The characterization of soluble t cell receptors specific for the parasite toxoplasma gondii

164 367 0
The characterization of soluble t cell receptors specific for the parasite toxoplasma gondii

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

THE CHARACTERIZATION OF SOLUBLE T CELL RECEPTORS SPECIFIC FOR THE PARASITE TOXOPLASMA GONDII TAN ZHEN WEI (B.Sc.(Hons.), NTU A THESIS SUBMITTED FOR THE DEGREE OF DOCTOR OF PHILOSOPHY NUS GRADUATE SCHOOL FOR INTEGRATIVE SCIENCES AND ENGINEERING NATIONAL UNIVERSITY OF SINGAPORE 2014 Declaration I hereby declare that this thesis is my original work and it has been written by me in its entirety I have duly acknowledged all the sources of information which have been used in the thesis This thesis has also not been submitted for any degree in any university previously Tan Zhen Wei 31 July 2014 I   Acknowledgements Being able to write this page would mean the end of a four and a half year journey, a journey which I will not be able to make alone Here, I would like to express my heartfelt thanks to these people First and foremost, I would like to thank my supervisor, Dr Gijsbert Grotenbreg This journey would not have started without him With his guidance, I could see myself grow over the years, learning the skill sets expected of a researcher He was a mentor but never appeared too far to approach and always willing to provide advice Next, I would also like to thank the members of the lab, past and present Special thanks to Ming Yan and Kai Yee, both of whom showed me the ropes when I first joined the lab To Ling yun, for offering help and support as a fellow post-grad To Jia Wei, for being in charge of peptide synthesis, taking care of the lab and making sure the lab is always in running order, not to forget being a very good gym buddy To Socks, for always willing to drive me to NTU for experiments, teaching me on SPR, as well as providing advice whenever needed To Kenneth and Lionel, for offering advice on experiments and football discussions To Joanna, Gladys, Ping Ping and all other members whom I did not list but have helped me in one way or the other in my journey Next, I would like to thank Dr Johnathan Rapley and Dr Rob Meijers (European Molecular Biology Laboratory, Hamburg Germany) for solving the crystal structure of the Rop7c1 TCR This allowed me to the supercharging part of my project I would also like to thank Dr Lee Kim Swee and Dr Hidde Ploegh (Whitehead Institute of Biomedical Research, Cambridge USA) for agreeing to share data on the effector function of the T cell clones, as well as II   the chance to collaborate on a publication currently under review I would also like to thank Dr David Thompson and Dr David Liu (Howard Hughes Medical Institute, Harvard University USA) for kindly sharing the AvNAPSA program for our supercharging experiments I would like to take this opportunity to thank Dr Markus Wenk and Dr Cynthia He as well, both of whom were ever so friendly and provided insightful advice during my TAC meetings with them I would like to thank my parents for their support during this time, for trusting in me in my decision to a Ph.D Last, I would like to thank my fiancée, Yi Zhen, for her support and her understanding that I was always late for our dates whenever I was in lab III   Table of Contents Summary……… ……………………………………………………… VII List of Tables……….………………………………… IX List of Figures………………………………………… X List of Illustrations………………………………………………… XII List of Abbreviations and symbols………………………………… XIII List of Publications…………………………………………………… XVII Chapter 1: Introduction .1 1.1 Adaptive immune system………………………………………………1 1.2 BCR recognition and diversity……………………………………… 1.3 BCR therapeutics: Monoclonal Antibodies………………………… 1.4 TCR recognition and diversity……………………………………… 14 1.5 TCR therapeutics: Adoptive T cell transfer…………………………18 1.6 TCR therapeutics: Soluble TCRs…………………………………….21 1.7 Supercharging to improve stabilities of soluble TCRs…………… 27 Chapter 2: Production and characterization of TCR clones with identical antigen specificities 2.1 Introduction…………………………………………………………… 31 2.2 Materials and Methods……………………………………………… 35 2.2.1 Cloning of TCR α and β chains…………………………………….35 2.2.2 Expression and purification of TCR α and β chains.…………….36 2.2.3 Refolding and purification of soluble TCRs……………………….36 IV   2.2.4 TCR-tetramer binding assay……………………………………….37 2.2.5 Surface Plasmon Resonance………………………………………38 2.3 Results………………………………………………………………….38 2.3.1 Cloning, expression and purification of individual TCR chains…38 2.3.2 Refolding and purification of the soluble TCR clones…………42 2.3.3 Binding specificities of refolded TCRs…………………………….48 2.3.4 Binding affinity between the three TCRs and Ld Rop7 MHC… 51 2.4 Discussion…………………………………………………………… 55 Chapter 3: Production and characterization of supercharged TCRs 3.1 Introduction………………………………………………………… 60 3.2 Materials and Methods……………………………………… 65 3.2.1 Design of supercharged Rop7c1 TCR chains………………… 65 3.2.2 Production and purification of supercharged TCR chains………66 3.2.3 Refolding of supercharged TCRs………………………………….66 3.2.4 Assessing functional avidities of supercharged TCRs………… 66 3.2.5 RAW cells staining with TCR tetramers………………………… 67 3.2.6 Stability assay of supercharged TCRs……………………………67 3.3 Results……………………………………………………………… 68 3.3.1 Crystal Structure of Rop7c1 TCR………………………………….68 3.3.2 Production of supercharged Rop7c1 TCR α and β chains…… 68 3.3.3 Production and purification of supercharged Rop7c1 TCRs……73 3.3.4 Functional avidity and specificity of supercharged TCRs……….84 3.3.5 Stabilities of supercharged Rop7c1 TCRs……………………… 88 3.4 Discussion…………………………………………………………… 98 V   Chapter 4: Comparing TCR binding affinity and effector function 4.1 Introduction……………………………………………………………105 4.2 Materials and Methods………………………………………………110 4.2.1 Surface