecules, the substrates were incubated in P-selectin solution (15 jg/mL in DPBS, unless stated otherwise) using a perfusion chamber (Electron Microscopy Sciences) for 3 h at room temperature to pattern the remaining areas with P-selectin. The surfaces were then backfilled with BSA (1 mg/mL in DPBS) for 1 h to block non-specific interactions. Wstep 1 PEGG PEG Step 2 P4kd -eed PEG moeueP-selectin Figure 3-2 Schematic diagram for patterning of P-selectin on a gold substrate involving microcontact printing. Step 1: Selective deposition of PEG molecules on the gold surface. Step 2: Filling in of the uncoated surface with P-selectin. Direct microcontact printing (sCP) of proteins has been used widely to control the geometry of protein patterns on various planar surfaces [24-29], including printing of Pselectin to study neutrophil rolling [30]. However, it is possible that the protein will denature or lose bioactivity during the PDMS stamping process [31]. Additionally, transfer of the stamp material (PDMS) from the stamp to the surface during pCP can contaminate the patterned areas [32]. In the present method, after selective deposition of 62 PEG molecules, the gold substrate is patterned with P-selectin in the solution phase so the possibility of denaturation due to protein drying can be ruled out. 3.2.4 Substrate Characterization Atomic force microscopy (Veeco Dimension 3100; Tapping mode; scan rate: 1 Hz) and scanning electron microscopy (JEOL 6700; acceleration voltage 3.5 kV) were used to characterize the patterned surface geometry. All substrates for AFM and SEM characterization were placed in a vacuum chamber overnight before imaging to minimize residual solvent on the surface; no further coating was employed for SEM imaging. AFM images of P-selectin patterned using the small line-patterned stamp (SS) show clearly defined 10 im-wide lines of P-selectin with well-resolved, straight edges (Figure 3-3 (a), (b)). The medium line-patterned stamp (MS) was used to prepare surfaces for studying edge track lengths and rolling velocities. The sharp contrast between P-selectin regions and PEG regions confirms that the resulting patterns had well-defined edges over large areas as revealed by SEM (Figure 3-3 (c), (d)). Figure 3-3 Characterization of P-selectin patterned substrates. AFM images of 10 [tm wide P-selectin lines separated by 15 ptm wide PEG bands (after step 2), displaying 63 the contrast between P-selectin and PEG regions in (a) height and (b) phase, respectively. The phase image indicates a difference between the mechanical properties of the surface in the two regions. SEM images of surfaces after PEG printing (c) (step 1) and after Pselectin adsorption (d) (step 2), respectively, showing uniformity of patterning (brighter areas correspond to PEG regions). Scale bars are 5 pm in (a) and (b) and 100 tm in (c) and (d). 3.3 3.3.1 Cell Rolling on Asymmetric Patterns in a Flow Chamber Cell Culture HL60 myeloid cells (ATCC) were maintained in Iscove's Modified Dulbecco's Medium supplemented with 20 % fetal bovine serum (FBS) as per ATCC recommendation. Cells were cultured in 75 cm 2 polystyrene tissue culture flasks (BD Falcon) with the cell density maintained between 105-106 cells/mL (passage number 26, 27 and 29 were used in this study). 3.3.2 Experiment Setup A suspension of HL60 cells was flowed over the patterned surfaces in a rectangular flow chamber (Glycotech, Inc; width w = 1.0 cm; length = 6 cm; height h = 0.005 inch) as shown in Figure 3-4 with inclination angles of the receptor pattern of either a = 50, 100, 150 or 200 (see Figure 3-1) at room temperature of 24.5"C. A syringe pump (World Precision Instruments (WPI), SP230IW) was used to generate different flow rates between 75 and 300 pL/min, with corresponding shear stresses of 0.5-2.0 dynes/cm 2 (~0.05 to 0.2 Pa). Flow was laminar (Reynold's number Re ~ 0.1-3) and shear stress r was calculated using the plane Poiseuille flow equation T = 6pQ/wh 2 , where p is the viscosity, Q is volumetric flow rate, w is width of the flow chamber, and h is height of the flow chamber. For confirming the non-fouling properties of PEG, the injected cell concentration was ~106 cells/mL (high density). For the studies of edge tracking lengths and rolling velocity, the injected cell concentration was ~105 cells/mL (low density). An inverted microscope (Nikon TE2000-U) with a mounted camera (Andor iXon 885) was used to record HL60 rolling interactions with adhesive P-selectin substrates using a 4x 64 objective, typically at a rate of 1 frame per second for durations of 300 s with exposure time of 0.01-0.05 s. For each shear stress magnitude, pattern inclination angle, and Pselectin concentration, three independent experiments were performed. Data are presented as mean and standard deviation of the average values obtained from each experiment. Figure 3-4 Rectangular flow chamber for cell rolling on multiple asymmetric pattern study. The flow chamber was attached to the P-selectin-patterned substrate via a vacuum manifold to assemble the device. The thickness of the gasket determines the height of the chamber, here, height = 0.005 inch). 3.3.3 Results and Data Analysis Poly(ethylene glycol) (PEG) has been widely used in developing surfaces that prevent fouling coming from the proteins [33, 34]. Although previous studies suggests that only the high molecular weigh PEG (MW > 3500 Da) can resist protein adsorption [35], Prime and Whitesides demonstrated that self-assembled monolayers (SAMs) made from shortchain alkanethiols such as HS(CH 2)1 1(OCH 2CH 2 )nOH can be used in designing highly protein-resistant surfaces and they also reported that the number of the ethylene oxide groups can be as few as two [36, 37]. In this chapter, HS(CH 2)1 1(OCH 2CH 2)40H was used. To confirm the anti-fouling properties of PEG, a suspension of HL60 cells with high density (10 6 cells/mL) was flowed over the patterned surfaces (prepared by LS) in the flow chamber. The HL60 cells were observed to roll specifically in the P-selectin patterned regions as shown in Figure 3-5. These results confirm that the PEGfunctionalized regions on either side of P-selectin lines could block P-selectin adsorption and that P-selectin molecules retained their activity after being adsorbed to the exposed gold. In addition, experiments to characterize cell adhesion on PEG surfaces and surfaces 65 coated with BSA were performed, and none of cell-surface interactions was observed, further confirming that the observed cell-rolling interactions were due to P-selectin. PEG region P-selectin region Figure 3-5 The interaction of HL60 cells with P-selectin/PEG patterned substrate in the flow chamber. The substrate was prepared using large line-patterned stamp (LS). The edge inclination angle was 100 and the shear stress was 0.5 dyn/cm 2. The HL60 cells only interact with the P-selectin patterned region, not PEG patterned region, which confirms the anti-fouling property of PEG. For studies of the effects of edge inclination angle, shear stress, and the P-selectin incubation density on the edge tracking lengths and rolling velocities, the patterned substrates were prepared by medium line-patterned stamp (MS). The P-selectin patterned substrates were incorporated into a flow chamber for studying the rolling behavior of HL60 cells under different condition. The injected cell concentration was ~105 cells/mL (low density) to prevent cell-cell interaction Tracking of Cells. The image sequences were analyzed using a customized Matlab (Mathworks, Inc.) program that utilized a particle tracking freeware [38] to detect the cells and generate tracks along the patterned line edges. The procedure used to generate tracks has been described in Karnik et al's previous work [39]. A tracking criterion that required the cell displacement between consecutive frames was set to be less than 10 pim. Tracks with total length <40 ptm were not included in analysis of the distribution of cell responses, as these short tracks predominantly represented artifacts on the substrate such 66 as pinholes and in some cases unlinked fragments of a single track. These settings allowed for selective tracking of only the rolling cells, since velocities of the non-adhered freely flowing cells were much higher. Tracks generated by the software were randomly selected and inspected manually by comparing with the images to ascertain their accuracy. Figure 3-6 shows an example of actual tracks of HL60 cells rolling on Pselectin patterns at an edge angle of 100, shear stress of 0.5 dyn/cm 2 , and incubation concentration of 15 [tg/mL. The tracks clearly depict the change in direction of rolling HL60 cells that occurred when the cells encountered the inclined edges. Identification of Tracks that Encountered an Edge. Positions of the patterned edges were identified using the difference in contrast between the PEG and P-selectin regions as imaged using optical microscopy. The procedure used to identify tracks that encountered the edge was developed by Suman Bose and described in Langmuir 2011, 27(1) 240-249 [10]. Tracks with endpoints within 10 pm of the nearest edge were identified as having encountered an edge. As shown in Figure 3-6 inset, the tracks were subdivided into two segments at intersection point Pi - one that represented rolling inside the P-selectin line (green line), and another representing rolling on the edge (black line). The length travelled along the patterned line edge was defined as edge tracking length, 4, and calculated from the distance between the Pi and the end of the track. Rolling velocity was calculated by dividing the length of the segment by the time taken to traverse it. For each experiment, an array of edge tracking length (le), velocity on edge (vp) and velocity on plain region (vp) with one data set corresponding to each track was generated and used for further analysis. 67 Figure 3-6 Tracks of HL60 cells rolling on P-selectin patterns. The tracks (blue lines) on P-selectin lines (pink) were obtained by analyzing 300 images acquired at 1 fps using a customized Matlab code. The edge inclination angle was 100 and the shear stress was 0.5 dyn/cm 2 . Inset shows a track corresponding to a cell that first rolled inside the Pselectin line (green) in the direction of fluid flow and then tracked along the edge (black). Hopping was observed in our study: After cells detached form the edges of the band, most of them will reattach onto successive downstream band. However, the customized Matlab program used in this study can only identify the tracks of the cells on a single band but not multiple bands. The following studies of the effect of substrate parameters including edge angle, shear stress, and P-selectin incubation density on the rolling behavior of HL60 cells are based on the data extracted from the tracks on a single band. 3.3.3.1 Effect of Edge Angle on the Rolling Behavior of HL60 Cells The effect of the edge inclination angle a subtended by edges of the P-selectin lines with respect to the direction of fluid flow on the edge tracking length 4, the lateral displacement d, and the rolling velocities v, an ve, respectively, is shown in Figure 3-7. At a=5', HL60 cells rolled an average distance of more than 135 pm along the edges before detachment at a shear stress of 0.5 dyn/cm2 . Figure 3-7(a) shows that, as the edge angle was increased from 5* to 20' in 50 increments, the average edge tracking length 1e 68 decreased significantly (ANOVA, F = 18.403, p = 0.001). In other words, the ability of the cells to roll along the edges was reduced with increasing edge inclination angle (and increasing component of the fluid force on the cell directed away from the edge). Comparison of data pairs (post-hoc t-test) demonstrated statistically significant differences in 1e for every 5' increase in a. In contrast, Figure 3-7(b) shows that the lateral displacement d (ANOVA, F = 3.075, p = = 1e sina, did not show a significant trend with increasing a 0.091), and varied from 7.0 to 12.5 pm over this range of a. However, a statistically significant difference was observed between the lateral displacements at a =10 and 200 (post hoc t-test). This behavior can be understood in that although sina increases with increasing edge inclination angle, edge tracking length 1e concomitantly decreases. It is the magnitude of this lateral displacement that is more relevant to separation of cells by rolling on such a patterned substrate. In addition to altering the direction of cell rolling, asymmetric receptor bands can also alter the rolling velocity of the cells [39]. To examine the effect of a on rolling velocity, we quantified the average rolling velocity of cells within and along the edges of the Pselectin lines as a function of a at a fixed shear stress magnitude of 0.5 dyn/cm 2 (Figure 3-7(c)). The rolling velocity within the P-selectin line was in the range of v, = 2.9 - 3.6 tm/s, and was always less than that along the edge regions (ve = 4.4 - 6.0 pm/s). This can be understood in terms of the expectation that, as a increases, surface area of interaction and adhesion resistance to rolling between the cell and the surface decreases, leading to an increase in rolling velocity. Pairwise (t-test) statistical analyses show a significant increase in ve at large edge angles (15' and 20') compared to v,, consistent with previous observations [39]. The average rolling velocity on the edge ve increased from 4.4 ptm/s for an edge inclination angle of 5' to 6.0 pm/s at an edge inclination angle of 200, though this trend did not reach a degree of statistical significance (ANOVA, F = 3.55, p=0.067). In contrast, ve,, (lateral velocity, defined previously as the edge velocity component in the direction of lateral displacement d) increased significantly from 0.4 pm/s to 2.1 pm/s as a increased from 5" to 200. Thus, receptor bands characterized by large edge inclination angles (a=15', 20') led to greater lateral displacement of cells over a given rolling duration. 69 (a) (b) 300 p= 0.0055 w 250 , 20 p= 0.0697 2 p= 0.01 D=0.0423 15 200 p= 0.007 150 a. 100 p= 0.027 s0 10 5 0 5 (C) 10 15 Edge Inclination angle, a () V. p= p. P80.16 p3.0O88 r_ =L 5 (d) 8 6 20 4 p=0v19p= 0.043 0.11 ~ 15 10 Edge Incdhon angle, a() 0 pu 0.0005 3 p= 4 71 20 0.0007 p= 0.0023 p= 0.024 2 pp=0.017 0 1 2 A 0 5 10 15 20 Edge inclination angle, a (*) 5 10 15 20 Edge inclination angle, a (*) Figure 3-7 Effect of edge inclination angle a on rolling behavior of HL60 cells at a fluid shear stress magnitude of 0.5 dyn/cm2. Variation of (a) edge tracking length, le; (b) lateral displacement, d; (c) rolling velocities v, and v, within the P-selectin lines and on the edge, respectively; and (d) lateral velocity, ve,, (component of the edge rolling velocity in the direction of lateral displacement). Error bars represent one standard deviation, with n = 3 replicate experiments for each condition. 3.3.3.2 Effect of Shear Stress on Rolling Behavior of HL60 Cells Figure 3-8 summarizes the edge tracking length l,, the lateral displacement d, and the rolling velocities v, an v, as a function of the shear stress (,r = 0.5 dyn/cm 2 to 2.0 dyn/cm 2 ) at a fixed edge angle of 5*. The edge tracking length le varied in the range of 118.6-173.1 im over r = 0.5 to 2.0 dyn/cm 2 . However, there was no statistically significant effect of shear stress on le (ANOVA, F=2.119, p=O.176) (Figure 3-8(a)). The lateral displacement d varied between 10.3-15.1 ptm, again with no statistically significant dependence on the shear stress (Figure 3-8(b)). Similarly, Figure 3-8(c) shows that the rolling velocity within the P-selectin lines and on the edge also did not vary 70 significantly with shear stress (ANOVA, p=0.917 and p=0.165, respectively). This lack of dependence on shear stress is not surprising, given that cell rolling involves mechanisms at the cellular and molecular levels to regulate the rolling response over a range of shear stresses [40]. Similar independence of rolling velocity with shear stress has been observed before in the case of HL60 cells rolling on E-selectin [41, 42]: the rolling velocity of HL60 cells has been observed to increase with shear stress at low shear stress (r <0.5 dyn/cm 2 ) and reach a plateau at higher shear stresses. Our experiments indicate that similar to rolling within the P-selectin line, shear stress also does not have a significant effect on the rolling behavior of HL60 cells on asymmetrically patterned edges within x = 0.5 to 2.0 dyn/cm 2. (b) (a) 250 200 .- ZU E 15 150 10 k 100 5 ~50 W 0 0 0.5 (c) 8 6 0.5 2.0 1.0 1.5 Shear stress, T (dyn/cm2) (d) o.*p-0.033 V13 Ip. 0.088 p 0.012 4 p= 0. 3 1.5 2.0 1.0 2 Shear stress, Y(dyn/cM ) 0.6 0.5 ~ 0.4 0.3 4 0.2 S2 0.1 -'0 0.5 1.5 1.0 Shear stress, Y (dyn/cm 2) 0.5 2.0 1.0 1.5 2.0 2 Shear stress, t (dyn/cm ) Figure 3-8 Effect of shear stress T on rolling behavior of HL60 cells at an edge inclination angle of 5'. Variation of (a) edge tracking length, 1,; (b) lateral displacement, d; (c) rolling velocities v, and v, within the P-selectin lines and on the edge, respectively; and (d) lateral velocity, ve,,y. The effect of shear stress on rolling behavior of HL60 cells is not statistically significant as observed in (a), (b), (c) and (d). Error bars represent one standard deviation, where n = 3 replicate experiments for each condition. 