Plasmon Resonance…………………………………….110 4.2.2 Structural modeling of Altered Peptide Ligands……………… 110 4.2.3 Stability assays for pMHCs……………………………………….110 4.3 Results……………………………………………………………… 111 4.3.1 Rop7c1, Rop7c2 and Rop7c3 recognize their cognate ligands differently…………………………………………………………….111 4.3.2 Stabilities of the Ld MHC presenting APLs…………………… 116 4.3.3 TCR binding affinity is not indicative of effector function………119 4.4 Discussion…………………………………………………………….122 Conclusion……………………………………………………………….130 Future Work………………………………………………………………131 Bibliography…………………………………………………………… 132 Annex A………………………………………………………………… 143 VI   Summary CD8+ T cells are important for resolving attacks by pathogens such as viruses and parasites Recently, transnuclear mice monoclonal for each of three TCRs recognizing the Rop7 antigenic peptide from the parasite Toxoplasma gondii were generated To better understand T cell immunity against the parasite, I characterized the binding affinities of these three T Cell Receptors (TCRs) against the Rop7 peptide MHC molecule I observed a range of binding affinities for these three TCRs Moreover, binding kinetic studies also revealed that they contact the peptide Major Histocompatibility Complex (pMHC) for different periods of time These data indicate that during an infection by the parasite, T cells expressing TCRs with a range of binding affinities are activated Thus, T cell activation is not solely dependent on the binding affinity of the TCR to its cognate ligand With the soluble TCRs generated from the binding affinity and kinetics studies, I sought to increase the stabilities of soluble TCRs so as to increase their attractiveness as a potential alternative immunoconjugate platform to monoclonal antibodies To this end, I selected the Rop7c1 TCR, which has the highest refolding yield as well as binding affinity, to try and improve its stability Increasing the surface charges of a protein have been shown to increase the thermostabilities of some proteins Thus, I increased the surface charges of the Rop7c1 TCR by selecting and mutating highly surface exposed residues to either positively or negatively charged residues However, increasing the surface charges did not improve the stability of the TCR VII   Several factors such as free energy and surface charge-charge interactions play a role in protein stability as well and thus, solely increasing the surface charges of the TCR may not be sufficient to improve its stability With our collaborator’s data, I was also able to compare the binding affinities of the three TCR clones with the effector function of the T cells expressing them I observed that the binding affinities of the TCR not correlate positively with the strength of their effector function This implies T cell effector function may not be dependent on TCR binding affinity but on other possible intrinsic factors More experiments will be required to identify these factors VIII   List of Tables Table 1: TCR gene usage .40 Table 2: AvNAPSA scores for Rop7c1 TCR 70 Table 3: Attempted refolds of supercharged TCRs………………………81 Table 4: Buffer conditions for stability assays……………………………90 IX   References 10 11 12 13 14 15 16 17 18 19 20 21 22 Cooper, M D & Alder, M N The evolution of adaptive immune systems Cell (2006) Starr, T K & Jameson, S C Positive and negative selection of T cells Annual review of … (2003) Davis, M M et al LIGAND RECOGNITION BY αβ T CELL RECEPTORS Annu Rev Immunol 16, 523–544 (1998) van der Merwe, P A & Davis, S J Molecular interactions mediating T cell antigen recognition Annu Rev Immunol 21, 659–684 (2003) Mempel, T R., Henrickson, S E & Andrian, von, U H T-cell priming by dendriticcells in lymph nodes occurs in three distinct phases Nature 427, 154–159 (2004) Neefjes, J., Jongsma, M L M., Paul, P & Bakke, O Towards a systems understanding of MHC class I and MHC class II antigen presentation Nat Rev Immunol 11, 823–836 (2011) Zhu, J & Paul, W E CD4 T cells: fates, functions, and faults Blood 112, 1557–1569 (2008) Weaver, C T., Harrington, L E., Mangan, P R., Gavrieli, M & Murphy, K M Th17: an effector CD4 T cell lineage with regulatory T cell ties Immunity 24, 677–688 (2006) Sakaguchi, S Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses Annu Rev Immunol 22, 531–562 (2004) Pattle, S B & Farrell, P J The role of Epstein-Barr virus in cancer Expert Opin Biol Ther 6, 1193–1205 (2006) Walboomers, J M et al Human papillomavirus is a necessary cause of invasive cervical cancer worldwide J Pathol 189, 12–19 (1999) Teicher, B A & Chari, R V J Antibody conjugate therapeutics: challenges and potential Clin Cancer Res 17, 6389–6397 (2011) Alley, S C., Okeley, N M & Senter, P D Antibody-drug conjugates: targeted drug delivery for cancer Curr Opin Chem Biol 14, 529–537 (2010) Schumacher, T N M T-cell-receptor gene therapy Nat Rev Immunol 2, 512–519 (2002) Köhler, G & Milstein, C Continuous cultures of fused cells secreting antibody of predefined specificity Nature (1975) Waldmann, T A Immunotherapy: past, present and future Nat Med 9, 269– 277 (2003) Jones, P T., Dear, P H., Foote, J., Neuberger, M S & Winter, G Replacing the complementarity-determining regions in a human antibody with those from a mouse Nature 321, 522–525 (1986) Queen, C et al A humanized antibody that binds to the interleukin receptor Proc Natl Acad Sci U.S.A 86, 10029–10033 (1989) Nelson, A & Dhimolea, E Development trends for human monoclonal antibody therapeutics Nat Rev Drug Discov (2010) Wu, A M & Senter, P D Arming antibodies: prospects and challenges for immunoconjugates Nat Biotechnol 23, 1137–1146 (2005) Casi, G & Neri, D Antibody-drug conjugates: Basic concepts, examples and future perspectives J Control Release (2012) doi:10.1016/j.jconrel.2012.01.026 Gura, T Therapeutic antibodies: magic bullets hit the target Nature 417, 584–586 (2002) 132   23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 Jain, R Tumor physiology and antibody delivery (Frontiers of radiation therapy and oncology, 1990) Roopenian, D C & Akilesh, S FcRn: the neonatal Fc receptor comes of age Nat Rev Immunol 7, 715–725 (2007) Chames, P., Van Regenmortel, M., Weiss, E & Baty, D Therapeutic antibodies: successes, limitations and hopes for the future Br J Pharmacol 157, 220–233 (2009) Adams, G P et al High affinity restricts the localization and tumor penetration of single-chain fv antibody molecules Cancer Res 61, 4750– 4755 (2001) Fujimori, K., Covell, D G., Fletcher, J E & Weinstein, J N A modeling analysis of monoclonal antibody percolation through tumors: a binding-site barrier J Nucl Med 31, 1191–1198 (1990) Laugel, B et al Design of soluble recombinant T cell receptors for antigen targeting and T cell inhibition J Biol Chem 280, 1882–1892 (2005) Wong, F Insulin-dependent diabetes mellitus and its animal models Current Opinion in Immunology (1999) Novellino, L., Castelli, C & Parmiani, G A listing of human tumor antigens recognized by T cells: March 2004 update Cancer Immunol Immunother 54, 187–207 (2005) Dahan, R & Reiter, Y T-cell-receptor-like antibodies - generation, function and applications Expert Rev Mol Med 14, e6 (2012) Robert, R et al Identification of human scFvs targeting atherosclerotic lesions: selection by single round in vivo phage display J Biol Chem 281, 40135–40143 (2006) Sullivan, D E et al Construction and characterization of an intracellular single-chain human antibody to hepatitis C virus non-structural protein J Hepatol 37, 660–668 (2002) Legastelois, I & Desgranges, C Design and intracellular activity of a human single-chain antibody to human immunodeficiency virus type conserved gp41 epitope J Virol 74, 5712–5715 (2000) Thomas, T C et al Inhibition of complement activity by humanized antiC5 antibody and single-chain Fv Mol Immunol 33, 1389–1401 (1996) Liu, S Y et al Follow-up of relapsed B-cell lymphoma patients treated with iodine-131-labeled anti-CD20 antibody and autologous stem-cell rescue J Clin Oncol 16, 3270–3278 (1998) Souriau, C & Hudson, P J Recombinant antibodies for cancer diagnosis and therapy Expert Opin Biol Ther (2001) Blažek, D & Celer, V The production and application of single-chain antibody fragments Folia microbiologica (2003) Segal, D M & Bast, B Production of bispecific antibodies Current protocols in immunology (1995) Chames, P & Baty, D Bispecific antibodies for cancer therapy: the light at the end of the tunnel? MAbs 1, 539–547 (2009) Kufer, P., Lutterbüse, R & Baeuerle, P A A revival of bispecific antibodies Trends Biotechnol 22, 238–244 (2004) Grosse-Hovest, L et al A recombinant bispecific single-chain antibody induces targeted, supra-agonistic CD28-stimulation and tumor cell killing Eur J Immunol 33, 1334–1340 (2003) Holliger, P., Prospero, T & Winter, G ‘Diabodies’: small bivalent and bispecific antibody fragments Proc Natl Acad Sci U.S.A 90, 6444–6448 (1993) Bühler, P et al A bispecific diabody directed against prostate-specific membrane antigen and CD3 induces T-cell mediated lysis of prostate cancer cells Cancer Immunol Immunother 57, 43–52 (2008) 133   45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 Galon, J., Costes, A., Sanchez-Cabo, F & Kirilovsky, A Type, density, and location of immune cells within human colorectal tumors predict clinical outcome Science (2006) Wahlin, B E., Sander, B., Christensson, B & Kimby, E CD8+ T-cell content in diagnostic lymph nodes measured by flow cytometry is a predictor of survival in follicular lymphoma Clin Cancer Res 13, 388–397 (2007) Begley, J & Ribas, A Targeted therapies to improve tumor immunotherapy Clinical Cancer Research (2008) Bargou, R et al Tumor regression in cancer patients by very low doses of a T cell-engaging antibody Science 321, 974–977 (2008) Koszinowski, U H., Reddehase, M J & Jonjic, S The role of CD4 and CD8 T cells in viral infections Current Opinion in Immunology 3, 471–475 (1991) Doherty, P C., Allan, W., Eichelberger, M & Carding, S R Roles of alphabeta and gammadelta T Cell Subsets in Viral Immunity Annu Rev Immunol 10, 123–151 (1992) Goedert, J The epidemiology of acquired immunodeficiency syndrome malignancies Seminars in oncology (2000) Shankaran, V., Ikeda, H., Bruce, A & White, J IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity Nature (2001) Mason, D A very high level of crossreactivity is an essential feature of the T-cell receptor Immunol Today 19, 395–404 (1998) Turner, S J., Doherty, P C., McCluskey, J & Rossjohn, J Structural determinants of T-cell receptor bias in immunity Nat Rev Immunol 6, 883–894 (2006) Altman, J D., Moss, P., Goulder, P & Barouch, D H Phenotypic analysis of antigen-specific T lymphocytes Science (1996) Toebes, M et al Design and use of conditional MHC class I ligands Nat Med 12, 246–251 (2006) Grotenbreg, G M et al Discovery of CD8+ T cell epitopes in Chlamydia trachomatis infection through use of caged class I MHC tetramers Proc Natl Acad Sci U.S.A 105, 3831–3836 (2008) Frickel, E.