71 3.3.3.3 Effect of P-selectin Incubation Concentration on Rolling Behavior of HL60 Cells Apart from shear stress and edge inclination angle, the P-selectin density on the surface may be expected to affect the trajectories of cells rolling on the inclined edges. Increasing P-selectin density on a glass surface under high shear stresses (~20 dyn/cm 2) has been observed to decrease the rolling velocity of HL60 cells, but receptor density did not modulate the rolling velocity under low shear stress (< 2 dyn/cm 2 ) [21]. When the Pselectin incubation concentration was varied in the range of 5 to 30 ptg/rnL while maintaining an incubation time of 3 h, no adhesion (5, 10 pg/mL), non-uniform rolling (11, 12, 13, and 14 pg/mL), robust rolling (15, 20, and 25 gg/mL), and firm adhesion at 30 pg/mL for a shear stress of 0.5 dyn/cm 2 were observed. Thus, the range of P-selectin incubation concentrations that resulted in a useful rolling response was 15 to 25 pg/mL. Therefore, the edge tracking length and cell rolling velocities in this range of P-selectin concentrations were then characterized as shown in Figure 3-9. Interestingly, there was no significant change in rolling velocity with change of P-selectin incubation concentration: the average rolling velocities inside the bands (vp) were 2.78±0.57, 2.44±0.64, and 2.67±0.67 pm/s for P-selectin concentrations of 15, 20, and 25 pg/mL at shear stress of 0.5 dyn/cm 2 , respectively. Similarly, there is no significant change in the cell behavior along the edge including edge tracking length 1e, lateral displacement d, edge rolling velocity ve, and lateral velocity ve,,y. These results are in agreement with previous observation of less change in rolling variation with a variation of ligand density under low shear stress (< 2 dyn/cm 2 ) by Dong et al [21]. These results indicate that when P-selectin is directly immobilized on a gold substrate, the rolling behavior along the edge cannot be controlled as easily as that by changing the edge inclination angle. 72 (a) (b) ~80 a= 100 a= 200 ~60 15 12 E a= 100 *= 200 n 9 CD 40 6 20 3 0 25 20 15 P-selectin Incubation concentration (pg/mL) 15 20 25 P-selectin Incubation concentration (pg/mL) (c) (d) 8 3 o=10 a a= 10* = 20' a =20* 7A 2 0 0 25 20 15 P-selectin incubation concentration (pg/mL) 25 20 15 P-selectin incubation concentration (pg/mi) Figure 3-9 Effect of P-selectin incubation concentration on rolling behavior of HL60 cells. Variation of (a) edge tracking length, le; (b) lateral displacement, d; (c) rolling velocities v, and v, within the P-selectin lines and on the edge, respectively; and (d) lateral velocity, ve,y at edge inclination angles a = 100 and 200 and shear stress of 0.5 dyn/cm 2 . Error bars represent one standard deviation, where n = 3 replicate experiments for each condition. 3.3.3.4 Detachment of Cells Rolling Along an Edge Can be Described by a Poisson Process. While the average edge tracking length and lateral displacement are useful to elucidate the effects of edge angle, shear stress, and P-selectin incubation concentration on cell rolling, knowledge of the distributions of the distance rolled along the edge and the lateral displacement is important for predicting the eventual distribution of a population of cells in a separation device. Similarly, this knowledge is required to elucidate the number of edge tracking events that must be observed to distinguish between cell phenotypes by observing rolling on the patterned substrates. Therefore, the distribution of the edge tracking lengths was examined, with the aim of developing a model that would 73 serve as a tool to predict cell rolling trajectories and their spread, with direct implications on analysis resolution and separation efficiency. For all the experiments conducted, distributions of the edge tracking length exhibited a decaying exponential characteristic similar to that expected from a Poisson process as shown in Figure 3-10. Poisson process occurs when individual events within the process are random in nature with uniform probability of occurrence independent of history. It is well known that cell rolling is a stochastic process involving discrete adhesive interactions between the cell and the surface [43-46]. Rolling cells continuously form adhesive contacts with the surface with receptors localized at the tips of extensible structures known as microvilli [47]. In the case of cells rolling along an edge, the region of overlap between the cell's contact area and the receptor-coated region is the site where new cell-surface adhesions are formed. As the cell tracks along an edge, new cell-surface adhesions are continuously formed in the region of overlap between the cell's contact area and the receptor-coated region. The cell will detach from the surface when a new adhesive interaction fails to form before the last adhesive interaction is broken under shear flow. If the probability of formation of new adhesive interactions in a given period is constant (i.e., independent of the past rolling history of each cell and/or uncorrelated among cells within a population), detachment is expected to be a random event and the distance traveled by each cell along the edge is expected to follow a Poisson distribution. To test this hypothesis, the cumulative distribution function (CDF) was calculated from the data and fit it with a Poisson distribution (Figure 3-10) given by: C(le)= I-exp( L (Eq. 3-1) Here, k is the mean value of the Poisson distribution. In the ideal case where detachment is a random process, k will approach the mean value of the edge tracking length. The CDF does not involve any arbitrary bin widths and therefore allows for an objective comparison of the actual distribution of the edge tracking lengths with that predicted for a Poisson process (Eq. 3-1). The CDFs displayed in Figure 3-10 do not begin at the origin 74 because edge tracking lengths <10 pm could not be resolved by the present video frame rate and magnification; these unresolved lengths were used for calculation of the CDF itself but were not used for fitting the Poisson function. The Poisson distribution described by Eq. 3-1 well fit the CDF for all edge inclination angles considered, and the mean value of k obtained by fitting to the CDF also accurately predicted the observed edge tracking length histograms (Figure 3-10(a), inset) at different edge inclination angles. Additionally, to confirm that the process of detachment of cells from the edge was indeed a Poisson process for a given cell, k under different experimental conditions was calculated and it was compared to the measured average edge tracking lengths (Table 31). A strong correspondence between k and the average edge tracking length was found from the experiments; even at different shear stress magnitudes, the maximum difference was found to be within 7%. These data suggest the fact that detachment of these HL60 cells rolling along an edge is indeed a random process. Interestingly, the average value of k obtained by fitting the CDF to the Poisson distribution showed an exponential decrease with increasing edge inclination angle (Figure 3-10(b)). This observation provides an empirical relation between the edge angle and the average edge tracking length, while the distribution of the lengths itself can be modeled using a Poisson process. The Poisson mean values ' obtained from the empirical fit (in Figure 3-10(b)) are summarized in Table 3-1 and they were compared to the measured average edge tracking lengths. The maximum difference was found to be within 5% which suggests that this exponential dependence combined with modeling cell detachment as a Poisson process can be used to interpolate the average edge tracking length as well as its distribution for any edge angle between 5' and 20'. The prediction of the variation of lateral displacement with edge inclination angle was made based on this exponential fitting that agreed well with the experimental data (Figure 3-10(c)). Interestingly, the curve predicts an optimal edge inclination angle between 50 and 100 that maximizes the lateral displacement for rolling along a single edge. This prediction explains why a statistically significant difference was not observed between lateral displacements at edge inclination angles of 5' and 100 (Figure3-6). The average 75 lateral displacement exhibits a maximal value at the edge inclination angle between 5' and 10'. (a) - 1.0 2G . W0 1150 I U 20 40 10 20 0 0.5 150 - C.) 0 200 400 1V 200 200 15* 400 2W 1501 100 100 so 50 W 0 Edge Ucking length, 0.0 200 0 400 800 Edge traddng length, Is (Jim) (b) (c) logX = -0.126a+5.489 R2=0.9%62 100 100 ) 1000 800 15 10F 5 O Experkmental data Pmedictio from Poisson process -- 10 50 le (Am) 0 5 15 10 Edge inclination angle, a 0 0 20 5 20 10 15 Edge inclination angle, a o 25 30 Figure 3-10 Detachment of cells rolling along an edge is well described by a Poisson distribution. (a) Cumulative distribution function of edge tracking lengths l, (filled triangles) was fitted to a Poisson distribution described by Eq. 3-1 (solid lines). Insets show the frequency distribution of the experimentally measured edge tracking lengths, along with that predicted by the Poisson distribution fit to the CDF (solid lines). Colors indicate different inclination angles a of the receptor pattern. Representative results are shown for only one experiment for each a. (b) Variation of the average value of k with edge inclination angle is well described by a linear fit on a semi-log plot (solid line). (c) Variation with the edge inclination angle of the average value of the lateral displacement (solid line) obtained from the empirical fit shown in (b) along with the experimental results (open circles). Error bars in (b) and (c) represent one standard deviation. Shear stress is 0.5 dyn/cm 2. Table 3-1 Comparison of the experimental average of 1e, Poisson average value of k, and the Poisson mean value ' from the empirical fit. n = 3 replicate experiments. 76 Data were obtained by fitting to a Poisson distribution with experimentally measured average edge tracking length. Shear stress is 0.5 dyn/cm 2 . Experimental Poisson mean value Edge angle average Poisson mean % Error from the empirical fit % Error a (0) /,±SD (jim) A±SD (pm) 100(A-I./)I. (pm) A'(pm) 100(A'-,/)I. (pm) 5 135.5±36.3 134.1±33.2 -0.98% 128.9 -4.87% 10 72.2±17.2 66.2±21.7 -8.35% 68.6 -4.92% 15 37.2±7.6 34.7±9.4 -6.60% 36.6 -1.72% 20 20.3±3.7 20.3±3.9 0.11% 19.5 -4.08% Prediction of cell trajectories on a receptor-patterned substrate. The experimental results obtained in this study enable development of a simulation tool for predicting the trajectories and distribution of cells rolling across a substrate patterned with asymmetric P-selectin lines. The experimental observations were incorporated into a simulation of the lateral displacement of HL60 cells as they encounter multiple edges of P-selectin. A Monte Carlo simulation was developed and performed by Suman Bose; the detailed procedure is described in Langmuir 2011, 27(1) 240-249 [10]. The simulated results agreed well with the experimental distribution as shown in Figure 3-11(a) despite the low number of experimental data points available. Although a comparison of distribution over more number of edges is desirable, the study was limited by the field of view of the camera, which could only accommodate approximately three edges of the patterned bands. A second Monte Carlo simulation was performed by Suman Bose again to examine the net lateral displacement and resulting distributions of the cells (injected at a particular location (0, 0)) at a longitudinal distance of 1 cm downstream of the origin (similar to Figure 1-6). The detailed procedure of this simulation can be found at Langmuir 2011, 27(1) 240-249 [10]. In the simulation study, the device was assumed to be patterned with parallel P-selectin lines having a width of 50 im and a gap of 70 ptm between adjacent lines (similar to the pattern used in experiments). The cells were assumed to roll on all the P-selectin lines that they encountered with the edge tracking lengths described by a Poisson distribution with the mean I as obtained earlier. Figure 3-11(b) summarizes the 77 probability density functions of the net lateral displacement, defined here as the sum of all displacements that a given cell acquired over the longitudinal travel distance of 1 cm. The net lateral displacement increased with increasing edge inclination angle a from 50 to 150, with little change as a increased from 15' to 200. The net lateral displacement showed maximum sensitivity to the edge inclination angles between 50 to 100. Increasing the edge inclination angle a increases the total number of edges encountered by the cells, but simultaneously decreases the mean lateral displacement on each edge (ksina) (Figure 3-10(c)). These opposing effects resulted in the net lateral displacement exhibiting an increase till a = 150, and little change between 150 and 200. With increasing number of edges encountered, the distribution of cells is seen to approach a Gaussian distribution, consistent with the central limit theorem. Interestingly, although a = 100 and 200 yield similar net lateral displacements, it is noteworthy that the Gaussian spread is smaller for a = 200 due to the larger number of edges encountered. It is noteworthy that although the distribution of cells approaches a Gaussian form, it is not exactly Gaussian as some of the cells in the distribution are rolling along the edge; these cells create "spikes" in the distribution at specific positions where the edges intersect the axis (at a longitudinal travel distance of 1 cm). 78 (a) 0.05 0aSimulated Result 0.04 51 V Expreimental Result 0.03 0.02 (L 0.01 LL-I - 0.00 20 0 rm 100 60 80 40 Net lateral displacement (pm) (b) 120 50 (81.64 ± 21.95 pm) 0.01 0.00 I-" I 0.01 0.00 I 100 (157.34± 38.30 pm) 150 (184.81± 37.18 pm) 0.01 C. 0.00 i 200 (190.89±33.59 pm) 0.01 0.00 0 50 200 250 100 150 Net lateral displacement (pm) 300-300 Figure 3-11 Prediction of cell trajectories on a receptor-patterned substrate (a) Probability distribution of net lateral displacement of HL-60 cells after rolling on three consecutive bands of P-selectin patterns as obtained from Monte Carlo simulations (shaded area) and experimental observations. a = 200 (shear stress 0.5 dyn/cm 2). (b) Prediction of the downstream distribution of HL60 cells rolling on patterned P-selectin in a separation device. (Courtesy of Suman Bose) The study of the effects of substrate parameters on the cell rolling trajectories has led to realization of a microfluidic device with multiple P-selectin bands of edge inclination 79 angle of 150 and this device has been demonstrated to be able to separate neutrophils directly from the blood [12]. 3.4 Optimization of Device Design Next, the results of cell rolling parameterization are used to predict optimal design features of the cell separation device that will maximize separation of a cell subpopulation that binds and rolls transiently along the inclined band pattern. Chapter II discussed how cells flowed inside the microchannel and settling distance (xs) was identified as a function of shear stress. An empirical equation was developed to predict cell capture probability from the free stream of fluid, as shown in Figure 3-12 (a) (left). In the first part of Chapter III, the edge tracking length for rolling cells on a single adhesive band was characterized. The effective chamber length (xe) was defined as the chamber length required for settled cells to be captured by the receptors and then deflected as shown in Figure 3-12 (a) (right). Both settling distance (x,) and effective chamber length (xe) should be considered when chamber length is designed. Based on experimental data from Chapter II and Chapter III, device capture efficiency (Pd), total lateral displacement (d,), and chamber length for a P-selectin-patterned microfluidic system can be predicted. The prediction enables study of sensitivity of device design parameters including edge inclination angle (a), band width (w), and shear stress 80 (T). pf= capture probability fron free stream by an incli ned pattern (a) x,= settling distance L (b) 4 V x, = effecttve chamber length L =chamber length= x,+x, Pd= device capture efficiency 2LJ W "P dt=total lateral displacement Device design (a, w, g, r) Figure 3-12 Optimization device design. (a) Schematic diagram of cell settling, capture, rolling on the patterns. (b) Schematic diagram of a device. Device capture efficiency, total lateral displacement, and required chamber length can be modeled based on different device design parameters. Device performance such as device capture efficiency (Pd), and total lateral displacement (dt) depends on two features: the number of the bands comprising the inclined pattern of a microfluidic device, and the capture efficiency of the cells by receptor bands. The number of the bands (n) inside a microfluidic device can be determined by: n ~Xe x (Eq. 3-2) sin a where x, is the effective chamber length, w is the band width, g is pattern gap, and a is edge angle as shown in Figure 3-12(a). 81 Device capture efficiency (Pd) is the capture efficiency of cells by a device or capture efficiency by "n" number of bands patterned inside the device and is defined as: pd =1-(Ipf)" (Eq. 3-3) where pf is the capture probability from the free stream by a single receptor band. The empirical equation (Eq. 2-7) developed in Chapter II can be applied to calculate pf here. To study total lateral displacement of a cell interacting with multiple bands, hopping of cells from one band to the next needs to be considered. This unique hopping behavior may affect the capture efficiency of the cells by receptor bands. The effect of hopping behavior can be studied by considering two extreme cases: (1) The first case is not to consider hopping, which means recapture probability by downstream bands (p,) is equal to capture probability from the free stream (pr). (2) The second case is to consider hopping and assume p,= 1.0 as shown in Figure 3-13 (a). This case implies that all cells hop to other downstream bands when detached from upstream bands When hopping events are not considered, the capture probability by any one of the bands inside the device is pf. The capture probability byj bands when a cell is travelling inside a device of n bands is Ci x pfj x (1- pf)"-1 . Total lateral displacement (d) when hopping events are not considered can be derived as: d, =< d>x (>Cj x Pfj'x (1 - Pf)n-ji xj f (j=0 (Eq. 3-4) where <d> is the average lateral displacement of a cell along a single band edge. The value of <d> assumed here was obtained from experimental observations discussed in this chapter. C' representsj combinations of n elements. Assuming cells will continue hopping and be recaptured by successive downstream bands after they detached from the edge of upstream bands (p, = 1.0), the probability of 82 captured by the first band is pf and the capture probability by ith band is (1- Pj )I1 X p1 . The number of the bands that a cell will roll on after it is captured by ih band is (n +1- i) . Therefore, the total lateral displacement (d,) can be derived as: d, =< d > x ( -p)'' xpf (Eq. 3-5) x(n +1-i) i=1 For the device design of band edge inclination angle a = 150, band width w = 10 pm, gap width g = 10 gm and shear stress T = 0.25 dyn/cm 2 , the probability of attachment of cells from the free stream can be predicted to be 0.03 from Eq. 2-7. Considering two cases: (1) P, = 1.0 and (2) pr = pr = 0.03, the predicted total lateral displacements at different effective chamber length are shown in Figure 3-13(b). It is clear that the hopping plays a critical role in enhancing the overall lateral displacement. The total lateral displacement can increase by several orders of magnitude depending on the chamber length. (a) P,= recapture probability by downstream pattern (hopping probability) P1 = cap ture probab ility from free stream by an pattern dt, total lateral displacement "W %VWIMWU"=IV VW (b) E I -" 6000 ".. 4000 C' 2000 Pr Pf= 0.03 --%185gm 0 PS 0 10 20 30 40 Effective Chamber Length, x, (cm) 83 50 Figure 3-13 (a) Schematic diagram showing cell hopping onto downstream band. (b) Variation of effective chamber length on total lateral displacement when hopping events were (pr = 1.0) and were not (pr = pf = 0.03) considered for the pattern design of a = 150, w = 10 ptm, g = 10 pIm, and T = 0.25 dyn/cm 2 . The objective of this section is to study optimization of the device design (a, w, and T) by integrating experimental data into device performance prediction. Since cell hopping among adhesive bands was observed from the experiments with HL60 cells in the flow chamber (Section 3-3), it is reasonable to consider hopping in this prediction study and to simplify the system, p, is considered to be 1.0 in the prediction model discussed in this chapter. The effect of a, w, or T on the device performance including device capture efficiency (Pd) and total lateral displacement (dt) can then be predicted by Eq. 3-3 and Eq.3-5, respectively. Since simple miniaturization and massive parallelization enable high-throughout microfluidic systems and enhance separation potential, one of the aims for many microfluidic systems is to minimize chamber length [48, 49]. Therefore, the minimum required chamber length (xe) for targeting certain Pd and d, was identified as a function of a, w, or r. The minimum x, was used as the indicator to determine the sensitivity and performance of the P-selectin patterned device. In other words, device design and operation parameters that would minimize device length and maximize separation of a specific cell population were quantified. 3.4.1 Effect of Band Width (w) on Device Performance The device capture efficiency (Pd) and the total lateral displacement (d) at different effective chamber length (xe) were predicted by Eq. 3-3 and Eq.3-5, respectively, at w varying from 10 pm to 50 tm and a = 15', g = 10 pm, and r =0.25 dyn/cm 2. The band width around 10 pm was the limit achievable via the microcontact printing used in this thesis. The predictions are shown in Figure 3-14. Pd increases as the adhesive band width increases, while d, decreases as the band width increases. To target at Pd > 90% and dt > 850 pm, for example, the required xe that meet each criterion and both criteria are listed 84 in Table 3-2. The criteria of device capture efficiency of 90% and total lateral displacement of 850 pm used in the prediction of optimal design were chosen arbitrarily to illustrate the effect of substrate parameters on device performance. It is clearly shown that the required effective chamber length that meets both criteria increases as band width increases. This study suggests that to simply miniaturize and massively parallelize the separation system, the band width should be minimized. However, the value of the minimum band width (w.), which allows the stable edge tracking, is not identified in this thesis. This value is expected to be related to the adhesive footprint (effective contact area) of the cells. For example, the neutrophil footprint is on the order of 10 pm [50]. Thus, although there is motivation to decrease patterned band width below the limits achievable via the microcontact printing used in this thesis, target widths will also be limited by cell-adhesive band interactions that require further study. (a,w, g, r)=(1s,10,10, O.s) (15, 20, 10, O.S) (15, 30, 10,0.5) (15, 40, 10, O.5) (15, 50,10,0.5) -8, 4-OQ + -Q+ 4000 - 100 E 80 3000 E _ - 2000 9JA ~40/ e20 j a -1000 0 0 0 4 8 12 16 20 24 28 32 Effective Chamber Length, x, (cm) Figure 3-14 The relationship between device capture efficiency (Pd) and effective chamber length (x,) and the relationship between total lateral displacement (dt) and effective chamber length (xe) when w varies from 10 pm to 50 pm with a = 150, g = 10 ptm, and T = 0.25 dyn/cm 2 . 85 Table 3-2 Prediction of required effective chamber lengths (x,). The range of x, which target at Pd > 90%, d, ? 850 pm and both criteria (Pd 90% and d, ? 850 pm) when w varies from 10 pm to 50 pm with a = 15*, g = 10 pm, and r = 0.25 dyn/cm 2 . 3.4.2 w (Pm) x, (cm) @ Pd = 90% x, (cm) @dt = 850 gm Required x, (cm) that meet both Pd (90%) and dt (850 pm) 15 20 30 40 50 6-8 4-6 4-6 4-6 4-6 8-10 12-14 14-16 18-20 22-24 8-10 12-14 14-16 18-20 22-24 Effect of Edge Inclination Angle on Device Performance The device capture efficiency (Pd) and the total lateral displacement (dt) at different effective chamber lengths (xe) were predicted when a varied from 50 to 250 with fixed w = g =10 pm, and T = 0.25 dyn/cm 2 and shown in Figure 3-15. The Pd is the same for all edge inclination angles considered, since n and pf are the same for all cases. The maximum d, per unit effective chamber length was identified to be between a = 150 and 200. To target at Pd > 90% and d, > 850 pm, for example, the required x, that meet each criterion and both criteria are listed in Table 3-3. It is clearly shown that the required effective chamber length that meet both criteria is minimized at a = 15'-20'. This conclusion is consistent with the results from Monte Carlo simulations described in 3.3.3.4 and suggests that using a device design of a = 15'-20' can maximize separation potentials. 86 (a, w,gr)=(5, 10, 10,0.s) - + (10,10, 10,0.5) (15, 10, 10,0.s)-r (20,10,10,0.5)-e (25,10,10,05) -0+ 4000E 100 .4 80 3000 C E ZI 60 2000 ILU u 20 - 000 1000.g > 0 0 -i 0 4 8 12 16 20 24 28 32 Effective Chamber Length, x, (cm) Figure 3-15 The relationship between device capture efficiency (Pd) and effective chamber length (xe) and the relationship between total lateral displacement (d,) and effective chamber length (xe) when a varies from 5 * to 250 with w = g =10 stm, and T = 0.25 dyn/cm 2 . Table 3-3 Prediction of required effective chamber lengths (xe). The range of x, which target at Pd ? 90%, d, 850 stm and both criteria (Pd ? 90% and d, > 850 sIm) when a varies from 5 0 to 250 with w = g =10 pm, and r = 0.25 dyn/cm2 . Required x, (cm) that a (0) Xe (cm) @ Pd = 90% Xe (cm) @dt = 850 pm meet both Pd (90%) and dt (850 ptm) 10 15 20 25 3.4.3 10-12 8-10 8-10 10-12 4-6 4-6 4-6 4-6 10-12 8-10 8-10 10-12 Effect of Shear Stress on Device Performance Figure 3-16 shows the relationship between device capture efficiency (Pd) and effective chamber length (xe), and the relationship between total lateral displacement (d,) and effective chamber length (xe), when r varies = 0.25 dyn/cm 2 to 1.0 dyn/cm 2 with fixed a = 150 and w = g =10 gm. Both Pd and d, decrease as shear stress increases. To target Pd > 90% and dt > 850 im, for example, the required effective chamber lengths that meet each 87 criterion and both criteria are listed in Table 3-4. It is shown that the required effective chamber length that meets both criteria increases by about 140 cm from T =0.25 dyn/cm 2 to r = 1.0 dyn/cm 2 for this particular design target. On the other hand, in the study of cell settling in the microfluidic channel (section 2.2), the result shows the settling distance increases as the shear stress increases. The total chamber length (L) is considered as the effective chamber length plus the settling distance as shown in Figure 3-12 (b). Both of the studies (in this section and section 2.2) clearly indicate that separation chamber length is very sensitive to shear stress. Specifically, lower shear stress results in higher capture efficiency and smaller settling distance and therefore better separation performance for P-selectin-based substrates. However, vacuum leaks made it difficult to examine shear stress below 0.25 dyn/cm 2 for this particular experimental setup used in this thesis. Although the optimum shear stress was not identified in this study, the shear stress of 0.25 dyn/cm 2 was experimentally identified to ensure cell rolling and separation occurs in current experimental system. On the other hand, it has been widely studied that Selectin molecules mediate shear threshold phenomenon. This phenomenon through P-selectin or E-selectin is less evident than through L-selectin [51-53]. Therefore, the conclusion drawn from current study system that smaller shear stress results in better device performance may not be valid for L-selectin-based systems. For example, Alon et al have demonstrated that leukocytes only accumulated on the L-selectin-coated surface above a threshold shear of 0.3 dyn/cm 2 and as shear stress increased above this threshold, the number of accumulation of cells increased and reached a maximum at an optimal shear about 2 dyn/cm 2 , and then decreased as shear increased further [53]. It can be inferred that the capture efficiency of a device would be affected by shear threshold phenomenon for a L-selectin-based system and it may maximize at shear stress around 2 dyn/cm 2 . However, the settling distance increases with increasing shear stress, which is independent of the molecules coated on the substrates and shear stress, may affect edge tracking length significantly. Therefore, to identify the optimum shear stress for a L-selectin-patterned microfluidic system, the 88 studies of cell capture by L-selectin-coated-substrates and edge tracking length at different shear stress by inclined L-selectin bands need to be analyzed. (a, w, g, r)= (15, 10, (15, 10, (15, 10, (15, 10, (a) 10, 0.25)*+'f 10, 0.50)+410, 0.75) "Q" 10, 1.0) +- S100 80 160 40 20 U 0 0 4 8 12 16 20 24 28 32 Effective Chamber Length, x, (cm) (b) ,~4000 E 3000 2000 o 1000 10 q 0 0 -0 4 8 12 16 20 24 28 32 Effective Chamber Length, x, (cm) Figure 3-16 The relationship between device capture efficiency (Pd) and effective chamber length (xe) and the relationship between total lateral displacement (d,) and effective chamber length (xe) when T varies from 0.25 dyn/cm 2 to 1.0 dyn/cm 2 with a = 150 and w = g =10 pim. Table 3-4 Prediction of required effective chamber lengths (xe). The range of x, which target at Pd > 90%, dt > 850 sm and both criteria (Pd > 90% and d, ? 850 Pim) when T varies from 0.25 dyn/cm 2 to 1.0 dyn/cm 2 with a = 15* and w = g =10 pm. 89 r (dyn/cm 2 ) Xe (cm) @ Pd = 90% 0.25 0.5 0.75 1 x, (cm) @d, = 850 rn 6-8 Required x, (cm) that meet both Pd (90%) and dt (850 prm) 8-10 12-14 44-46 142-144 10-12 18-20 30-32 8-10 12-14 44-46 142-144 Optimization design parameters including edge inclination angle a, band width w, gap width g, and shear stress T, for a miniaturized device of a chamber length of L, with device capture efficiency Pd, and total lateral displacement d, were studied in this thesis (Figure 3-17). Some future guidelines for the device optimization based on the studies of optimization design parameters are: 1. Decreasing shear stress increases device capture efficiency, which in turn minimizes chamber length. 2. Increasing probability of attachment from the free stream can enhance device capture efficiency. For example, using flow-focusing design to force cells to interact with substrates. 3. Device capture efficiency and total lateral displacement can be increased by increasing the number of bands. Decreasing band width and/or gap width increases the number of bands within a device for a given chamber length. 4. Further reductions in band width and gap width require additional studies of minimums required for both efficient edge tracking and hopping. 5. Optimum edge inclination angle is a = 15'-20', which allows maximizing total lateral displacement for a given chamber length or minimizing chamber length for targeting at certain total lateral displacement. 90 Device design w, g, r) Pd= device capture efficiency hopping d =total lateral displacement (r = shear stress) L = chamber length = x,+x, Figure 3-17 Device optimization parameters including edge inclination angle a, band width w, gap width g, and shear stress T, for a miniaturized device of a chamber length of L, with device capture efficiency P, and total lateral displacement d,. x, is settling distance and xe is effective chamber length. 3.5 Conclusion In summary, adhesive patterned substrates with multiple well-defined P-selectin patterns have been designed by a facile, versatile method utilizing microcontact printing. These substrates were incorporated in a flow chamber for studying HL60 cell rolling behavior including quantification of edge tracking lengths and cell rolling velocities. Among the parameters (edge inclination angle, shear stress, and P-selectin incubation density) considered, the pattern edge inclination angle modulated the cell rolling trajectory most strongly; in fact, the edge tracking length decreased exponentially with increasing edge inclination angle. In addition, the nature of cell rolling and detachment along the edge was consistent with a Poisson process. This correlation suggests that detachment of rolling cells from receptor-functionalized edges is a random process that is not disrupted measurably by cell-surface interactions over the device timescales considered. Experimental characterization of cell rolling enabled the development of a computational Monte Carlo tool to simulate the trajectories of cells, and showed that the optimum edge inclination angle is 15'-20'. Experimental characterization of edge tracking length on a single edge (in Chapter III) combined with study of capture probability from the free stream (in Chapter II) allowed the device performance to be predicted. The prediction results suggest that reducing shear stress, decreasing band width (and/or gap width), and 91 using an optimum edge angle of 15 -20' can enhance overall efficiency of cell separation for a miniaturized device. 3.6 Chapter Reference [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] G. I. Bell, Models for Specific Adhesion of Cells to Cells. Science, 1978. 200(4342): p. 618. R. Alon, D. A. Hammer, and T. A. Springer, Lifetime of the P-SelectinCarbohydrate Bond and Its Response to Tensile Force in Hydrodynamic Flow. Nature, 1995. 374(6522): p. 539. D. N. Granger and P. Kubes, The Microcirculation and Inflammation Modulation of Leukocyte-Endothelial Cell-Adhesion. Journal of Leukocyte Biology, 1994. 55(5): p. 662. L. A. Lasky, Selectin-Carbohydrate Interactions and the Initiation of the Inflammatory Response. Annual Review of Biochemistry, 1995. 64: p. 113. B. T. Marshall, M. Long, J. W. Piper, T. Yago, R. P. McEver, and C. Zhu, Direct Observation of Catch Bonds Involving Cell-Adhesion Molecules. Nature, 2003. 423(6936): p. 190. T. F. Tedder, D. A. Steeber, A. Chen, and P. Engel, The Selectins - Vascular Adhesion Molecules. Faseb Journal, 1995. 9(10): p. 866. G. A. Zimmerman, Two by Two: The Pairings of P-Selectin and P-Selectin Glycoprotein Ligand 1. Proc Natl Acad Sci U S A, 2001. 98(18): p. 10023. R. P. McEver and C. Zhu, Rolling Cell Adhesion. Annu Rev Cell Dev Biol, 2010. 26: p. 363. R. Karnik, S. Hong, H. Zhang, Y. Mei, D. G. Anderson, J. M. Karp, and R. Langer, Nanomechanical Control of Cell Rolling in Two Dimensions through Surface PatterningofReceptors. Nano Lett, 2008. 8(4): p. 1153. C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Examining the Lateral Displacement of H160 Cells Rolling on Asymmetric P-Selectin Patterns. Langmuir, 2011. 