-M et al Parasite stage-specific recognition of endogenous Toxoplasma gondii-derived CD8+ T cell epitopes J Infect Dis 198, 1625– 1633 (2008) Chang, C X L et al Conditional ligands for Asian HLA variants facilitate the definition of CD8+ T-cell responses in acute and chronic viral diseases Eur J Immunol 43, 1109–1120 (2013) Newell, E W et al Combinatorial tetramer staining and mass cytometry analysis facilitate T-cell epitope mapping and characterization Nat Biotechnol 31, 623–629 (2013) Kirak, O., Frickel, E., Grotenbreg, G & Suh, H Transnuclear mice with predefined T cell receptor specificities against Toxoplasma gondii obtained via SCNT Science (2010) Kvistborg, P et al TIL therapy broadens the tumor-reactive CD8(+) T cell compartment in melanoma patients Oncoimmunology 1, 409–418 (2012) Hershkovitz, L., Schachter, J & Treves, A Focus on adoptive T cell transfer trials in melanoma Clinical and … (2010) Nauerth, M et al TCR-Ligand koff Rate Correlates with the Protective Capacity of Antigen-Specific CD8+ T Cells for Adoptive Transfer Sci Transl Med 5, 192ra87 (2013) Barrett, D M., Singh, N & Porter, D L Chimeric Antigen Receptor Therapy for Cancer Annual review of … (2014) 134   66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 Krause, A et al Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes J Exp Med 188, 619–626 (1998) Cheng, L E., Ohlén, C., Nelson, B H & Greenberg, P D Enhanced signaling through the IL-2 receptor in CD8+ T cells regulated by antigen recognition results in preferential proliferation and expansion of responding CD8+ T cells rather than promotion of cell death Proc Natl Acad Sci U.S.A 99, 3001–3006 (2002) Charo, J et al Bcl-2 overexpression enhances tumor-specific T-cell survival Cancer Res 65, 2001–2008 (2005) Kalos, M et al T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia Sci Transl Med 3, 95ra73 (2011) Sadelain, M., Rivière, I & Brentjens, R Targeting tumours with genetically enhanced T lymphocytes Nat Rev Cancer 3, 35–45 (2003) Riddell, S R & Greenberg, P D Principles for adoptive T cell therapy of human viral diseases Annu Rev Immunol 13, 545–586 (1995) Sewell, A K Why must T cells be cross-reactive? Nat Rev Immunol 12, 669–677 (2012) Humphries, C Adoptive cell therapy: Honing that killer instinct Nature 504, S13–5 (2013) Linette, G P et al Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma Blood 122, 863–871 (2013) Morgan, R A et al Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2 Mol Ther 18, 843–851 (2010) Hinrichs, C S et al Adoptively transferred effector cells derived from naive rather than central memory CD8+ T cells mediate superior antitumor immunity Proc Natl Acad Sci U.S.A 106, 17469–17474 (2009) Berger, C et al Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates J Clin Invest 118, 294–305 (2008) Tuohy, V K et al The epitope spreading cascade during progression of experimental autoimmune encephalomyelitis and multiple sclerosis Immunol Rev 164, 93–100 (1998) Kent, S C et al Expanded T cells from pancreatic lymph nodes of type diabetic subjects recognize an insulin epitope Nature 435, 224–228 (2005) Song, Q et al Novel autoimmune hepatitis-specific autoantigens identified using protein microarray technology J Proteome Res 9, 30–39 (2010) Rossini, A., Greiner, D & Friedman, H Immunopathogenesis of diabetes mellitus (Diabetes Rev, 1993) Mason, U et al CD4 coating, but not CD4 depletion, is a predictor of efficacy with primatized monoclonal anti-CD4 treatment of active rheumatoid arthritis J Rheumatol 29, 220–229 (2002) Sia, C Imbalance in Th cell polarization and its relevance in type diabetes mellitus Rev Diabet Stud 2, 182–186 (2005) Pauza, M E., Neal, H., Hagenbaugh, A., Cheroutre, H & Lo, D T-cell production of an inducible interleukin-10 transgene provides limited protection from autoimmune diabetes Diabetes 48, 1948–1953 (1999) Zaccone, P et al Interleukin-13 prevents autoimmune diabetes in NOD mice Diabetes 48, 1522–1528 (1999) Stokkers, P C F & Hommes, D W New cytokine therapeutics for inflammatory bowel disease Cytokine 28, 167–173 (2004) Kumar, V et al Recombinant T cell receptor molecules can prevent and 135   88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 reverse experimental autoimmune encephalomyelitis: dose effects and involvement of both CD4 and CD8 T cells J Immunol 159, 5150–5156 (1997) Accardi, L & Di Bonito, P Antibodies in single-chain format against tumour-associated antigens: present and future applications Curr Med Chem 17, 1730–1755 (2010) Jäger, E et al Simultaneous humoral and cellular immune response against cancer-testis antigen NY-ESO-1: definition of human histocompatibility leukocyte antigen (HLA)-A2-binding peptide epitopes J Exp Med 187, 265–270 (1998) Renkvist, N., Castelli, C., Robbins, P F & Parmiani, G A listing of human tumor antigens recognized by T cells Cancer Immunol Immunother 50, 3– 15 (2001) Boulter, J M & Jakobsen, B K Stable, soluble, high-affinity, engineered T cell receptors: novel antibody-like proteins for specific targeting of peptide antigens Clin Exp Immunol 142, 454–460 (2005) Meissner, M., Reichert, T E., Kunkel, M & Gooding, W Defects in the human leukocyte antigen class I antigen processing machinery in head and neck squamous cell carcinoma: association with clinical outcome Clinical cancer … (2005) Epel, M., Ellenhorn, J D., Diamond, D J & Reiter, Y A functional recombinant single-chain T cell receptor fragment capable of selectively targeting antigen-presenting cells Cancer Immunol Immunother 51, 565– 573 (2002) Card, K F et al A soluble single-chain T-cell receptor IL-2 fusion protein retains MHC-restricted peptide specificity and IL-2 bioactivity Cancer Immunol Immunother 53, 345–357 (2004) Halin, C et al Synergistic therapeutic effects of a tumor targeting antibody fragment, fused to interleukin 12 and to tumor necrosis factor alpha Cancer Res 63, 3202–3210 (2003) Topham, D J., Tripp, R A & Doherty, P C CD8+ T cells clear influenza virus by perforin or Fas-dependent processes (1997) Tenter, A M., Heckeroth, A R & Weiss, L M Toxoplasma gondii: from animals to humans International Journal for Parasitology 30, 1217–1258 (2000) Khan, I A., Smith, K A & Kasper, L H Induction of antigen-specific parasiticidal cytotoxic T cell splenocytes by a major membrane protein (P30) of Toxoplasma gondii J Immunol 141, 3600–3605 (1988) Suzuki, Y., Orellana, M A., Schreiber, R D & Remington, J S Interferongamma: the major mediator of resistance against Toxoplasma gondii Science 240, 516–518 (1988) Andrade, R M., Wessendarp, M., Gubbels, M.