27(1): p. 240. C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Studying Cell Rolling Trajectorieson Asymmetric Receptor Patterns.J Vis Exp, 2011(48). S. Bose, R. Singh, C. Shen, M. Hanewich-Hollatz, C. H. Lee, D. M. Dorfman, J. M. Karp, and R. Karnik, Affinity Flow Fractionation of Cells Via Transient Interactionswith Asymmetric Molecular Patterns. Scientific Reports, 2013(3): p. 2329. G. C. Ibbotson, C. Doig, J. Kaur, V. Gill, L. Ostrovsky, T. Fairhead, and P. Kubes, Functional Alpha4-Integrin: A Newly Identified Pathway of Neutrophil Recruitment in CriticallyIll Septic Patients.Nat Med, 2001. 7(4): p. 465. K. L. Davenpeck, M. E. Brummet, S. A. Hudson, R. J. Mayer, and B. S. Bochner, Activation of Human Leukocytes Reduces Surface P-Selectin Glycoprotein Ligand-1 (Psgl-1, Cd162) and Adhesion to P-Selectin in Vitro. Journal of Immunology, 2000. 165(5): p. 2764. 92 [15] [16] [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] D. E. Lorant, R. P. Mcever, T. M. Mcintyre, K. L. Moore, S. M. Prescott, and G. A. Zimmerman, Activation of Polymorphonuclear Leukocytes Reduces Their Adhesion to P-Selectin and Causes Redistribution of Ligands for P-Selectin on Their Surfaces. Journal of Clinical Investigation, 1995. 96(1): p. 171. P. H. Reinhardt and P. Kubes, Differential Leukocyte Recruitment from Whole Blood Via EndothelialAdhesion Molecules under Shear Conditions. Blood, 1998. 92(12): p. 4691. N. Charles, J. L. Liesveld, and M. R. King, Investigating the Feasibilityof Stem Cell Enrichment Mediated by Immobilized Selectins. Biotechnology Progress, 2007. 23(6): p. 1463. S. D. Narasipura, J. C. Wojciechowski, N. Charles, J. L. Liesveld, and M. R. King, P-Selectin-Coated Microtube for Enrichment of Cd34(+) Hematopoietic Stem and Progenitor Cells from Human Bone Marrow Hematology. Clinical Chemistry, 2008. 54(1): p. 77. K. E. Norman, K. L. Moore, R. P. Mcever, and K. Ley, Leukocyte Rolling in-Vivo Is Mediated by P-Selectin Glycoprotein Ligand-1. Blood, 1995. 86(12): p. 4417. M. B. Lawrence, G. S. Kansas, E. J. Kunkel, and K. Ley, Threshold Levels of Fluid Shear Promote Leukocyte Adhesion through Selectins (Cd62l,PE). Journal of Cell Biology, 1997. 136(3): p. 717. C. Dong and X. X. Lei, Biomechanics of Cell Rolling: Shear Flow, Cell-Surface Adhesion, and Cell Deformability.Journal of Biomechanics, 2000. 33(1): p. 35. L. Wu, B. T. Xiao, X. L. Jia, Y. Zhang, S. Q. Lu, J. Chen, and M. Long, Impact of CarrierStiffness and Microtopology on Two-Dimensional Kinetics of P-Selectin and P-Selectin Glycoprotein Ligand-1 (Psgl-1) Interactions.Journal of Biological Chemistry, 2007. 282(13): p. 9846. W. C. Chang, L. P. Lee, and D. Liepmann, Biomimetic Technique for AdhesionBased Collection and Separation of Cells in a Microfluidic Channel. Lab on a Chip, 2005. 5(1): p. 64. C. D. James, R. C. Davis, L. Kam, H. G. Craighead, M. Isaacson, J. N. Turner, and W. Shain, Patterned Protein Layers on Solid Substrates by Thin Stamp MicrocontactPrinting.Langmuir, 1998. 14(4): p. 741. A. Bernard, E. Delamarche, H. Schmid, B. Michel, H. R. Bosshard, and H. Biebuyck, PrintingPatternsofProteins.Langmuir, 1998. 14(9): p. 2225. J. L. Tan, J. Tien, and C. S. Chen, MicrocontactPrintingof Proteins on Mixed Self-Assembled Monolayers. Langmuir, 2002. 18(2): p. 519. D. J. Graber, T. J. Zieziulewicz, D. A. Lawrence, W. Shain, and J. N. Turner, Antigen Binding Specificity of Antibodies Patterned by Microcontact Printing. Langmuir, 2003. 19(13): p. 5431. J. 0. Foley, E. Fu, L. J. Gamble, and P. Yager, MicrocontactPrintedAntibodies on Gold Surfaces: Function, Uniformity, and Silicone Contamination. Langmuir, 2008. 24(7): p. 3628. M. Ghosh, C. Alves, Z. Tong, K. Tettey, K. Konstantopoulos, and K. J. Stebe, Multifunctional Surfaces with Discrete Functionalized Regions for Biological Applications. Langmuir, 2008. 24(15): p. 8134. 93 [30] [31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] [43] [44] D. Lee and M. R. King, MicrocontactPrintingof P-Selectin Increases the Rate of Neutrophil Recruitment under Shear Flow. Biotechnology Progress, 2008. 24(5): p. 1052. A. Folch and M. Toner, Microengineering of Cellular Interactions. Annual Review of Biomedical Engineering, 2000. 2: p. 227. K. Glasmastar, J. Gold, A. S. Andersson, D. S. Sutherland, and B. Kasemo, Silicone Transfer During Microcontact Printing. Langmuir, 2003. 19(13): p. 5475. I. Banerjee, R. C. Pangule, and R. S. Kane, Antifouling Coatings: Recent Developments in the Design of Surfaces That Prevent Fouling by Proteins, Bacteria, and Marine Organisms. Adv Mater, 2011. 23(6): p. 690. M. Salta, J. A. Wharton, P. Stoodley, S. P. Dennington, L. R. Goodes, S. Werwinski, U. Mart, R. J. Wood, and K. R. Stokes, Designing Biomimetic Antifouling Surfaces. Philos Trans A Math Phys Eng Sci, 2010. 368(1929): p. 4729. W. R. Gombotz, G. H. Wang, T. A. Horbett, and A. S. Hoffman, Protein Adsorption to Poly(Ethylene Oxide) Surfaces. J Biomed Mater Res, 1991. 25(12): p. 1547. K. L. Prime and G. M. Whitesides, Adsorption of Proteins onto Surfaces Containing End-Attached Oligo(Ethylene Oxide): A Model System Using SelfAssembled Monolayers. J. Am. Chem. Soc., 1993. 115(23): p. 10714. K. L. Prime and G. M. Whitesides, Self-Assembled Organic Monolayers: Model Systems for Studying Adsorption of Proteins at Surfaces. Science, 1991. 252(5009): p. 1164. http://physics.georgetown.edu/matlab/. R. Karnik, S. Hong, H. Zhang, Y. Mei, D. G. Anderson, J. M. Karp, and R. Langer, Nanomechanical Control of Cell Rolling in Two Dimensions through Surface PatterningofReceptors. Nano Letters, 2008. 8(4): p. 1153. T. Yago, A. Leppanen, H. Y. Qiu, W. D. Marcus, M. U. Nollert, C. Zhu, R. D. Cummings, and R. P. McEver, Distinct Molecular and Cellular Contributions to Stabilizing Selectin-MediatedRolling under Flow. Journal of Cell Biology, 2002. 158(4): p. 787. S. Q. Chen, R. Alon, R. C. Fuhlbrigge, and T. A. Springer, Rolling and Transient Tethering of Leukocytes on Antibodies Reveal Specializations of Selectins. Proceedings of the National Academy of Sciences of the United States of America, 1997. 94(7): p. 3172. M. B. Lawrence and T. A. Springer, Neutrophils Roll on E-Selectin. J Immunol, 1993. 151(11): p. 6338. M. J. Smith, E. L. Berg, and M. B. Lawrence, A Direct Comparison of SelectinMediated Transient, Adhesive Events Using High Temporal Resolution. Biophysical Journal, 1999. 77(6): p. 3371. K. C. Chang and D. A. Hammer, Adhesive Dynamics Simulations of SialylLewis(X)/E-Selectin-Mediated Rolling in a Cell-FreeSystem. Biophysical Journal, 2000. 79(4): p. 1891. 94 [45] [46] [47] [48] [49] [50] [51] [52] [53] S. Jadhav, C. D. Eggleton, and K. Konstantopoulos, A 3-D ComputationalModel Predicts That Cell Deformation Affects Selectin-Mediated Leukocyte Rolling. Biophysical Journal, 2005. 88(1): p. 96. C. B. Korn and U. S. Schwarz, Dynamic States of Cells Adhering in Shear Flow: From Slipping to Rolling. Physical Review E, 2008. 77(4). J. Y. Shao and R. M. Hochmuth, Micropipette Suctionfor MeasuringPiconewton Forces of Adhesion and Tether Formation from Neutrophil Membranes. Biophysical Journal, 1996. 71(5): p. 2892. S. Vyawahare, A. D. Griffiths, and C. A. Merten, Miniaturization and Parallelizationof Biological and ChemicalAssays in Microfluidic Devices. Chem Biol, 2010. 17(10): p. 1052. S. Baratchi, K. Khoshmanesh, C. Sacristan, D. Depoil, D. Wlodkowic, P. McIntyre, and A. Mitchell, Immunology on Chip: Promises and Opportunities. Biotechnol Adv, 2013. P. Sundd, E. Gutierrez, M. K. Pospieszalska, H. Zhang, A. Groisman, and K. Ley, Quantitative Dynamic Footprinting Microscopy Reveals Mechanisms of Neutrophil Rolling. Nat Meth, 2010. 7(10): p. 821. E. B. Finger, K. D. Puri, R. Alon, M. B. Lawrence, U. H. von Andrian, and T. A. Springer, Adhesion through L-Selectin Requires a Threshold Hydrodynamic Shear. Nature, 1996. 379(6562): p. 266. T. Yago, V. I. Zarnitsyna, A. G. Klopocki, R. P. McEver, and C. Zhu, Transport Governs Flow-Enhanced Cell Tethering through L-Selectin at Threshold Shear. Biophys J, 2007. 92(1): p. 330. R. Alon, S. Chen, R. Fuhlbrigge, K. D. Puri, and T. A. Springer, The Kinetics and Shear Threshold of Transient and Rolling Interactions of L-Selectin with Its Ligand on Leukocytes. Proc Natl Acad Sci U S A, 1998. 95(20): p. 11631. 95 This page is intentionally left blank 96 4 Identification of Adhesion Molecules with Weak Affinity via Phage Display from M13 pVIII Library 4.1 Introduction Chapter IV aims to discuss and demonstrate how to identify specific receptors with low affinity to the target molecules to broaden the application range for cell separation devices based on transient cell-receptor interaction. Selectin is one of the families of cell adhesion molecules (CAMs). Three known members (P-, E- and L-selectin) of the selectin family have similar calcium-dependent lectin-like domain, which recognizes certain carbohydrate groups [1-4]. For example, Pselectin expressed on endothelial cells binds to P-selectin Glycoprotein Ligand- 1 (PSGL1) highly expressed on the neutrophils during the leukocytes adhesion cascade [5, 6]. The nature of P-selectin's low affinity to PSGL-1 allows the leukocytes to be captured and roll on the blood vessel wall and enables the feasibility of label-free netrophil separation via cell rolling on P-selectin patterns in microfluidic devices [7-10]. However, there exists a need for applying this technology to different receptors other than selectin family. The antibody is commonly used for identifying an unique target, called antigen, and it has been reported to show high specificity and high affinity (nanomolar ~subnanomolar range) [11-13]. Antibody-based microfluidic devices were reported to show high-purity capture of cells [14, 15]. However, high affinity resulting in stable and strong binding of the cells makes it difficult retrieving captured cells within the device which hinders some application where the viable cells are required, and prevents easy enumeration of cells. Biopanning is an affinity selection technique used for identifying peptides that bind to a target. Selected peptides have been reported to show affinity to the target within micromolar range [16], which opens the possibility of using selected peptides as weak affinity receptors for cell separation based on cell rolling. One of the most commonly used libraries is the M13 phage display library. M13 is a well-known filamentous 97 bacteriophage or virus and it is harmless to mammalian cells [ 17]. The length of the M 13 phage is around 880 nm and the diameter is around 6-7 nm. M13 has five coat proteins (pIII, pVI, pVII, pVIII, and pIX). Figure 4-1 shows the schematic diagram of M13 bacteriophage and the summary of the residues and number of copies of each type of coat protein. Among the five coat proteins, pVIII is the major one, which has 2,700 copies per phage. The whole genome of M13 has been revealed and studied for years. Its coat protein groups can be independently altered by engineering the viral genome. Because of this advantage, the M13 phage can be easily engineered to serve as a powerful biological toolkit. M13 phage display library is one of the examples [18-23]. A phage display library can be constructed by displaying random peptides on the certain position of the proteins of the phage. For example, pVIII library is the phage pool where the random peptides were inserted on pVIII coat protein. The phage library is then exposed to the target to find out which peptide sequence has affinity to the target. pVll, piX P1 pVi,. pill -- 6-7 nm 880 nm Protein Residues # of Copies pVll 33 5 piX 32 -5 pVil 73 2,700 pill 427 3-5 pVI 112 ~5 Figure 4-1 Schematic diagram of M13 bacteriophage and the summary of coat protein. (adapted from Dr. Chung-Yi Chiang from Prof. Angela Belcher's lab) In Chapter IV, biopanning using pVIII library was studied to investigate whether it can be used to identify molecules with low affinity with CD4 chosen as the target. CD4 is a type of protein molecules expressed on the surface of the mature Th cells (helper T cells), named CD4+ T cells [24]. It is a receptor for Glycoprotein gpl20 of human immunodeficiency virus (HIV) [25]. Binding of gp120 to CD4 is the first step of viral 98 entry, which leads to the fusion of viral and cell membranes. As the virus infects the cells, the number of CD4+ T cells decreases resulting in weakening the immune system. CD4+ T cells count is also an indicator of the success or the failure of anti-retroviral therapy (ARV) [26]. In addition to application of diagnosis and immune monitoring, sorting and analysis of pure, viable CD4+ T cells can be applied in studies on proliferation, apoptosis and regulation of CD4+ T cells [27-29]. In this Chapter, pVIII library was applied to select phage with affinity to CD4 protein. The selected phage was chemically immobilized on a gold substrate and the capture efficiencies of the CD4 expressing cells (SUPTI) and non-CD4 expressing cells (K562) were quantified. The phage-engineered substrate was blocked with CD4 to demonstrate the specificity of the selected phage to CD4. 4.2 4.2.1 Biopanning of pVIII Library Phage display library of random peptides The phage display library is a pool of bacteriophage of millions of random peptide sequences with different functionalities [18-21, 23]. Synthetic oligonucleotides of fixed length and random condons are fused to genes VIII of M13 vectors. The fusions are expressed as random peptide on the capsid or shell proteins of M 13 to form phage display library. The libraries are then used to screen peptides for targeting the materials of interest. To select peptides for targeting the materials of interest, the phage library is exposed to the target. Phage with affinity to the target can bind to the target. Unbound phage is then washed off from the target and the bound phage is then eluted from the target surface by changing the pH value or temperature. The sequences of the peptide motifs on the eluted phage are then characterized. The eluted phage is amplified to serve as the sub-library for the next round of the panning process. The panning process is repeated until the consensus peptides are found. 99 4.2.2 Biopanning Procedure Biopanning with the M13 pVIII library was conducted in collaboration with Dr. Nimrod Heldman and Dr. Rana Gosh in Prof. Angela M. Belcher's lab at MIT (DMSE and BE). Materials. Recombinant human CD4 receptor protein was purchased from Protein Science Corp and diluted into 100 pg/mL in phosphate buffered saline (PBS, Mediatech Inc). The pVIII library used in this chapter was constructed by Dr. Paul Widboom (Belcher's group at MIT) and the diversity of this particular library is 106. Bovine serum albumin (BSA) was purchased from Rockland Immunochemicals, Inc. Tris-HC buffer (Mallinckrodt Chemicals) was titrated to 1 M, pH=9.1. Glycine-HCl (Sigma-Aldrich) was titrated to 0.2 M, pH=2.2. 96-well standard microplate were purchased from Falcon@. Methods. A schematic diagram of identifying peptide motif with affinity to CD4 by biopanning method is shown in Figure 4-2. 400 pL of the diluted CD4 protein solution was added into 4 wells (100 pL/well; 10 pg CD4/well) of 96-well standard microplate. The plate was incubated overnight at 4'C in the refrigerator. Next day, the plate was taken out to room temperature. The CD4 solution was poured off. The wells were cleaned by filling each well with PBS and then slapped face down onto the clean paper towel ten times. Blocking solution was prepared by dissolving 3wt% BSA in PBS. 1200 ptL of the diluted BSA solution was added into the CD4-coated wells (300 pL/well) to block nonspecific interaction and kept at room temperature for 3 hours. The BSA solution was poured off and the wells were cleaned again by filling each well with PBS and then slapped face down onto the clean paper towel 10 times. 400 pL phage solution containing 400 copies of each sequence was prepared and added to four wells (100 pL/well; 100 copies of each sequence /well). The microplate was then kept overnight in the 4'C refrigerator. Next day, the microplate was taken out to room temperature. The unbound phage solution was poured off. The wells were cleaned by filling each well with PBS and then slapping it face down onto the clean paper towel ten times before bound phage was eluted. After the washing steps, 100 pL of Glycine-HCl 100 (pH=2.2; 0.2 M) containing 1 mg/mL BSA were added to each well and incubated for 10 minutes to elute the bound phage. The elution process was then repeated once. All the eluted phage in Glycine-HCl (eluate) were added to 150 ptL Tris-HCl (1 M, pH=9.1). The eluate was titered and the sequences of the peptide motif on the phage was sequenced. The phage solution was then amplified and purified to serve as the sub-library. The sublibrary was titered again and the sequences of the peptide motif on the sub-library were sequenced. The sub-library was then used for the next round of the panning process. The panning process was repeated three times. For the second round and the third round of biopanning, the initial input concentration of phage is 1011 per well (4 wells) and sequential steps were the same as described in the first round of biopanning. pVIII Phage Library _Livrsity-16) Amp &P Ug tion cation P P miUnbound Phages DNA sequencing Figure 4-2 Schematic diagram for identifying peptide motifs of affinity to CD4 protein by biopanning using pVIII library. The pVIII library was exposed to the CD4coated well plate. Unbound phage was then washed off from the target and the bound phage was eluted from the surface by acid solution. The eluted phage was amplified to serve as the sub-library for the next round of the panning process. The peptide sequences of the eluted and amplified phage was identified by titering and DNA sequencing. The panning process was repeated until the consensus peptides were found. 