-J., Striepen, B & Subauste, C S CD40 induces macrophage anti-Toxoplasma gondii activity by triggering autophagy-dependent fusion of pathogen-containing vacuoles and lysosomes J Clin Invest 116, 2366–2377 (2006) Plaksin, D., Polakova, K., McPhie, P & Margulies, D H A three-domain T cell receptor is biologically active and specifically stains cell surface MHC/peptide complexes J Immunol 158, 2218–2227 (1997) Novotny, J et al A soluble, single-chain T-cell receptor fragment endowed with antigen-combining properties Proc Natl Acad Sci U.S.A 88, 8646– 8650 (1991) Chung, S., Wucherpfennig, K W., Friedman, S M., Hafler, D A & Strominger, J L Functional three-domain single-chain T-cell receptors Proc Natl Acad Sci U.S.A 91, 12654–12658 (1994) Stone, J D., Chervin, A S., Schreiber, H & Kranz, D M Design and 136   105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 characterization of a protein superagonist of IL-15 fused with IL-15Rα and a high-affinity T cell receptor Biotechnol Prog (2012) doi:10.1002/btpr.1631 Belmont, H J et al Potent antitumor activity of a tumor-specific soluble TCR/IL-2 fusion protein Clin Immunol 121, 29–39 (2006) Willcox, B E et al Production of soluble [alpha][beta] T-cell receptor heterodimers suitable for biophysical analysis of ligand binding Protein Sci 8, 2418–2423 (1999) Boulter, J M et al Stable, soluble T-cell receptor molecules for crystallization and therapeutics Protein Eng 16, 707–711 (2003) Miklos, A E et al Structure-Based Design of Supercharged, Highly Thermoresistant Antibodies Chemistry & Biology 19, 449–455 (2012) Lawrence, M S., Phillips, K J & Liu, D R Supercharging Proteins Can Impart Unusual Resilience J Am Chem Soc 129, 10110–10112 (2007) Caldwell, G W., Ritchie, D M., Masucci, J A., Hageman, W & Yan, Z The new pre-preclinical paradigm: compound optimization in early and late phase drug discovery Curr Top Med Chem 1, 353–366 (2001) Schellekens, H Bioequivalence and the immunogenicity of biopharmaceuticals Nat Rev Drug Discov 1, 457–462 (2002) Hardy, J & Selkoe, D J The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics Science 297, 353–356 (2002) Wang, W Protein aggregation and its inhibition in biopharmaceutics Int J Pharm 289, 1–30 (2005) Loeb, J CHEMICAL AND PHYSICAL BEHAVIOR OF CASEIN SOLUTIONS J Gen Physiol 3, 547–555 (1921) Rationalization of the effects of mutations on peptide andprotein aggregation rates (2003) at Fink, A L Natively unfolded proteins Curr Opin Struct Biol (2005) Vucetic, S., Brown, C J & Dunker, A K Flavors of protein disorder Proteins: Structure (2003) Pawar, A P., Dubay, K F., Zurdo, J & Chiti, F Prediction of “aggregationprone” and “aggregation-susceptible” regions in proteins associated with neurodegenerative diseases Journal of molecular … (2005) Dosztányi, Z., Csizmók, V., Tompa, P & Simon, I The pairwise energy content estimated from amino acid composition discriminates between folded and intrinsically unstructured proteins J Mol Biol 347, 827–839 (2005) Gudiksen, K L et al Eliminating positively charged lysine epsilon-NH3+ groups on the surface of carbonic anhydrase has no significant influence on its folding from sodium dodecyl sulfate J Am Chem Soc 127, 4707–4714 (2005) McNaughton, B R., Cronican, J J., Thompson, D B & Liu, D R Mammalian cell penetration, siRNA transfection, and DNA transfection by supercharged proteins Proc Natl Acad Sci U.S.A 106, 6111–6116 (2009) Cronican, J J et al Potent delivery of functional proteins into Mammalian cells in vitro and in vivo using a supercharged protein ACS Chem Biol 5, 747–752 (2010) Hitsuda, T et al A protein transduction method using oligo-arginine (3R) for the delivery of transcription factors into cell nuclei Biomaterials 33, 4665–4672 (2012) Sun, J & Braciale, T J Role of T cell immunity in recovery from influenza virus infection Current opinion in virology 3, 425–429 (2013) Spence, P J & Langhorne, J T cell control of malaria pathogenesis 137   126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 Current Opinion in Immunology (2012) Gigley, J P., Bhadra, R & Khan, I A CD8 T Cells and Toxoplasma gondii: A New Paradigm Journal of Parasitology Research 2011, 1–9 (2011) Hewitt, E W The MHC class I antigen presentation pathway: strategies for viral immune evasion Immunology 110, 163–169 (2003) Gras, S., Kjer-Nielsen, L & Burrows, S T-cell receptor bias and immunity Current opinion in … (2008) Argaet, V P et al Dominant selection of an invariant T cell antigen receptor in response to persistent infection by Epstein-Barr virus J Exp Med 180, 2335–2340 (1994) Moss, P A et al Extensive conservation of alpha and beta chains of the human T-cell antigen receptor recognizing HLA-A2 and influenza A matrix peptide Proc Natl Acad Sci U.S.A 88, 8987–8990 (1991) Kjer-Nielsen, L., Clements, C & Purcell, A A structural basis for the selection of dominant αβ T cell receptors in antiviral immunity Immunity (2003) Sullivan, W J., Jr & Jeffers, V Mechanisms of Toxoplasma gondii persistence and latency FEMS Microbiol Rev 36, 717–733 (2011) Weiss, L M & Dubey, J P Toxoplasmosis: A history of clinical observations International Journal for Parasitology 39, 895–901 (2009) Torgerson, P R & Mastroiacovo, P The global burden of congenital toxoplasmosis: a systematic review Bull World Health Organ 91, 501–508 (2013) Suzuki, Y & Remington, J S The effect of anti-IFN-gamma antibody on the protective effect of Lyt-2+ immune T cells against toxoplasmosis in mice J Immunol 144, 1954–1956 (1990) Brown, C R & McLeod, R Class I MHC genes and CD8+ T cells determine cyst number in Toxoplasma gondii infection J Immunol 145, 3438–3441 (1990) Wilson, D C., Grotenbreg, G M., Liu, K & Zhao, Y Differential regulation of effector-and central-memory responses to Toxoplasma gondii infection by IL-12 revealed by tracking of Tgd057-specific CD8+ T cells PLoS Pathog (2010) Blanchard, N et al Immunodominant, protective response to the parasite Toxoplasma gondii requires antigen processing in the endoplasmic reticulum Nat Immunol 9, 937–944 (2008) Klock, H E., Koesema, E J., Knuth, M W & Lesley, S A Combining the polymerase incomplete primer extension method for cloning and mutagenesis with microscreening to accelerate structural genomics efforts Proteins 71, 982–994 (2008) Sørensen, H P Towards universal systems for recombinant gene expression Microb Cell Fact 9, 27 (2010) Rudolph, R In vitro folding of inclusion body proteins The FASEB Journal (1996) De Bernardez Clark, E Refolding of recombinant proteins Curr Opin Biotechnol (1998) Garboczi, D N et al Structure of the complex between human T-cell receptor, viral peptide and HLA-A2 Nature 384, 134–141 (1996) Colf, L., Bankovich, A., Hanick, N & Bowerman, N How a single T cell receptor recognizes both self and foreign MHC Cell (2007) Antos, J M et al Site-specific N- and C-terminal labeling of a single polypeptide using sortases of different specificity J Am Chem Soc 131, 10800–10801 (2009) Rudolph, M G., Stanfield, R L & Wilson, I A How TCRs bind MHCs, peptides, and coreceptors Annu Rev Immunol 24, 419–466 (2006) 138   147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 Zhu, X et al Visualization of p53264–272/HLA-A* 0201 complexes naturally presented on tumor cell surface by a multimeric soluble singlechain T cell receptor (2006) Stone, J D., Chervin, A S & Kranz, D M T-cell receptor binding affinities and kinetics: impact on T-cell activity and specificity Immunology 126, 165–176 (2009) Rosette, C et al The impact of duration versus extent of TCR occupancy on T cell activation: a revision of the kinetic proofreading model Immunity 15, 59–70 (2001) McKeithan, T W Kinetic proofreading in T-cell receptor signal transduction Proc Natl Acad Sci U.S.A 92, 5042–5046 (1995) Blander, J M., Sant'Angelo, D B., Bottomly, K & Janeway, C A Alteration at a single amino acid residue in the T cell receptor alpha chain complementarity determining region changes the differentiation of naive CD4 T cells in response to antigen from T helper cell type (Th1) to Th2 J Exp Med 191, 2065–2074 (2000) Valitutti, S., Müller, S., Cella, M., Padovan, E & Lanzavecchia, A Serial triggering of many T-cell receptors by a few peptide-MHC complexes Nature 375, 148–151 (1995) Matsui, K., Boniface, J J., Steffner, P., Reay, P A & Davis, M M Kinetics of T-cell receptor binding to peptide/I-Ek complexes: correlation of the dissociation rate with T-cell responsiveness Proc Natl Acad Sci U.S.A 91, 12862–12866 (1994) Corr, M et al T cell receptor-MHC class I peptide interactions: affinity, kinetics, and specificity Science 265, 946–949 (1994) Valitutti, S., Dessing, M., Aktories, K., Gallati, H & Lanzavecchia, A Sustained signaling leading to T cell activation results from prolonged T cell receptor occupancy Role of T cell actin cytoskeleton (1995) Valitutti, S & Lanzavecchia, A Serial triggering of TCRs: a basis for the sensitivity and specificity of antigen recognition Immunol Today 18, 299– 304 (1997) Irvine, D J., Purbhoo, M A., Krogsgaard, M & Davis, M M Direct observation of ligand recognition by T cells Nature 419, 845–849 (2002) Holler, P D & Kranz, D M Quantitative analysis of the contribution of TCR/pepMHC affinity and CD8 to T cell activation Immunity 18, 255–264 (2003) Li, Y et al Directed evolution of human T-cell receptors with picomolar affinities by phage display Nat Biotechnol 23, 349–354 (2005) Kalergis, A M et al Efficient T cell activation requires an optimal dwelltime of interaction between the TCR and the pMHC complex Nat Immunol 2, 229–234 (2001) Rabinowitz, J D., Beeson, C., Lyons, D S., Davis, M M & McConnell, H M Kinetic discrimination in T-cell activation (1996) Cole, D K et al Human TCR-binding affinity is governed by MHC class restriction J Immunol 178, 5727–5734 (2007) Gras, S et al A structural basis for varied αβ TCR usage against an immunodominant EBV antigen restricted to a HLA-B8 molecule The Journal of Immunology 188, 311–321 (2012) Davis-Harrison, R L., Armstrong, K M & Baker, B M Two different T cell receptors use different thermodynamic strategies to recognize the same peptide/MHC ligand J Mol Biol 346, 533–550 (2005) Purbhoo, M A et al Quantifying and imaging NY-ESO-1/LAGE-1-derived epitopes on tumor cells using high affinity T cell receptors (2006) Stefani, M & Dobson, C M Protein aggregation and aggregate toxicity: new insights into protein folding, misfolding diseases and biological 139   167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 evolution J Mol Med 81, 678–699 (2003) Thomas, T., Thomas, G., McLendon, C., Sutton, T & Mullan, M betaAmyloid-mediated vasoactivity and vascular endothelial damage Nature 380, 168–171 (1996) Kelly, J W The alternative conformations of amyloidogenic proteins and their multi-step assembly pathways Curr Opin Struct Biol 8, 101–106 (1998) Dobson, C M The structural basis of protein folding and its links with human disease Philos Trans R Soc Lond., B, Biol Sci 356, 133–145 (2001) Frokjaer, S & Otzen, D E Protein drug stability: a formulation challenge Nat Rev Drug Discov 4, 298–306 (2005) Pace, C N., Shirley, B A., McNutt, M & Gajiwala, K Forces contributing to the conformational stability of proteins FASEB J 10, 75–83 (1996) Simeonov, P., Berger-Hoffmann, R., Hoffmann, R., Sträter, N & Zuchner, T Surface supercharged human enteropeptidase light chain shows improved solubility and refolding yield Protein Eng Des Sel 24, 261–268 (2011) Chiti, F., Stefani, M., Taddei, N., Ramponi, G & Dobson, C M Rationalization of the effects of mutations on peptide and protein aggregation rates Nature 424, 805–808 (2003) Liu, Y & Kuhlman, B RosettaDesign server for protein design Nucleic Acids Res 34, W235–8 (2006) Bishop, J