101 4.2.3 4.2.3.1 Phage Titering Lysogeny Broth (LB) Medium Preparation 1 L Lysogeny broth (LB) medium were prepared by mixing 25 g Difco LB Broth (BD Biosciences) and 1 L water. The medium was then autoclaved and stored at room temperature for further use. 4.2.3.2 Tetracyline (Tet) Stock Solution Preparation Tetracycline (OmniPur) was dissolved in Methanol (Pharmco-AAPER) to a final concentration of 20 mg/mL. The Tet stock solution was then stored in dark at -20'C for further use. 4.2.3.3 IPTG/Xgal Stock Solution Preparation 1.25g isopropyl P-D-thiogalactoside (IPTG, Sigma-Aldrich) and ig 5-Bromo-4-chloro-3indolyl- P-D-galactoside (Xgal, Sigma-Aldrich) were mixed in 25 mL Dimethylformamide (Sigma-Aldrich) and stored at 4C in the dark for further use. 4.2.3.4 LB/IPTG/Xgal Plates Preparation 15 g agar (BD Biosciences) and 1 L LB medium were mixed, autoclaved, and then cooled to <70'C. 1 mL Tet stock solution and 1 mL pre-made IPXG/Xgal were add to agar/LB solution. The solution was then poured on Petri dishes (BD Falcon) (1 5mL per dish). The dishes were then served as LB/IPTG/Xgal plates after they were solidified at room temperature. The LB/IPTG/Xgal plates were sealed and stored at 4C in the dark for further use. 4.2.3.5 Agarose Top 5 g Bacto-Tryptone (BD Biosciences), 2.5 g yeast extract (BD Biosciences), 2.5 g NaCl (Sigma-Aldrich), 0.5 g MgCl 2 6H 2 0 (Alfa Aesar), 3.5 g Agarose (OmniPur) were 102 dissolved and mixed in 500 mL water. The solution was then autoclaved and stored at room temperature for further use. 4.2.3.6 Titration Procedure The phage was titered on LB/IPTF/Xgal plates. 5 mL LB medium and a single colony of ER2738 (New England Biolabs) or XLlBlue (Agilent Technologies) were incubated overnight at 37*C with shaking serving as overnight cell culture. 10-fold serial dilutions of the phage in LB medium were prepared. For unamplified panning eluates, the dilution range is 101-104. For amplified phage supernatants, the dilution range is 108-1014. Agarose Top were pre-melted at 50'C. LB/IPTG/Xgal plates were pre-warmed at 37.5'C. 10 gL of each dilution were added to 200 pL of overnight cell culture to infect cells and incubated at room temperature for 5 minutes. The infected cells were then added to 3 mL pre-warmed Agarose Top and all the solution were poured onto the LB XGal/IPTG plates. After the plates were cooled and the Agarose Top was solidified at room temperature, the plates were then incubated overnight at 37 'C. Next day, the blue plaques on the plates (as shown in Figure 4-3) were randomly picked for sequencing. 103 Figure 4-3 Titration of phage on LB/IPTG/Xgal plates (dilution range is from 10' to 104). The phage was titered on LB/IPTF/Xgal plates and the blue plaque was then randomly selected for sequencing. 4.2.4 4.2.4.1 Amplification of M13 bactriophage PEG/NaCl Stock Solution Preparation 200 g PEG8000 (Amreco) and 146 g NaCl (Sigma-Aldrich) were dissolved and mixed in 1L water. The stock solution was then autoclaved and stored at room temperature for further use. 4.2.4.2 Amplification between Rounds of Panning 400 pL overnight cell culture, 40 pL Tet stock solution, and 400 pL the phage eluates were added to 40 mL autoclaved LB media. The culture solution was then incubated at 37'C for 4.5 to 5 hours with shaking (MaxQ 4000 shaker, Thermo Scientific). The culture solution was transferred to a centrifuge tube and spun for 20 minutes at 3500 rpm (Beckman Coulter JA25.5) at 4 'C. The supernatant was then transferred to a fresh tube. 1/6 volume of PEG/NaCl stock solution were added to the tube with supernatant and kept at 4'C overnight to allow the phage to precipitate. Next day, PEG precipitate was spun down for 20 minutes at 10,000 rpm at 4'C. The supernatant was then discarded. The pellet was suspended in 1 mL PBS and then transferred to 1.5 mL microcentrifuge tube. The solution was then spun for 5 minutes at 3500 rpm in a table-top microcentrifuge (Beckman Coulter) to pellet residual cells. The supernatant was then transferred to fresh microcentrifuge tube and re-precipitated with 1/6 volume of PEG/NaCl at ice for 2 hours. PEG precipitate was then spun down for 20 minutes at 10,000 rpm. The supernatant was then discarded. The pellet was suspended in 100 piL PBS to serve as sub-library. The phage concentration was characterized by spectrophotometer (Nanodrop@ ND 1000). The relationship between phage concentration and absorption is given by the following equation derived by G. Smith [30]: 104 phages/ mLIL(A = number where (A2 , - A 320 )x 6.-10" (26 2 ,6 -A 32 )x 6.-10" 30)x10JA29-A30x-017- of nucleotides / phage 7200 bases! phage (Eq. 4-1) A2 69 : the absorbance at 269 nm which reflects the DNA content in the solution A 32 0 : the absorbance at 320 nm which corrects for the naturally high baseline value of the solution The amplified phage sub-library was then titered for sequencing as described above. 4.2.5 DNA extraction for sequence identification After each round of biopanning and amplification, DNA samples of phage was prepared for sequencing and the DNA sequences were then translated into peptide sequences to see if there are multiple occurrences of the same sequence, which indicates dominant binders. Plaque Amplification. A blue plaque on LB/IPTG/Xgal plates were picked randomly by sterile pipette tip and were then transferred to a tube containing diluted culture (mixture of 30 piL overnight cell culture, 3 piL Tet stock solution, and 3.5mL LB). The culture solution was then incubated in the 37'C for 4.5 to 5 hours with shaking. Multiple plaques were picked. DNA Extraction. The schematic diagram is shown in Figure 4-4. All the buffers and columns used for DNA extraction were purchased from Qiagen (QlAprep Spin Miniprep Kit) without further purification. The culture amplified solution was transferred to a centrifuge tube and spun for 20 minutes at 3500 rpm. The supernatant was discarded. The pelleted bacterial cells were resuspended in 250 piL Buffer P1 and then transferred to a microcentrifuge tube. 250 ptL Buffer P2 was added to the solution and the tube was gently inverted 5 times to mix. 350 tL Buffer N3 was then added to the solution and the tube was inverted gently 5 times. The solution was then spun 10 minutes at 13000 rpm in a table-top microcentrifuge (Beckman Coulter). The supernatants were then applied to the QUAprep spin column. The solution was then spun 1 minute at 13000 rpm and the flow- 105 through was discarded. 0.75 mL Buffer PE was added to QlAprep spin column and the solution was spun 1 minute at 13000rpm. The flow-through was discarded and the column was placed in the microcentrifuge and spun down for 1 minute to remove residual Buffer PE. QlAprep column was then placed in a clean 1.5 mL microcentrifuge tube and 50 pL Buffer EB (10 mM Tris-Cl, pH8.5) was added to the column to elute DNA. The solution was then spun 1 minute at 13000 rpm. The sample for sequencing was prepared by adding 8.8 pL eluted DNA to 3.2 pL 96 gIll sequencing primer (5'-. HOCCC TCA TAG TTA GCG TAA CG -3', 1 pmol/pl; New England BioLabs). The samples were then analyzed by polymerase chain reaction (PCR, LightCycler 480 Real-time PCR system, Roche Applied Science). The DNA sequences obtained from PCR characterization of the phage was then translated into peptide sequences. Suspend bacteria in P buffer Spin down bacteria Lyse bacteria using P2 Neutralize the sarnple using N3 buffer buffer Spin down, apply MLSFAAAYDTNMDSD PAKAAFNSLQASATEY supernatants Into QIA- IGYAWAMVVVIVGAT IGIKLFKKFTSKAS coun Translation into amino acids PCR sequencing Elute DNA by adding EB buffer Discard flow-though, wash with PE buffer, centrifuge, and discard flow-through Figure 4-4 Schematic diagram of DNA extraction for sequence identification. DNAs of the phage was extracted from the bacteria. The bacteria were lysed and the DNAs were isolated from the lysed bacteria and then eluted using QlAprep Spin Miniprep Kit. 106 4.2.6 Biopanning Results The number of phage per well of input and output solutions and the recovery for each round of screening are summarized in Table 4-1. The large recovery from the first round may come form the nonspecific binding among phage in the library. The enrichment was observed between the second and the third round. The peptide sequences of the phage screened from the the first round of panning, the first round of amplification, the second round of panning, the second round of amplification, and the the third round of panning are summarized in Table 4-2. Although some clones appears multiple times, they may be fast growers. For example, VNTTVSGE didn't appear in the first and the second round of biopanning but appear twice (out of ten samples) after the second amplification. To explore wider range of possibility of identifying specific peptide motifs, twelve clones were selected and were named as RK1, RK2, RK3.... and RK12. . Peptide properties of RK1-RK12 are shown in Table 4-3 (negatively charged amino acids in red; positively charged amino acids in blue; polar amino acid without charged in yellow; hydrophobic amino acid in gray; aromatic amino acids in green; Tyrosine in orange). RK3 and RK12 show similar charge distribution. RK4, RK7, RK8, and RK9 also have similar charge distribution. Each clone was then amplified as described above for cell capture experiment. Table 4-1 Input and output concentration of phage and the recovery of each round of screening. The concentration is defined as the number of the phage per well. For each round of screening, four wells coated with CD4 were used. Round of screening Input phage (#/well) Output phage (#/well) Recovery (Input/Output) 108 1.66x10-s 7.12x10-8 9.5x10-6 1 2 1011 1.66x1O3 7.12x10 3 3 1011 9.5x105 Table 4-2 CD4 protein binding sequences from biopanning of pVIII library. 107 Occurrence Sequence Round1 panning A A A D D D D D D D G G G Q S Y G N N P V W Y D G L D Y D K G L N Y N D N D G T G T Y V Q I N G L R S D S S N L S Y Y S P M L P D S D M N S G N I G N G G T L V D G I D D D T N T M M P S S E P S S T E A E P S Round1 amplification A D D D G V V Q S W W G N S D Y N S N S T T S G D F Q D S T P A T G L S S G G A I S L M T G D D R P P E E T T E A A D D E E V V V S Y S P S N N S Y D Y Y F R P S S T T T S Y S G Q N T M N T A S S G G G N D S D D G I S D S S M G V T D E E E M P A P E T T D G V V V T G N N S Y D S T S S S Q T N T P G V G S G I S S L M D G E P P T E T V S S S M G D T V S S S M G D T A A P Y D D N T D M S I N S P E Round 2 panning Round 2 amplification Round 3 panning Q 108 X2 X2 X2 X2 X3 X2 Table 4-3 Selected peptide sequences and their properties. (negatively charged amino acids in red; positively charged amino acids in blue; polar amino acid without charged in yellow; hydrophobic amino acid in gray; aromatic amino acids in green; Tyrosine in orange). RK 2 RK 3 RK 6 S RK7S RK8 RK 8 RK RK RK 10 11 12 S S S P NT $ T T S P 4.3 Immobilization of phage on Au-coated glass slide Gold-coated glass slides were purchased from EMF Corp. All slides were cleaned with piranha solution prior to use (3:1 mixture of sulfuric acid (Sigma-Aldrich) and 30% hydrogen peroxide (Sigma-Aldrich)). A schematic diagram of the immobilization of phage on Au-coated glass slides is shown in Figure 4-5. Step 1: Dithiobis (succinimidyl propionate) (DSP, Sigma-Aldrich) was diluted in absolute ethanol (Pharmco-AAPER) to a concentration of 5 mM. The cleaned Au-coated slides were inbucated in 5 mM DPS for 3 hours at room temperature with shaking. The surface was then rinsed with ethanol and dried under a stream of N2. Step 2: Each clone was diluted in PBS to a concentration of 1013 /mL. The substrates were then incubated in phage solution (101 3 /mL in PBS, unless stated otherwise) using a perfusion chamber (Electron Microscopy Sciences) at 4'C overnight. The surfaces were then backfilled with BSA (3 wt% in PBS) for 3 h to block nonspecific interactions. 109 DSP d94 'T'- Au-coated glass slide Step 1 Step 2 Figure 4-5 Schematic diagram for immobilizing phage on a gold substrate. Step 1: Functionalization of DSP onto the gold surface. Step 2: Immobilization of phage on the reactive gold surface. 4.4 Substrate Characterization Atomic force microscopy (Veeco Dimension 3100, tapping mode, 1 Hz scan rate was used to characterize the surface. All substrates for AFM characterization were placed in a vacuum chamber overnight before imaging to minimize residual solvent on the surface. AFM height images are shown in Figure 4-6. Although all the samples were prepared at the incubation concentration of 1013 phage/mL, the surface density of the phage varies among the samples. For example, RK4, 6, 8 and 9 show bundle-like structures. It is possible that those clones have more hydrophobic motifs and tend to aggregate because of hydrophobic interaction. However, the surface density of RK6 is not as high as them although RK6 has similar charge distribution as RK4, RK8, and RK9. On the other hand, RK1 1 and RK12 show very low density on the gold substrates and those clones have more negatively charged motifs. A single phage may repel one another because of negative charges and may result in low surface density of the phage on the gold substrates. 110 III wuo WU S"7T wu 0 WU SIT wu 0 WU SIT wu 0 wu S wu 0 wu 0 wu ST wu S*L- wu 0 wu S-LwuS wu 0 WU ST 15 nm (j) RK10 0 nm 15 nm 0 nm 20 nm ) 0 nm 17.5 nm Onm Figure 4-6 Surface characterization of phage-engineered substrates. AFM height images of phage immobilized on the gold-coated substrate. All the images are 1 Om X 10 m. 4.5 4.5.1 Cell Capture Experiments Cell Culture SUPTI cells and K562 cells (both purchased from ATCC) were maintained in Iscove's modified Dulbecco's medium supplemented with 20% fetal bovine serum according to the ATCC recommendation. Cells were cultured in 75 cm2 polystyrene tissue culture flasks (BD Falcon@) with the cell density maintained between 105- 106 cells/mL. 4.5.2 Cell Staining All the cells were stained in sterile condition (inside the Bio-Hood) before the experiments. K562 cells were transferred from the flash into a 15mL conical tube and were spun at 1700 rpm for 5 min. The supernatant was discarded and the pellet was 112 suspended in PBS at a final concentration of 106 cell/mL. The Cell trackerM Green (CMFDA, 5-Chloromethylfluorescein Diacetate; Life Technologies) was then added to the solution to the final concentration of 1pM and incubated inside the water bath of 37.5'C for 20 min. The cells were spun down at 1800 rpm for 5 min and the pellet was then suspended in appropriate cell media to a final concentration of 106 cells/mL. The solution was then transferred to the wells of 6-well plate (Falcon@) (2 mL per well) and incubated inside the incubator for 1 h at 37.5'C with 5% CO 2. The solution was transferred to 15mL centrifuge tube and spun down at 1800 rpm for 5 min. The supernatant was discarded and the pellet was suspended in the media to the concentration of 106 cells/mL. The solution was then transferred into the well plate and kept in the incubator at 37.5'C with 5% CO 2 for further use. 4.5.3 Cell Capture Experiment in Flow Chamber An inverted fluorescent microscope (Nikon TE2000-U) with a mounted camera (Andor iXon 885) was used to record the interaction of cells and the phage-immobilized substrates using a 4x objective, typically at a rate of 1 frame/s with exposure time of 0.1-1 s. A suspension of cells was allowed to flow over the phage-coated surfaces in a rectangular flow chamber (Glycotech, Inc.; width w=1.0 cm, length l=6 cm, and height h=0.005 in.) at room temperature of 24.5'C. A syringe pump (World Precision Instruments (WPI), SP230IW) was used to generate different flow rates of between 15 and 75 iL/min, with corresponding shear stresses of 0.1-0.5 dyn/cm 2 (~ 0.01-0.05 Pa). The flow was laminar (Reynold's number Re ~ 0.1-3), and the shear stress T was calculated using the plane Poiseuille flow equation r=6puQ/wh 2 , where p is the kinematic viscosity, Q is the volumetric flow rate, w is the width of the flow chamber, and h is the height of the flow chamber. The cells were first flowed slowly (flow rate ~ 15 pL/min) into the flow chamber until they were shown on the window of the view. The flow was then stopped for 5 min to allow the cells to settle down and interact with the substrate. The flow was started again and parts of the cells were detached from the surface. 113 4.6 Cell Capture Efficiency 4.6.1 Phage-engineered Surface A suspension of the mixture of roughly equal amounts of non-stained SUPTI cells and stained K562 cells (~ 105 cells/mL) were flowed over the phage-engineered substrate. The fluorescent images of the cells inside the flow chamber before the flow started and after the flow started over the RK1-immobilized substrate are shown in Figure 4-7. The number of the settled cells before the flow started and the number of the cells attached on the substrate after the flow started for 60 s were counted. (a) Before starting the flow (b) After starting the flow 1~(=O.1 25 dyn/cm2) 20p Figure 4-7 Fluorescent microscope images of the cells on the RK1-engineered gold substrate (A) before the flow was started (B) after the flow was started for 60 s. The white dots are stained K562 cells and black dots are non-stained SUPTI cells. The capture efficiency (CE) was defined as: N CE - -R-xl00% where NR Ns: (Eq. 4-2) : Number of the cells remaining on the substrates after the flow started Number of the cells settling on the substrates before the flow started Figure 4-8 shows the results of capture efficiency (CE) of different clones (RK1-RK12) 114 at shear stress of 0.1 dyn/cm 2 . Since SUPTI is CD4 expressing cells and K562 is nonCD4 expressing cells, ideally, the SUPTI CE is higher than K562 CE. However, for some clones (RK2, RK3, RK5, RK7, RK8, RK10, RK11, and RK12), the K562 CE is higher than that of SUPTI CE. In some cases, RK 4, RK6, and RK9, for example, there is no significant difference between SUPTI CE and K562 CE although the average values of CE of SUPTI is higher than that of K562. Among these clones, RKl (DSYSTSMP) has most difference between the SUPTI and K562 capture efficiencies; however, the difference was not statistically significant (p=0.06). On the other hand, there is no strong evidence showing that surface density of phage relates to capture efficiency. 100 l JSUPT1 U K562 80 60 L 4 -J- 20 0 1 2 3 4 5 6 7 8 9 10 11 12 Clone Number Figure 4-8 Capture efficiencies of RK1-RK12-immobilized substrates at shear stress of 0.1 dyn/cm2. Error bars represent one standard deviation, where n = 4 replicate experiments for each condition. To study the effect of shear flow on the CE, RK1 were immobilized on the substrate and shear stresses of 0.1, 0.2, 0.3, 0.4, and 0.5 dyn/cm 2 were generated by syringe pump as described before. Figure 4-9(a) shows the SUPTI and K562 capture efficiencies at different shear stresses. For both SUPT1 and K562, CEs decrease with increasing shear stresses. However, difference between SUPTI and K562 CEs does not become more significant as the shear stress increases. 115 100 0 SUPT1 *EK562 .8 (U1 0 60 - ul a) p=0.06 40 p=0.30 20 0 0.1 r-i p=0.24 p=0.47 p=O.50 0.2 0.3 0.4 0.5 Shear Stress, T (dyn/cm 2) Figure 4-9 Capture efficiencies of RK1-immobilized substrates. The effect of different shear stress on the capture efficiencies of the cells on the RKl-immobilized substrates. Error bars represent one standard deviation, where n = 4 replicate experiments for each condition. 4.6.2 CD4-blocked Phage-engineered Surface To confirm that the interaction between the phage and the cells was specific to CD4. The CD4 proteins were used to block the interaction of phage to the cells. The phageimmobilized slides were incubated with CD4 proteins (5gg/mL) at room temperature for 3 h and then incorporated with the flow chamber. A suspension of SUPT1 cells (~ 105 cells/mL) was flowed over the CD4-blocked RK1-engineered substrate. The microscopic images of the cells on the substrate inside the flow chamber before the flow started and after the flow started over the RK1-immobilized substrate are shown in Figure 4-10. It was observed that the cell-surface interaction was reduced by blocking the surface with CD4, further confirming that the previously observed interaction was due to CD4. 116 (a) Before starting the flow Figure 4-10 Microscope images of the SUPT1 cells captured on the CD4-blocked RK1-engineered gold substrate (a) before the flow was started (b) after the flow was started for 60 sec. 4.7 Peptide Sequences Characterization of pVIII Library The sequence characterization of pVIII library was done after the three rounds of biopanning were completed to identify the possible cause for unsuccessful results. 96 samples were selected and sequenced (the procedure is the same as described before in section 4.2) and the results are shown in Table 4-4. Some unexpected biases have been found. For example, RK 7(VSSNGSET) appeared three times (out of 96 samples). This indicates that this library may not be a good screening pool. Table 4-4 Peptide sequences from biopanning of pVIII library. (96 samples were selected) 117 Sequence Sequence D N S M E G S G D D G D S D M T D M N M S S V E M p p A K p K E E K S D E p p p Y G N M Occurrence Occurrence Note Note X3 RK2 X2 RK10 P X2 D G p w A V K L A P V A S S N D I Y A S Y S D Y Y A S A S D M D S D L M P E S T M P S N T L S N M I P N D s N T S A X3 I I 118 RK5 X2 0 m V m Ci M -i x o x -I M) K () Z M) j 0 V (A) Z V) r- m z Zo 0 0 v)--j - (An (A -a -Z-I--Zn ZZ m - m C)nQi KM m )-0Z 0 0 -< V-C)( -- -0< (An Z 0m 0 MM0 4> Ln (A -n mn , -- 1 C > *-< m V > o o m 0 0 Z V -< -< -< m L .-u -< > *)< o mm m mm > C)C)C)C)C)C)C)C)C)C)C)C)nm M < < < << m(A-Ir-Im < C) A A(A- ZZA -V v Z < << << (A (A L < << -.) -V << -< 4.8 Conclusion In this chapter, the goal was to identify specific phage (or peptides) with affinity to CD4 and being able to capture CD4-expressing cells by phage-engineered substrate. 12 clones of phage was selected by biopanning using pVIII library as the screening pool and the CD4 protein as the target. Cell-capture experiments were performed on phage-engineered surfaces and the cells can be eluted from the surface by shear flow. 12 clones show different surface morphology after they were immobilized on the surface. Among 12 clones, RKl (DSYSTSMP) has most difference between the SUPTI and K562 capture efficiencies, although this was not statistically significant (p = 0.06). It was observed that the RK1 phage interaction is specific to CD4 expressed on the cells; however, the SUPTI capture efficiency of RK1 is only around 21% and this may not be able to provide sufficient capture efficiency for cell separation application. There are two possible reasons why the biopanning used in this chapter was not successful: (1) pVIII library used for biopanning has bias; (2) CD4 immobilized onto the well plate as the target for biopanning may not be oriented correctly and actively. It has been shown that the pVIII library used in this chapter is not a good screening pool because of the present of biased clones. A library containing a wide diversity of sequences without biases is needed for screening process to increase the possibility of selecting specific binders. Moreover, the biopanning target for this study, CD4 protein, was immobilized on the polystyrene surface. The immobilization of proteins on the polystyrene has been studied extensively and the interaction is mainly based on physical adsorption [31]. The CD4 may be adsorbed on the polystyrene well plate without specific orientation. Proteins may be denatured after they are physically adsorbed on the surface [31, 32]. To serve as the target for biopanning, CD4 should be immobilized onto the substrate with correct orientation. 4.9 [1] Chapter Reference R. P. McEver and C. Zhu, Rolling Cell Adhesion. Annu Rev Cell Dev Biol, 2010. 26: p. 363. 120 [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] S. M. Lucila and U. H. von Andrian, Immunological Adhesion and Homing Molecules, in Els200 1, John Wiley & Sons, Ltd. L. A. Lasky, Selectin-Carbohydrate Interactions and the Initiation of the Inflammatory Response. Annual Review of Biochemistry, 1995. 64: p. 113. R. P. McEver, Selectins: Lectins That Initiate Cell Adhesion under Flow. Curr Opin Cell Biol, 2002. 14(5): p. 581. K. L. Moore, K. D. Patel, R. E. Bruehl, F. Li, D. A. Johnson, H. S. Lichenstein, R. D. Cummings, D. F. Bainton, and R. P. McEver, P-Selectin Glycoprotein Ligand1 Mediates Rolling of Human Neutrophils on P-Selectin. J Cell Biol, 1995. 128(4): p. 661. G. A. Zimmerman, Two by Two: The Pairings of P-Selectin and P-Selectin Glycoprotein Ligand 1. Proc Natl Acad Sci U S A, 2001. 98(18): p. 10023. C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Examining the Lateral Displacement of H160 Cells Rolling on Asymmetric P-Selectin Patterns. Langmuir, 2011. 27(1): p. 240. C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Karnik, Studying Cell Rolling Trajectories on Asymmetric Receptor Patterns.J Vis Exp, 2011(48). C. Edington, H. Murata, R. Koepsel, J. Andersen, S. Eom, T. Kanade, A. C. Balazs, G. Kolmakov, C. Kline, D. McKeel, Z. Liron, and A. J. Russell, Tailoring the Trajectory of Cell Rolling with Cytotactic Surfaces. Langmuir, 2011. 27(24): p. 15345. S. Choi, J. M. Karp, and R. Karnik, Cell Sorting by Deterministic Cell Rolling. Lab Chip, 2012. 12(8): p. 1427. S. Lin, S.-Y. Lee, C.-C. Lin, and C.-K. Lee, Determinationof Binding Constant and Stoichiometry for Antibody-Antigen Interaction with Surface Plasmon Resonance. Current Proteomics, 2006. 3(12): p. 271. C. A. Borrebaeck, A. C. Malmborg, C. Furebring, A. Michaelsson, S. Ward, L. Danielsson, and M. Ohlin, Kinetic Analysis of Recombinant Antibody-Antigen Interactions: Relation between Structural Domains and Antigen Binding. Biotechnology (N Y), 1992. 10(6): p. 697. S. Hearty, P. Leonard, and R. O'Kennedy, MeasuringAntibody-Antigen Binding Kinetics Using Surface Plasmon Resonance. Methods Mol Biol, 2012. 907: p. 411. K. T. Kotz, W. Xiao, C. Miller-Graziano, W. J. Qian, A. Russom, E. A. Warner, L. L. Moldawer, A. De, P. E. Bankey, B. 0. Petritis, D. G. Camp, 2nd, A. E. Rosenbach, J. Goverman, S. P. Fagan, B. H. Brownstein, D. Irimia, W. Xu, J. Wilhelmy, M. N. Mindrinos, R. D. Smith, R. W. Davis, R. G. Tompkins, and M. Toner, Clinical Microfluidicsfor Neutrophil Genomics and Proteomics. Nat Med, 2010. 16(9): p. 1042. E. A. Warner, K. T. Kotz, R. F. Ungaro, A. S. Abouhamze, M. C. Lopez, A. G. Cuenca, K. M. Kelly-Scumpia, C. Moreno, K. A. O'Malley, J. D. Lanz, H. V. Baker, L. C. Martin, M. Toner, R. G. Tompkins, P. A. Efron, and L. L. Moldawer, Microfluidics-Based Capture of Human Neutrophils for Expression Analysis in Blood and BronchoalveolarLavage. Lab Invest, 2011. 91(12): p. 1787. B. K. Kay, J. Kasanov, and M. Yamabhai, Screening Phage-Displayed CombinatorialPeptideLibraries.Methods, 2001. 24(3): p. 240. 121 [17] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] M. A. Hemminga, W. L. Vos, P. V. Nazarov, R. B. Koehorst, C. J. Wolfs, R. B. Spruijt, and D. Stopar, Viruses: Incredible Nanomachines. New Advances with FilamentousPhages. Eur Biophys J, 2010. 39(4): p. 541. D. R. Wilson and B. B. Finlay, Phage Display: Applications, Innovations, and Issues in Phage and Host Biology. Can J Microbiol, 1998. 44(4): p. 313. D. J. Rodi and L. Makowski, Phage-DisplayTechnology--Findinga Needle in a Vast Molecular Haystack. Curr Opin Biotechnol, 1999. 10(1): p. 87. S. S. Sidhu, EngineeringM13for Phage Display. Biomol Eng, 2001. 18(2): p. 57. M. Szardenings, Phage Display of Random Peptide Libraries: Applications, Limits, and Potential.J Recept Signal Transduct Res, 2003. 23(4): p. 307. U. 0. Seker and H. V. Demir, Material Binding Peptidesfor Nanotechnology. Molecules, 2011. 16(2): p. 1426. U. Kriplani and B. K. Kay, Selecting Peptides for Use in Nanoscale Materials Using Phage-DisplayedCombinatorialPeptide Libraries. Curr Opin Biotechnol, 2005. 16(4): p. 470. R. V. Luckheeram, R. Zhou, A. D. Verma, and B. Xia, Cd4(+)T Cells: DifferentiationandFunctions. Clin Dev Immunol, 2012. 2012: p. 925135. M. W. Cloyd, J. J. Chen, P. Adeqboyega, and L. Wang, How Does Hiv Cause Depletion of Cd4 Lymphocytes? A Mechanism Involving Virus Signaling through Its Cellular Receptors. Curr Mol Med, 2001. 1(5): p. 545. L. N. Makoae, C. J. Portillo, L. R. Uys, P. S. Dlamini, M. Greeff, M. Chirwa, T. W. Kohi, J. Naidoo, J. Mullan, D. Wantland, K. Durrheim, and W. L. Holzemer, The Impact of Taking or Not Taking Arvs on Hiv Stigma as Reported by Persons Living with Hiv Infection in Five African Countries. AIDS Care, 2009. 21(11): p. 1357. M. Xydia, Y. Ge, U. Quitsch, and P. Beckhove, Cd4Ol Co-Stimulationfrom Cd8+ to Cd4+ Effector Memory T Cells Supports Cd4+ Expansion. Immunol Cell Biol, 2011. 89(6): p. 670. A. H. Patki, S. P. Zielske, S. F. Sieg, and M. M. Lederman, PreferentialS Phase Entry and Apoptosis of Cd4(+) T Lymphocytes of Hiv-1 -Infected Patients after in Vitro Cultivation. Clin Immunol, 2000. 97(3): p. 241. L. Pan, W. J. Weng, L. H. Xu, J. Wei, and J. P. Fang, Separation and Amplification of Cd4(+)Cd25(+) Regulatory T Cells from Sensitized Mice. Zhongguo Shi Yan Xue Ye Xue Za Zhi, 2012. 20(2): p. 500. http://www.biosci.missouri.edu/smithGp. J. E. Butler, L. Ni, R. Nessler, K. S. Joshi, M. Suter, B. Rosenberg, J. Chang, W. R. Brown, and L. A. Cantarero, The Physicaland FunctionalBehavior of Capture Antibodies Adsorbed on Polystyrene. J Immunol Methods, 1992. 150(1-2): p. 77. V. V. Hlady and J. Buijs, Protein Adsorption on Solid Surfaces. Curr Opin Biotechnol, 1996. 7(1): p. 72. 122 5 Identification of Adhesion Molecules with Weak Affinity via Phage Display from M13 pIII Library 5.1 Introduction In the previous chapter (Chapter IV) CD4 adhesion peptides selected from biopanning using M13 pVIII library were studied. The selected peptides were demonstrated to exhibit CD4-dependent adhesion. This chapter discusses study of the selected peptides obtained from bacteriophage-based biopanning, using a new protocol modified from the one studied in Chapter IV. This approach demonstrates the binding specificity of the selected peptides to the CD4 protein. Two main changes in the biopanning protocol discussed in this chapter are: (1) the screening pool, and (2) the target and the immobilization method used in the biopanning. Chapter IV established that the specific pVIII library used was not a good screening pool because of the presence of biased clones. In this chapter, a commercially available M13 pIll library is applied in selecting peptides with affinity to CD4 protein. This pIll library for M13 phage has been shown to contain a wide diversity of peptide sequences without population biases [1]. On the other hand, the CD4 protein used in Chapter IV as the biopanning target was immobilized on polystyrene well plates. The immobilization of proteins on the polystyrene has been studied extensively, and the interaction is mainly based on physical adsorption [2]. Thus, it is possible that the CD4 proteins may be adsorbed on the polystyrene well plate without specific orientation, as shown in Figure 51 (a). Proteins may also be denatured after they are physically adsorbed on the surface [2, 3]. To serve as the target for biopanning, CD4 should be immobilized onto the substrate with optimal orientation with respect to ligand binding. Nickel (1)-nitrilotriacetic acid (NTA) chelation has been widely used in isolation and purification of histidine-tagged proteins [4-9]. The coordination interaction between the metal ion and the histidine residue provides the possibility of appropriate orientation of the histidine-tagged protein on the metal ion coated substrates. On the other hand, CD4 has four extracellular domains (D1, D2, D3, and D4) where D1 is the outmost. Using peptides selected against 123 uppermost DI and D2 domains may increase capture efficiency of CD4+ cells. Moreover, DI and D2 domains play an important role in Envelope Glycoprotein GP120 (gp120) binding during Human Immunodeficiency Virus Type 1 (HIV-1) infection [10-12]. Indentifying a peptide sequence that binds specifically to CD4 presented by T cells may enable development of HIV monitoring via T cell counts [13, 14]. (a) CD4 (Dr-D4) _ Regular PS well plate (b) H 9H 9 R-N -C-N-CH-C-R 62 &2 (His-tagged D1 and D2) Ni (II) well plate NNNf)-NTA Figure 5-1 (a) CD4 protein physically adsorbed on regular polystyrene surface. (b) His-tagged CD4 protein immobilized on Ni (II)-NTA-coated surface with specific orientation. Nickel(II)-nitrilotriacetic acid (NTA) chelation has been widely used in isolation and purification of proteins tagged with oligo-histidine (His) [4-9]. Figure 5-1(b, left) shows the chelation between the histidine residues and the Ni(II)-NTA. The NTA can coordinate the Ni(II) ion with four valencies, and two remaining coordination sites are available for interaction with the imidazole ring from the histidine residues [7]. Knecht et al. have comparatively analyzed the binding properties of histidine(His)-tag with various lengths based on the surface plasmon resonance (SPR) measurements (see Table 5-1) [8]. The association constant (KD) values decrease from 62.7 pM for dihistidine to 0.014 pM for hexahistidine; however, a further increase in the length of the histidine does not improve the affinity: the KD values increase from 0.016 FM for heptahistidine to 0.07 pM for decahistidine [8]. Hence, hexahistidine has been empirically identified as the most 124 effective affinity tag and was used here. Table 5-1 Binding affinity (dissociation constant (KD) of oligo-histidine peptides to Ni(II). [81 Oligo-His Length Kd (pM) H-(His) 2-OH 62.7 ± 3.4 H-(His) 3-OH 2.23 ± 0.15 H-(His) 4-OH 0.313 ± 0.031 H-(His) 5-OH 0.024 ± 0.002 H-(His) 6-OH 0.014 ± 0.001 H-(His) 7-OH 0.016 ± 0.001 H-(His) 8 -OH 0.030 ± 0.002 H-(His)9 -OH 0.047 ± 0.002 H-(His)10-OH 0.070 ± 0.007 In this Chapter, hexa(His)-tagged CD4 proteins were immobilized onto the Ni(IL)-NTA coated substrates to serve as the target as shown in Figure 5-1(b) and M13 pIll library were used as the screening pool for biopanning. ELISA analysis indicates that the selected phage have specificity to CD4 proteins compared with wild type phage. The specificity of the corresponding synthetic peptide to the CD4 proteins was confirmed by staining the CD4-immobilized beads with dye-conjugated peptide. 5.2 Biopanning of pIII Library Biopanning with the M13 pIII library was conducted in collaboration with Dr. Nimrod Heldman and Dr. Rana Gosh in Prof. Angela M. Belcher's lab at MIT (DMSE and BE). 