R., Schuksz, M & Esko, J D Heparan sulphate proteoglycans fine-tune mammalian physiology Nature 446, 1030–1037 (2007) Niesen, F H., Berglund, H & Vedadi, M The use of differential scanning fluorimetry to detect ligand interactions that promote protein stability Nat Protoc 2, 2212–2221 (2007) Schellman, J A Temperature, stability, and the hydrophobic interaction Biophys J (1997) Ni, C Z et al Crystal structure of the MS2 coat protein dimer: implications for RNA binding and virus assembly Structure 3, 255–263 (1995) Low, J L et al Binding of TCR multimers and a TCR-like antibody with distinct fine-specificities is dependent on the surface density of HLA complexes PLoS ONE 7, e51397 (2012) Der, B S et al Alternative computational protocols for supercharging protein surfaces for reversible unfolding and retention of stability PLoS ONE 8, e64363 (2013) Makhatadze, G I., Loladze, V V., Ermolenko, D N., Chen, X & Thomas, S T Contribution of surface salt bridges to protein stability: guidelines for protein engineering J Mol Biol 327, 1135–1148 (2003) Shivu, B et al Distinct β-sheet structure in protein aggregates determined by ATR-FTIR spectroscopy Biochemistry 52, 5176–5183 (2013) Street, A G & Mayo, S L Intrinsic beta-sheet propensities result from van der Waals interactions between side chains and the local backbone Proc Natl Acad Sci U.S.A 96, 9074–9076 (1999) Leaver-Fay, A et al ROSETTA3: an object-oriented software suite for the simulation and design of macromolecules Meth Enzymol 487, 545–574 (2011) Rohl, C A., Strauss, C & Misura, K Protein structure prediction using Rosetta Meth Enzymol (2004) Roca, M., Messer, B & Warshel, A Electrostatic contributions to protein stability and folding energy FEBS Lett 581, 2065–2071 (2007) Joughin, B A., Green, D F & Tidor, B Action-at-a-distance interactions enhance protein binding affinity Protein Sci 14, 1363–1369 (2005) Zhou, H.-X & Dong, F Electrostatic contributions to the stability of a 140   189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 thermophilic cold shock protein Biophys J 84, 2216–2222 (2003) Dill, K A Dominant forces in protein folding Biochemistry (1990) Strickler, S S et al Protein stability and surface electrostatics: a charged relationship Biochemistry 45, 2761–2766 (2006) Chan, C.-H., Wilbanks, C C., Makhatadze, G I & Wong, K.-B Electrostatic contribution of surface charge residues to the stability of a thermophilic protein: benchmarking experimental and predicted pKa values PLoS ONE 7, e30296 (2012) Sanchez-Ruiz, J M & Makhatadze, G I To charge or not to charge? Trends Biotechnol 19, 132–135 (2001) Callan, M F et al Large clonal expansions of CD8+ T cells in acute infectious mononucleosis Nat Med 2, 906–911 (1996) Butz, E A & Bevan, M J Massive expansion of antigen-specific CD8+ T cells during an acute virus infection Immunity 8, 167–175 (1998) Zhang, N & Bevan, M J CD8(+) T cells: foot soldiers of the immune system Immunity 35, 161–168 (2011) Clements, C S., Dunstone, M A., Macdonald, W A., McCluskey, J & Rossjohn, J Specificity on a knife-edge: the alphabeta T cell receptor Curr Opin Struct Biol 16, 787–795 (2006) Deng, L & Mariuzza, R A Recognition of self-peptide-MHC complexes by autoimmune T-cell receptors Trends Biochem Sci 32, 500–508 (2007) Nicholson, M J., Hahn, M & Wucherpfennig, K W Unusual features of self-peptide/MHC binding by autoimmune T cell receptors Immunity 23, 351–360 (2005) Wucherpfennig, K W., Call, M J., Deng, L & Mariuzza, R Structural alterations in peptide-MHC recognition by self-reactive T cell receptors Current Opinion in Immunology 21, 590–595 (2009) Tynan, F E et al T cell receptor recognition of a ‘super-bulged’ major histocompatibility complex class I-bound peptide Nat Immunol 6, 1114– 1122 (2005) Garcia, K C., Adams, J J., Feng, D & Ely, L K The molecular basis of TCR germline bias for MHC is surprisingly simple Nat Immunol 10, 143– 147 (2009) Dai, S., Huseby, E S., Rubtsova, K & Scott-Browne, J Crossreactive T Cells spotlight the germline rules for αβ T cell-receptor interactions with MHC molecules Immunity (2008) Burrows, S R et al Hard wiring of T cell receptor specificity for the major histocompatibility complex is underpinned by TCR adaptability (2010) Godfrey, D & Rossjohn, J The fidelity, occasional promiscuity, and versatility of T cell receptor recognition Immunity (2008) Wu, L C., Tuot, D S., Lyons, D S., Garcia, K C & Davis, M M Twostep binding mechanism for T-cell receptor recognition of peptide–MHC Nature (2002) Liu, Y C et al The energetic basis underpinning T-cell receptor recognition of a super-bulged peptide bound to a major histocompatibility complex class I molecule J Biol Chem 287, 12267–12276 (2012) Boulter, J M et al Potent T cell agonism mediated by a very rapid TCR/pMHC interaction Eur J Immunol 37, 798–806 (2007) Ely, L K et al Antagonism of antiviral and allogeneic activity of a human public CTL clonotype by a single altered peptide ligand: implications for allograft rejection J Immunol 174, 5593–5601 (2005) Balendiran, G K et al The three-dimensional structure of an H-2Ldpeptide complex explains the unique interaction of Ld with beta-2 microglobulin and peptide Proc Natl Acad Sci U.S.A 94, 6880–6885 (1997) 141   210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 Smith, C A & Kortemme, T Backrub-like backbone simulation recapitulates natural protein conformational variability and improves mutant side-chain prediction J Mol Biol 380, 742–756 (2008) Tian, S., Maile, R & Collins, E J CD8+ T cell activation is governed by TCR-peptide/MHC affinity, not dissociation rate The Journal of … (2007) Falk, K., Rötzschke, O., Stevanović, S., Jung, G & Rammensee, H G Allele-specific motifs revealed by sequencing of self-peptides eluted from MHC molecules Nature 351, 290–296 (1991) Saper, M A., Bjorkman, P J & Wiley, D C Refined structure of the human histocompatibility antigen HLA-A2 at 2.6 A resolution J Mol Biol 219, 277–319 (1991) Corr, M et al Endogenous peptides of a soluble major histocompatibility complex class I molecule, H-2Lds: sequence motif, quantitative binding, and molecular modeling of the complex J Exp Med 176, 1681–1692 (1992) Mandl, J N., Monteiro, J P., Vrisekoop, N & Germain, R N T cellpositive selection uses self-ligand binding strength to optimize repertoire recognition of foreign antigens Immunity 38, 263–274 (2013) Persaud, S P., Parker, C R., Lo, W.