5.2.1 Biopanning Procedure Materials. Hexa(Histidine) tagged recombinant human CD4 molecules were purchased from Creative Biomart. and was diluted into 1 Opg/mL in phosphate buffered saline (PBS, Mediatech Inc). The phage screening pool was the PhD.-7TM pIII library (New England 125 Biolab) of the diversity ~10 9. Nickel(II) coated 96-well plates were purchased from Thermo Scientific. Other materials employed in this chapter can be referred to the text in Chapter IV unless otherwise specified. Methods. A schematic diagram of identifying peptide motif with affinity to CD4 by biopanning using M13 pIll library is shown in Figure 5-2. The methods (including biopanning procedure, phage titerring, phage amplification, and DNA extraction) were the same as described in Chapter IV (section 4.2), except: - CD4 is Histidine tagged; - 96-well plate is Ni(II) coated; - Screening pool is PhD.-7TM pIII library; - Four rounds of biopaning were completed; - For the first round of biopanning, the initial input concentration of phage was 100 copies per sequence in each well (1.28x 1011 phage per well and 4 wells in total). pill Phage Library (Diversity -109) CD4 protein late W Am g tion &Pcation UN DNA sequencing Bound P ages Unbound Phages Figure 5-2 Schematic diagram for identifying peptide motifs of affinity to CD4 by biopanning using pIII library. The pIll library was exposed to the CD4-coated well plate. Unbound phage was then washed off from the target and the bound phage was 126 eluted from the surface by acid solution. The eluted phage was amplified to serve as the sub-library for the next round of the panning process. The peptide sequences of the eluted and amplified phage was identified by titering and DNA sequencing. The panning process was repeated until the consensus peptides were found. 5.2.2 Biopanning Results The number of phage per well of input and output solutions and the recovery for each round of screening are summarized in Table 5-2. The enrichment was observed from the first round to the fourth round of biopanning for positive selection (i.e., adhesive sequences). The sequences screened from four panning rounds are listed in Table 5-3. After the fourth round, there are four peptide motifs with consensus sequences: (1) FYSHSAETV (2) KVWSLDH (3) KLWTLDW and (4) SPSTHWK. FYSHSAETV is wild type (WT) phage. Although SPSTHWK appeared multiple times (3 times out of 36 samples) in the fourth round of biopanning, it is identified as a fast grower: it appears once (out of 29 times) in the third round of biopanning and four times (out of 12 samples) in the third amplification. The possibility of occurrences increases greatly after amplification so there is a high possibility that SPSTHWK is a fast grower. Two remaining clones were then selected (KVWSLDH and KLWTLDW) and named as AMB1 and AMB2, respectively, and amplified for further study. Peptide properties of AMB 1 and AMB2 are shown in Table 5-4. It is interesting that the AMB 1 and AMB2 have similar charge distribution. The first six amino acids in the order are positively charged, hydrophobic, aromatic, polar, hydrophobic, and negatively charged for both phage. This indicates that they may have similar binding interaction to CD4 proteins. Table 5-2 Input and output concentration of phage and the recovery of each round of screening. The concentration is defined as the number of the phage per well. For each round of screening, four wells coated with CD4 were used. Round of screening 1 2 3 4 Input phage (#/well) 1.28x10" Output phage (#/well) 2.38x10 6 Recovery (Input/Output) 5.57x10-6 1011 2.85x106 2.85x1O-s 1011 1.02x10 6 1.02x10-5 4.75x10 6 4.75x10-5 1011 127 Table 5-3 CD4 protein binding sequences from biopanning of pIII library. Round 1 panning Sequence F L G H G S G S L R S H T L V G V L Y L Y S Ocurec Occurrence N H A L V Y E W N P Y H P R I H Q K I V K V D M N A L P L S K I S Round I amplification N G V R L Y L T G L F P K S T S K R A H G S K G N S D T G H R V P T R K L A L Round 2 panning L P X2 T T w N P Q N K L S X2 R H R L V L Round 2 amplification 128 X2 X2 Round 3 panning Round 3 amplification Sequence SeauenceOcunc H A A P L A M A D F V D L D I H E Y P F A F H K I H I E L L P L S L M R D N R E N H M N N w Q N H S S D I S F K S P C I S T V T N T T P V D L A N S x Y H K Y N L Occurence Y H L K R T S Q Y L S H w A T S T E T K T w T K K T S V E L P S s T T G T T T A E L A Q H R T Y T T V G Y H Y N R Q L M E N T S M M H G P L G V V R A V I I Q S T T x T R G L x w L K A T L w A w E E F N Q S Y Round 4 panning H w T S G S H T Y H Q P S T R w T F N 0 T Q A L L S D D L A A D F H K K K L N R R S S T T T T V W H w N F G S V 129 X2 T V X2 X4 T V X4 X7 X6 X3 Table 5-4 Selected peptide sequences and their properties. (negatively charged amino acids in red; positively charged amino acids in blue; polar amino acid without charged in yellow; hydrophobic amino acid in gray; aromatic amino acid in green; Histidine in orange). AMB 1 AMB 2 S VT L 5.3 Enzyme-linked Immunosorbent Assay (ELISA) The enzyme-linked immunosorbent assay (ELISA) is a common technique to detect the presence of antigen [15-17]. After the antigen is immobilized on the surface (usually a well plate), the detection antibody conjugated to an enzyme (or the antibody can be detected by a secondary antibody which is conjugated to an enzyme) is added, forming a complex with the antigen. Several washing steps are repeated between each ELISA step to remove unbound materials. Finally, the plate is developed by adding and enzymatic substrate to produce a colored end product, which provides visible signal. The signal correlates to the amount of antigen present in the well plate and can be qualitatively analyzed. ELISA was used here to test the above peptides for specific binding to CD4 proteins 5.3.1 5.3.1.1 Surface Density of CD4 on Well Plate as the Target of Biopanning Materials Anti-CD4 antibody (MEM- 115, a mouse monoclonal IgG 2a; Santa Cruz Biotechnology, Inc) was prepared at a 1:1000 dilution in PBS (with 2% BSA) and this antibody detects an epitope in the DI domain of CD4. Rabbit Anti-Mouse IgG 2 a - horseradish peroxidase (HRP) was phurased from Life technologies and was prepared at a 1:1000 dilution in PBS (with 2% BSA). Turbo 3,3',5,5'-Tetramethylbenzidine (TMB) was purchased from Thermo Scientific. IM sulfuric acid (H2 SO 4) was purchased from Sigma-Aldrich. 130 5.3.1.2 Method ELISA was used to characterize the surface density of CD4 protein on well plates, which were served as the target of biopanning in previous and this chapters. Preparation of the CD4-coated well plates was discussed in the sections of 4.2.2 and 5.2.1. Three wells of each immobilization method were prepared. 100 pL anti-CD4 IgG 2a dilutions were then added to into each well and then incubated for 1 h at room temperature. The antibody solution was poured off. The wells were then cleaned by filling each well with PBS and slapped face down onto the clean paper towel ten times. Next, 100 pL anti-Mouse IgG 2 a - HRP dilutions were then added to into each well and then incubated for 1 h at room temperature. The wells were then washed ten times. Next, 100 gL Turbo TMB was added to each well and then incubated for 30 min at room temperature. 100 pl 1 M H2SO 4 was then added to each well. TMB substrates yield a blue color when detecting HRP and the color changes to yellow with the addition of H2 SO 4 at the maximum absorbance wavelength of 450 nm. 5.3.1.3 ELISA Analysis The intensities of coloration at wavelength of 450 nm (yellow) were measured using microplate reader (Tecan M1000) and the results were compared with the intensity characterized from the enzyme substrate reaction with different antibody-HRP concentrations. The surface density for the CD4 (with Dl-D4 domains) physically adsorbed on the polystyrene well plate used in previous chapter (Chapter IV) was calculated to be around 4.7-9.3x 109 /cm 2 . The surface density for the target used in this chapter was about 3.4-6.8x 10'2 /cm 2 . The results suggests that the coordination interaction between the Ni(II) and the histidine provided appropriate orientation of histidine-tagged CD4 protein on the Ni(II) substrates and greatly enhanced the binding density of the CD4 protein on substrate by about three orders of magnitude. 131 5.3.2 Specificity of the Selected Phage to CD4 Protein 5.3.2.1 Materials Bovine serum albumin (BSA, Rockland Immunochemicals, Inc.) solution was prepared by BSA in PBS to a final concentration of 3 wt%. AMBI, AMB2 and Wild type (WT) phage was amplified (procedure was the same as what has been described in section 4.2) for this study. HRP-conjugated anti-M13 monoclonal antibody (GE Healthcare) was prepared at a 1:10000 dilution in PBS (with 2% BSA); this antibody recognizes and epitope on pVIII protein of M13 phage. Other materials can be referred to the text in sections of 5.2.1 and 5.3.1.1. 5.3.2.2 Methods ELISA was used to confirm the specific affinity of the screened phage to the CD4 protein molecules. Three independent experiments were performed. Quantitative data were presented as mean and standard deviation of the values obtained from each experiment. A schematic diagram of ELISA procedure is shown in Figure 5-3. The detailed procedure is described below: Immobilization of CD4 protein on Ni(II) well plate. The diluted CD4-His protein solution was added into Ni(II) well plate in the well number Al-Gi, A3-G3 and A5-G5 (100 pL/well; 1 pgCD4 /well). The plate was incubated overnight at 4'C in the refrigerator. Next day, the plate was taken out to room temperature. The CD4-His solution was poured off. The wells were cleaned by filling each well with PBS and then slapped face down onto the clean paper towel ten times. The BSA solution was then added into Al-Gl, A2-G2, A3-G3, A4-G4, A5-G5, and A6-G6 (300 pL/well) to block non-specific interaction and kept at room temperature for 3 hours. The BSA solution was poured off and the wells were cleaned again by filling each well with PBS and then slapped face down onto the clean paper towel 10 times. Exposure of the phage onto the CD4-coated well plate. Different dilutions (1013, 101, 101, 1010, 101, and 108/mL) of phage solutions were prepared and added to the coated 132 wells (100 pL/well). Column 1 and column 2 of the plate were incubated with AMB 1. Column 3 and Column 4 were incubated with AMB2. Column 5 and Column 6 were incubated with wild type (WT) phage. In Row A, B, C, D, E, F, G, the phage concentration is 1012, 1011 10,10 , 108, 10 7 , and 0 per well, respectively. The microplate was then kept overnight in the 4'C refrigerator. Next day, the microplate was taken out to the room temperature. The wells were cleaned by filling each well with BSA solution and slapped face down onto the clean paper towel 5 times. The plate was then cleaned by filling each well with PBS and slapped face down onto the clean paper towel 10 times. Detection of phage by anti-M13-HRP antibody. 100 tL Anti-M13-HRP dilutions were added to into each well (Al-Gi, A2-G2, A3-G3, A4-G4, A5-G5, and A6-G6) and then incubated for 1 h at room temperature. The antibody solution was poured off. The wells were then cleaned by filling each well with PBS and slapped face down onto the clean paper towel ten times. Enzyme-substrate reaction. 100 ptL Turbo TMB was added to each well (Al-G1, A2G2, A3-G3, A4-G4, A5-G5, and A6-G6) and then incubated for 30 minutes at room temperature. The TMB substrate yielded a blue color when HPR were detected. Next, 100 ptL 1 M sulfuric acid was then added to each well (Al-Gi, A2-G2, A3-G3, A4-G4, A5-G5, and A6-G6). The solution changed it color to yellow after sulfuric acid was added. 133 Sam NI(lI) surface CD4-HIs Phages Colored product HRP-conjugated o0 TMB substrate AntI-M13 00 0 sa TMB substrate Figure 5-3 Schematic diagram of ELISA procedure. CD4-His protein was immobilized on Ni (II) coated well plate. The phage was then exposed to the CD4-coated well plate. HRP-conjugated anti-M13 antibody binds specifically to the target (pVIII protein of the phage). Several washing steps were repeated between each ELISA step to remove unbound materials. Substrate (TMB) was added and the colored end product from enzyme-substrate reaction is produced. The color signal correlates to the amount of the phage present in the well plate. 5.3.2.3 ELISA Analysis ELISA analysis was used to identify the affinity of the selected phage (AMBl and AMB2) to CD4 proteins. The ELISA qualitative and quantitative results are shown in Figure 5-4(a) and Figure 5-4(b), respectively. Three 96-well plates were used to perform the experiment. For each well plate, column 1, 3, and 5 were coated with CD4 proteins then BSA, while column 2, 4, and 6 were coated with BSA only as control. Three clones 134 of phage (AMB 1, AMB2, and WT) were then added into the wells. AMB 1 phage was added to column 1 and 2. AMB2 phage was added to column 3, and 4. WT phage used as control was added to column 5 and 6. The ELISA qualitative results are shown in Figure 5-4(a). The phage was titered and the input concentration were labeled in the Figure 54(a). It was observed that the wells coated with CD4 protein are darker than the wells coated with only BSA and the color of the well becomes darker as the input concentration of the phage increases. The darker wells indicate higher concentration of anti-M13-HRP, which also means higher concentration of phage. To qualitatively compare the AMB1, AMB2 and WT phage, the intensities of coloration at wavelength of 450 nm (yellow) were measured using microplate reader (Tecan M1000); The ELISA quantitative results are shown in Figure 5-4(b). The detection limits for AMB1 and AMB2 are around 101-109 peptides per well. At the input concentration of 1012 per well, high absorbance may result from non-specific interaction between the phage and the coated molecules. It can be seen from the comparisons of the results of three types of phage at the input concentration around 1014-10" per well, the binding of AMBI and AMB2 to CD4 protein is more effective than that of WT phage. Moreover, it was demonstrated that AMB 1 and AMB2 are more specific to CD4 proteins than to the control molecules (BSA). These results also suggest that the biopanning process results in good affinity selection. 135 AMBi (a) Phage conc. [number/well] :CD4/ AMB2 ICD4/ IsA BSA BS s A I CD4/ I' BSA BSA IA A B 1010 C 109 D 103 E 10" F 0 G (b) -0- AMB1-CD4/BSA 00 ABM1-BSA AMB2-CD4/BSA + ABM2-BSA WT-CD4/BSA WT-BSA 1 0.8 C 0.6 .0 0.4 A. 0.2 0 6 7 g 9 12 13 Log [phage conc]/well Figure 5-4 ELISA assay output. (a) Qualitative results. Representative results are shown for only one experiment at each condition. Wells in column 1, 3, and 5 were coated with CD4 and then BSA. Wells in column 2, 4, and 6 were coated with only BSA as control. Phage with different input concentration were added. Phage AMBi were incubated in column 1 and 2. Phage AMB2 were incubated in column 3 and 4. WT phage was incubated in column 5 and 6; (b) Quantitative results. The relationship between the absorbance of the colored end product and the input concentration of the phage. Error bars represent one standard deviation, with n = 3 replicate experiments for each condition. 136 5.4 5.4.1 Specificity of the Synthetic Peptide to CD4 protein Synthetic peptide The custom peptide was synthesized by GenScript Corp. The sequence of the peptide is KLWTLDWGGGS-(CH 2 CH 2O) 6 -C (from N-terminus to C-terminus). KLWTLDW is the sequence of AMB2. The sequence of GGGS was designed to function as the spacer. The six repeat units of poly(ethylene oxide) were added to prevent further non-specific interaction between the peptide and other molecules. Cysteine containing of the thiol functional group at the C-terminus was designed for further use in functionalization. 5.4.2 Conjugation of Peptide to the Alexa Fluor 488 Dye Alexa Fluor@ 488 (AF 488) C5 Maleimide (C 3oH 2 5N4 NaOi 2 S2 ) is thiol-reactive and was purchased from Life Technologies. Figure 5-5 shows the reaction scheme of the dyemaleimide with the thiol-peptide. The peptide stock solution was made by dissolving the peptide in PBS to a concentration of 100 pM. AF 488 C5 Maleimide was added to the peptide stock solution to a concentration of 1 mM. The reaction was then allowed to proceed for 2 h at room temperature. This AF 488 dye-conjugated peptide solution may contain free AF 488 dye, non-conjugated peptide and AF 488 dye-conjugated peptide and needs further purification. o Alexa Fluor' F0 Figure 5-5 Reaction of the Alexa Fluor@ 488 C5 Maleimide with a thiol-peptide. 137 5.4.3 Purification of Dye-conjugated Peptides A polyacrylamide desalting column (gravity-flow column; Thermo Scientific) was used for recovering the molecules of the molecular weight larger than 1800 Da and the small compounds can be removed fully. The molecular weight of the peptide is 1684 and the molecular weight of the dye-conjugated peptide is 2405 and thus this column can be used for separating the dye-conjugated peptide form the solution. Some PBS buffer was first added into the column to equilibrate the column. The AF 488 dye-conjugated peptide solution (-5mL) was then added to the column. Several collection tubes were used to collect the drained samples (see Figure 5-6). Figure 5-6 Collected drained samples. The first five fractions were collected before the dye-conjugated peptide solution drained through and thus contained mostly buffer (colorless). The orange color came from dye-conjugated peptide. The non-conjugated dye shows yellow color. Fraction #8 was selected as the stock solution for further study. The concentration of this AF 488 dye-conjugated peptide was characterized by spectrophotometer (Nanodrop@ ND1000). The concentration was determined to be 250 gM by Eq. 5-1 (Beer-Lambert Law). Awhere A: C: S: C: -c-l the absorbance at 488 nm extinction coefficient (71000 M-1 cm~) the concentration of the dye-conjugated peptide path length 138 (Eq. 5-1) 5.4.4 Staining the CD4-functionalized Beads with the Dye-conjugated Peptides Materials. Ni(II)-NTA agarose beads (average bead size ~ 34 pm) were purchased from QLAgen. Alexa Fluor 647@ conjugated bovine albumin serum (AF 647 dye-conjugated BSA) were purchased from Life Technologies and diluted to 5 mg/mL in PBS. Blocking solution was prepared by dissolving BSA in PBS to a final concentration of 5 wt%. Incubating BSA-coated Ni(II) Beads with Dye-conjugated Peptides (as Control) Ni(II) beads were spun down and the supernatant was discarded. The pellet was suspended in suspended in blocking solution (BSA, 5 mg/mL in PBS) and incubated at room temperature for 3 h. Several washing steps were then repeated to remove free BSA. The beads were then incubated with 10 piM dye-conjugated peptide at room temperature for 1 h. Several washing steps were then repeated to remove unbound peptides. Functionalization of Ni(II) Beads with CD4 Protein Ni(II) beads were transferred into 1 mL microtube and were spun down for 15-30 s. The supernatant was discarded and the pellet was suspended in CD4-His solution (100 pg/mL in PBS) at a final concentration of 106 beads/mL. The solution was then incubated at 4'C overnight. Several washing steps were then repeated to remove unbound CD4-His molecules. The beads were then incubated in blocking solution at room temperature for 3 h. Functionalization of Ni(II) Beads with Dye-conjugated BSA Protein (as Control) Ni(II) beads were spun down and the supernatant was discarded. The pellet was suspended in AF 647 dye-conjugated BSA (5 mg/mL in PBS) at a final concentration of 106 beads/mL. The solution was then incubated at room temperature for 10 min. The beads were then spun down again and the pellet was then suspended in blocking solution and incubated at room temperature for 3 h. Staining Functionalized Beads with Dye-conjugated Peptides 139 The specificity of the synthetic peptides to the CD4 protein was confirmed by staining the functionalized beads with dye-conjugated peptides. The schematic diagram is shown in Figure 5-7. Two types of functionalized beads (CD4-coated beads and AF 647-BSAcoated beads) were mixed and incubated with BSA for 3 h at room temperature to block non-specific interaction. Several washing steps were applied and the mixed beads were then incubated with 10 pM AF 488-conjugated peptides at room temperature for 1 h. Several washing steps were then repeated to remove unbound peptides. AF 488-conjugated peptide KLWTLDWGGGS-(CH 2CH 20)sW CD4-HIs --- BSA 00 BSA-AF 647 Figure 5-7 Schematic diagram for staining the CD4-coated beads with dyeconjugated peptide. The Ni(II) beads were incubated with CD4-His and AF 647 conjugated BSA, separately. The beads were then mixed and incubated with BSA to block non-specific interaction. The mixed CD4 and AF 647-BSA-coated beads were then incubated with AF 488-conjugated peptide. Several washing steps were repeated between each step to remove unbound materials before the samples were imaged. 5.4.5 Characterization of Binding Specificity between Peptide and the Functionalized Beads An inverted fluorescence microscope (Nikon TE2000-U) with a mounted camera (Andor iXon 885) was used to examine autofluorescence of CD4-immobilized beads, and binding specificity of between peptides and the functionalized beads. A 40x objective was used. Different wavelengths were applied to image the mixture of the beads (see Figure 5-8). To identify if CD4 protein shows autofluorescence, CD4-immobilized beads were first imaged under bright field (Figure 5-8 (a)) and FITC channel (Figure 5-8 (b)). No autofluorescence was observed from FITC channel. Moreover, to characterize if 140 selected peptides bind to BSA, BSA-coated beads stained with AF 488 dye-conjugated peptides were imaged under bright field (Figure 5-8 (c)) and FITC channel (Figure 5-8 (d)). No fluorescence was observed from FITC channel, which suggests that there was no binding between BSA-coated beads and peptides. Finally, to confirm the specificity of selected peptide to the CD4 protein, mixed beads (beads coated with CD4 protein and beads coated with AF 647 dye-conjugated BSA) were stained with AF 488 dyeconjugated peptides and then imaged (Figure 5-8(e) ~ Figure 5-9(h)). Eleven such beads were first imaged under bright field (Figure 5-8(e)) and the Cy5 channel (Figure 5-8(f)). By comparing images of Figure 5-8(e) and Figure 5-8(f), beads number 1, 2, 6, and 11 were identified as the ones coated with AF 647 dye-conjugated BSA, which also suggests that number 3, 4, 5, 7, 8, 9, and 10 were coated with CD4 proteins. The mixed beads were then imaged under FITC channel Figure 5-9(g) and it shows the AF 488 dye-conjugated peptides bound to the CD4 coated beads (number 3, 4, 5, 7, 8, 9, and 10). Figure 5-9(h) is the Cy5/FITC channel merged image. These images indicated that selected peptide bound to only CD4-immobilized beads, which confirm the specificity of screened peptides to the CD4 protein. 141 (a) (C) (d) (e) (f) (h) (g) 100 Figure 5-8 Fluorescence microscope analysis. (a-b) CD4-immobilized beads. Images of (a) bright field and (b) FITC channel. CD4-immobilized beads do not show autofluorescence. (c-d) BSA-coated beads that were incubated with AF 488 dyeconjugated peptides. Images of (c) bright field and (d) FITC channel. AF 488 dyeconjugated peptides do not show non-specific binding to BSA-coated beads. (e-f) The mixture of the CD4-immobilized beads and the AF 647 dye-BSA-coated beads stained with AF 488 dye-conjugated peptides. Images of (e) bright field; (f) Cy5 channel; and (g) FITC channel; (h) is the Cy5/FITC channel merged image. AF 488 dye shows bright, green fluorescence and AF 647 shows bright, red fluorescence. Beads number 1, 2, 6, and 11 were coated with BSA and beads number 3, 4, 5, 7, 8, 9, and 10 142 were immobilized with CD4 proteins. The dye-conjugated peptide bound only to CD4immobilized beads. 5.5 Conclusion Biopanning using the M13 pIII library as screening pool and His-tagged CD4 on Ni(II) substrates as target were performed. The modified immobilization method resulted in an increase in the surface density of the target. Two consensus peptides were found, and selected phage and corresponding synthetic peptide were demonstrated to show specific binding to CD4 proteins. This initial identification of peptides specific to CD4 proteins prompts further studies to demonstrate that CD4+ cells or CD4 protein-functionalized beads can roll or be captured on peptide-functionalized surfaces. 5.6 Reference [1] [2] [3] [4] [5] [6] [7] [8] New England Biolabs, Ph.D. - 7TM Phage Display Peptide Library Kit Instruction Manual. J. E. Butler, L. Ni, R. Nessler, K. S. Joshi, M. Suter, B. Rosenberg, J. Chang, W. R. Brown, and L. A. Cantarero, The Physicaland FunctionalBehavior of Capture Antibodies Adsorbed on Polystyrene. J Immunol Methods, 1992. 150(1-2): p. 77. V. V. Hlady and J. Buijs, Protein Adsorption on Solid Surfaces. Curr Opin Biotechnol, 1996. 7(1): p. 72. K. Terpe, Overview of Tag Protein Fusions: From Molecular and Biochemical Fundamentalsto Commercial Systems. Appl Microbiol Biotechnol, 2003. 60(5): p. 523. L. Nieba, S. E. Nieba-Axmann, A. Persson, M. Hamalainen, F. Edebratt, A. Hansson, J. Lidholm, K. Magnusson, A. F. Karlsson, and A. Pluckthun, Biacore Analysis of Histidine-TaggedProteins Using a Chelating Nta Sensor Chip. Anal Biochem, 1997. 252(2): p. 217. X. Y. Dong, X. D. Feng, and Y. Sun, His-Tagged ProteinPurificationby MetalChelate Affinity Extraction with Nickel-Chelate Reverse Micelles. Biotechnol Prog, 2010. 26(4): p. 1088. H. Block, B. Maertens, A. Spriestersbach, N. Brinker, J. Kubicek, R. Fabis, J. Labahn, and F. Schafer, Immobilized-Metal Affinity Chromatography (Imac): A Review. Methods Enzymol, 2009. 463: p. 439. S. Knecht, D. Ricklin, A. N. Eberle, and B. Ernst, Oligohis-Tags:Mechanisms of Binding to Ni2+-Nta Surfaces. J Mol Recognit, 2009. 22(4): p. 270. 143 [9] [10] [11] [12] [13] [14] [15] [16] [17] M. De, S. Rana, and V. M. Rotello, Nickel-Ion-Mediated Control of the Stoichiometry of His-Tagged Protein/Nanoparticle Interactions. Macromol Biosci, 2009. 9(2): p. 174. L. Li, X. Shi, Q. Lu, S. Zhang, X. Wang, X. Jiang, Y. Liu, G. Wang, W. Zhu, R. Lei, and H. Wu, Role of Human Cd4 Dld2 Domain in Hiv-1 Infection. Immunol Invest, 2013. 42(2): p. 106. L. J. Matthias, P. T. Yam, X. M. Jiang, N. Vandegraaff, P. Li, P. Poumbourios, N. Donoghue, and P. J. Hogg, Disulfide Exchange in Domain 2 of Cd4 Is Required for Entry of Hiv-1. Nat Immunol, 2002. 3(8): p. 727. D. Sharma, M. M. Balamurali, K. Chakraborty, S. Kumaran, S. Jeganathan, U. Rashid, P. Ingallinella, and R. Varadarajan, Protein Minimization of the Gp120 Binding Region ofHuman Cd4. Biochemistry, 2005. 44(49): p. 16192. X. H. Cheng, D. Irimia, M. Dixon, K. Sekine, U. Demirci, L. Zamir, R. G. Tompkins, W. Rodriguez, and M. Toner, A Microfluidic Device for Practical Label-Free Cd4+ T Cell Counting of Hiv-Infected Subjects. Lab on a Chip, 2007. 7(2): p. 170. X. Cheng, A. Gupta, C. Chen, R. G. Tompkins, W. Rodriguez, and M. Toner, Enhancing the Performance of a Point-of-CareCd4+ T-Cell Counting Microchip through Monocyte Depletionfor Hiv/Aids Diagnostics. Lab Chip, 2009. 9(10): p. 1357. A. Voller, A. Bartlett, and D. E. Bidwell, Enzyme Immunoassays with Special Reference to Elisa Techniques. J Clin Pathol, 1978. 31(6): p. 507. A. Voller, The Enzyme-Linked Immunosorbent Assay (Elisa) (Theory, Technique and Applications). Ric Clin Lab, 1978. 8(4): p. 289. R. M. Lequin, Enzyme Immunoassay (Eia)/Enzyme-Linked ImmunosorbentAssay (Elisa). Clin Chem, 2005. 51(12): p. 2415. 144 6 Conclusion and Future Work Weak but specific interaction play an important role in many biological and physiological processes such as bacterial adhesion [1], cell homing [2, 3], and immune surveillance [1, 4]. Selectin and their ligand interaction resulting in rolling motion of leukocytes, stem cells and cancer cells has been widely studied. Patterning selectin with multiple inclined bands inside microfluidic devices opens new possibilities for separating cells by diverting the rolling direction of cells[5]. Therefore, systematic study of how cells flow inside the microfluidic channel, how cells get captured onto the patterned substrates, how substrates parameters affect cell rolling trajectories or separation potential, and how to identify new receptor-ligand system is prerequisite for the design of the substrates involving transient interactions between the cells and the receptor-patterned substrates. In this thesis, HL60 cells and P-selectin were used as a model system to study development of adhesive substrates and biopanning was used as the strategy to identify new receptor-ligand system. The specific contributions of the thesis are: - Identified shear stress controlled the transport of cells inside microfluidic device and capture efficiency of cells by receptor-functionalized substrates from the free stream. - Developed empirical equations to predict capture efficiency by adhesive substrates. - Developed a standard protocol using microcontact printing to create biomimetic adhesive substrate. - First observation of hopping of cells on successive inclined patterns. - Identified edge inclination angle affected edge tracking length of rolling cells on a single receptor band significantly. - Developed a standard approach to predict device performance and provided guidelines for future device design - Identified anti-CD4 peptides, which potentially could enable cell rolling, using biopanning. 145 The biomimetic adhesive microfluidic substrates for a new method to isolate cells in a continuous manner based on transient interaction of specific ligands on cell surface with inclined receptor bands have been developed and this study has led to realization of microfluidic device done by Bose el al for sorting leukocytes directly from blood using P-selectins in continuous flow [6]. The fact that separation length is most sensitive to shear stress since shear stress limits settling distance and capture efficiency of cells by P-selectin from the free stream in this technique. This thesis work suggests to lower shear stress in order to enhance separation potential for P-selectin-patterned microfluidic substrates. Moreover, it has been demonstrated that shear stress has no significant effect on edge tracking length after cells were captured. However, these statements may not be true for other adhesion molecules. For example, for L-selectin-based microfluidic devices, lower shear stress may not result in increasing capture efficiency because of shear threshold phenomenon. Detail studies of cell attachment and substrate parameters are needed for developing microfluidic systems based on new adhesion molecules. On the other hand, hopping behavior over successive patterns was observed in both flow chamber and device [6, 7] and it greatly increases the overall total lateral displacement of deflected cells, and therefore enhances device separation efficiency. However, the mechanism why cells hopped after they detached from the edge was not clear. What parameters may affect hopping behavior has not been identified. Moreover, study of how to increase hopping probability would help enhance device performance. For example, will decreasing gap width significantly increase the hopping probability? If yes, what will be the minimum gap width which leads to maximum hopping probability without making cell cross over the patterns? Therefore, understanding and quantification of hopping behavior would be useful for development of device design. To characterize hopping behavior, parallel receptor bands aligned perpendicular to flow direction can be designed as shown in Figure 6-1. Receptor bands are patterned on the substrate with different gap widths. The cells flow over the pattern and the minimum gap width without making cell cross over the pattern can be identified by studying the interaction between cells and the 146 pattern. Studying instantaneous flowing velocity of cells after they detach from the receptor bands may be interesting, too. For example, how the instantaneous flowing velocity changes from the moment the cell detach until it captures by a downstream band. Such study may help clarify the mechanism of hopping behavior of cells. I 0 I I U- )ping pi I 0. Z CL 0 Q U_ G1 G3 G2 (G1 < G2 < G3) Figure 6-1 Experimental design for characterizing hopping behavior of cells. Cells flow over parallel receptor bands with different gap widths aligned perpendicular to flow direction. The minimum gap width without making cell cross over the patterns can be identified. Since hopping probability was observed to be higher than capture probability from the free stream, increasing capture probability from the free stream will enhance device performance. 3D vertical hydrodynamic flow-focusing device was developed to synthesize polymer particle [8]. Similar design may help force cells to interact with substrates and therefore increase capture efficiency. Settling distance may reduce by such design as well. Using only known biological molecules such as selectins [9, 10] that enable cell to roll limits the application of biomimetic adhesive substrates studied in this thesis. To explore potential application for current technique, design or evolution of synthetic adhesion molecules against arbitrary surface marker is needed. While the present work does not 147 directly address the feasibility of separating different types of cells that interact with synthetic adhesion molecules, it lays the groundwork for future studies in this direction. Reference: [1] [2] [3] [4] [5] [6] [7] [8] [9] [10] W. E. Thomas, V. Vogel, and E. Sokurenko, Biophysics of Catch Bonds. Annu Rev Biophys, 2008. 37: p. 399. P. J. Quesenberry and P. S. Becker, Stem Cell Homing: Rolling, Crawling, and Nesting. Proc Natl Acad Sci U S A, 1998. 95(26): p. 15155. T. Lapidot, A. Dar, and 0. Kollet, How Do Stem Cells Find Their Way Home? Blood, 2005. 106(6): p. 1901. R. P. McEver and C. Zhu, Rolling Cell Adhesion. Annu Rev Cell Dev Biol, 2010. 26: p. 363. R. Karnik, S. Hong, H. Zhang, Y. Mei, D. G. Anderson, J. M. Karp, and R. Langer, Nanomechanical Control of Cell Rolling in Two Dimensions through Surface PatterningofReceptors. Nano Lett, 2008. 8(4): p. 1153. S. Bose, R. Singh, C. Shen, M. Hanewich-Hollatz, C. H. Lee, D. M. Dorfman, J. M. Karp, and R. Karnik, Affinity Flow Fractionationof Cells Via Transient Interactionswith Asymmetric Molecular Patterns.Scientific Reports, 2013(3): p. 2329. C. H. Lee, S. Bose, K. J. Van Vliet, J. M. Karp, and R. Kamik, Examining the Lateral Displacement of H160 Cells Rolling on Asymmetric P-Selectin Patterns. Langmuir, 2011. 27(1): p. 240. M. Rhee, P. M. Valencia, M. I. Rodriguez, R. Langer, 0. C. Farokhzad, and R. Karnik, Synthesis of Size-Tunable Polymeric Nanoparticles Enabled by 3d Hydrodynamic Flow Focusing in Single-Layer Microchannels. Adv Mater, 2011. 23(12): p. H79. M. E. Taylor and K. Drickamer, Paradigmsfor Glycan-Binding Receptors in Cell Adhesion. Curr Opin Cell Biol, 2007. 19(5): p. 572. S. Hakomori, Le(X) and Related Structures as Adhesion Molecules. Histochem J, 1992. 24(11): p. 771. 148