-L., Weber, K S & Allen, P M Intrinsic CD4+ T cell sensitivity and response to a pathogen are set and sustained by avidity for thymic and peripheral complexes of self peptide and MHC Nat Immunol 15, 266–274 (2014) van der Merwe, P A & Dushek, O Mechanisms for T cell receptor triggering Nat Rev Immunol (2010) Ernst, B., Lee, D S., Chang, J M., Sprent, J & Surh, C D The peptide ligands mediating positive selection in the thymus control T cell survival and homeostatic proliferation in the periphery Immunity 11, 173–181 (1999) Krogsgaard, M., Li, Q., Sumen, C., Huppa, J B & Huse, M Agonist/endogenous peptide–MHC heterodimers drive T cell activation and sensitivity Nature (2005) Lo, W.-L et al An endogenous peptide positively selects and augments the activation and survival of peripheral CD4+ T cells Nat Immunol 10, 1155– 1161 (2009) Kirberg, J., Berns, A & Boehmer, von, H Peripheral T cell survival requires continual ligation of the T cell receptor to major histocompatibility complex-encoded molecules J Exp Med 186, 1269–1275 (1997) Azzam, H S et al CD5 expression is developmentally regulated by T cell receptor (TCR) signals and TCR avidity J Exp Med 188, 2301–2311 (1998) Moran, A E et al T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse Journal of Experimental Medicine 208, 1279–1289 (2011) Corse, E., Gottschalk, R A., Krogsgaard, M & Allison, J P Attenuated T cell responses to a high-potency ligand in vivo PLoS Biol 8, (2010) Slansky, J E & Jordan, K R The Goldilocks model for TCR—too much attraction might not be best for vaccine design PLoS Biol (2010) Irving, M et al Interplay between T cell receptor binding kinetics and the level of cognate peptide presented by major histocompatibility complexes governs CD8+ T cell responsiveness J Biol Chem (2012) doi:10.1074/jbc.M112.357673 142   Annex A Amino acid sequence, molecular weight and isoelectric point (p.I.) of TCR chains: Rop7c1α MGQQVQQSPASLVLQEGENAELQCNFSTSLNSMQWFYQRPGGSLVSL FYNPSGTKQSGRLTSTTVIKERRSSLHISSSQTTDSGTYLCAMGDTNAY KVIFGKGTHLHVLPNIQNPEPAVYQLKDPRSQDSTLCLFTDFDSQINVP KTMESGTFITDKCVLDMKAMDSKSNGAIAWSNQTSFTCQDIFKETNA TYPSS Predicted p.I.: 5.88 Molecular weight: 21.71 kDa Rop7c1β MEAAVTQSPRNKVTVTGGNVTLSCRQTNSHNYMYWYRQDTGHGLR LIHYSYGAGNLQIGDVPDGYKATRTTQEDFFLLLELASPSQTSLYFCAS SEAGDTEVFFGKGTRLTVVEDLRNVTPPKVSLFEPSKAEIANKQKATL VCLARGFFPDHVELSWWVNGKEVHSGVCTDPQAYKESNYSYALSSR LRVSATFWHNPRNHFRCQVQFHGLSEEDKWPEGSPKPVTQNISAEAW GRALPETGGLNDIFEAQKIEWHE   Predicted p.I.: 6.14 Molecular weight: 29.03 kDa Rop7c2α MGQQVQQSPASLVLQEGENAELQCNFSTSLNSMQWFYQRPGGSLVSL FYNPSGTKQSGRLTSTTVIKERRSSLHISSSQTTDSGTYLCAMEQGNNR IFFGDGTQLVVKPNIQNPEPAVYQLKDPRSQDSTLCLFTDFDSQINVPK TMESGTFITDKCVLDMKAMDSKSNGAIAWSNQTSFTCQDIFKETNAT YPSS Predicted p.I.: 5.14 Molecular weight: 21.66 kDa Rop7c2β MEAAVTQSPRNKVAVTGGKVTLSCNQTNNHNNMYWYRQDTGHGLR LIHYSYGAGSTEKGDIPDGYKASRPSQENFSLILELATPSQTSVYFCASG DETKSSYEQYFGPGTRLTVLEDLRNVTPPKVSLFEPSKAEIANKQKATL VCLARGFFPDHVELSWWVNGKEVHSGVCTDPQAYKESNYSYALSSR LRVSATFWHNPRNHFRCQVQFHGLSEEDKWPEGSPKPVTQNISAEAW GRALPETGGLNDIFEAQKIEWHE   Predicted p.I.: 6.31 Molecular weight: 29.18 kDa Rop7c3α 143   MAQKVQQSPESLSVPEGGMASLNCTSSDRNFQYFWWYRQHSGEGPK ALMSIFSDGDKKEGRFTAHLNKASLHVSLHIRDSQPSDSALYFCAVSA GGSNAKLTFGKGTKLSVKSNIQNPEPAVYQLKDPRSQDSTLCLFTDFD SQINVPKTMESGTFITDKCVLDMKAMDSKSNGAIAWSNQTSFTCQDIF KETNATYPSS Predicted p.I.: 6.89 Rop7c3β Molecular weight: 22.01 kDa MGGIITQTPKFLIGQEGQKLTLKCQQNFNHDTMYWYRQDSGKGLRLI YYSITENDLQKGDLSEGYDASREKKSSFSLTVTSAQKNEMAVFLCASS RDWGYEQYFGPGTRLTVLEDLRNVTPPKVSLFEPSKAEIANKQKATLV CLARGFFPDHVELSWWVNGKEVHSGVCTDPQAYKESNYSYALSSRLR VSATFWHNPRNHFRCQVQFHGLSEEDKWPEGSPKPVTQNISAEAWGR ALPETGGLNDIFEAQKIEWHE Predicted p.I.: 6.40 Molecular weight: 29.40 kDa Molecular Weights and p.I.s of refolded TCRs TCR Rop7c1 Rop7c2 Rop7c3 Molecular Weight (kDa) 50.72 50.82 29.40 p.I 6.08 5.97 6.40 Amino acid sequence, molecular weight and p.I of supercharged TCR chains: Rop7c1α (-25) MGQQVEQSPASLVLQEGEDAELQCNFSTSLNSMQWFYQRPGGSLVSL FYNPSGTKESGRLTSTTVIDEERSSLHISSSQTTDSGTYLCAMGDTNAY KVIFGKGTHLHVLPNIEDPEPAVYQLKDPDSDDSTLCLFTDFDSDIEVP DTMESGTFITDKCVLDMEAMDSKSNGAIAWSEETSFTCQDIFEETDAT YPSS Predicted p.I.: 3.98 Molecular weight: 21.63 kDa Rop7c1α (-16) MGQQVQQSPASLVLQEGENAELQCNFSTSLNSMQWFYQRPGGSLVSL FYNPSGTKQSGRLTSTTVIDERRSSLHISSSQTTDSGTYLCAMGDTNAY KVIFGKGTHLHVLPNIEDPEPAVYQLKDPDSDDSTLCLFTDFDSQIEVP DTMESGTFITDKCVLDMEAMDSKSNGAIAWSNQTSFTCQDIFEETNAT YPSS Predicted p.I.: 4.23 Molecular weight: 21.65 kDa 144   Rop7c1α (+6) MGQQVQQSPASLVLQEGENAELQCNFSTSLNSMQWFYQRPGGSLVSL FYNPSGTKQSGRLTSTTVIKKRRSSLHISSSQTTDSGTYLCAMGDTNAY KVIFGKGTHLHVLPNIKRPEPAVYQLKDPRSRKSTLCLFTDFDSQIKVP KTMESGTFITDKCVLDMKAMDSKSNGAIAWSNQTSFTCQDIFKETNA TYPSS Predicted p.I.: 9.10 Molecular weight: 21.81 kDa Rop7c1α (+23) MGQQVRQSPASLVLQEGRKARLQCNFSTSLNSMQWFYQRPGGSLVSL FYNPSGTKRSGRLTSTTVIKKRRSSLHISSSQTTDSGTYLCAMGDTNAY KVIFGKGTHLHVLPNIKRPRPAVYQLKDPRSRKSTLCLFTDFDSRIKVP KTMESGTFITDKCVLDMKAMKSKSNGAIAWSRKTSFTCQDIFKKTKA TYPSS Predicted p.I.: 10.32 Molecular weight: 22.05 kDa Rop7c1β (-11) MEAAVTQSPRNKVTVTGGNVTLSCRQTNSHNYMYWYRQDTGHGLR LIHYSYGAGNLQIGDVPDGYKATRTTQEDFFLLLELASPSQTSLYFCAS SEAGDTEVFFGKGTRLTVVEDLDNVTPPKVSLFEPSKAEIANKQKATL VCLARGFFPDHVELSWWVNGEEVHSGVCTDPQAYKESEYSYALSSRL RVSATFWHNPRNHFRCQVQFHGLSEEDEWPEGSPKPVTQNISAEAWG RALPETGGLNDIFEAQKIEWHE Predicted p.I.: 5.27 Molecular weight: 29.00 kDa Rop7c1β (-25) MEAAVTQSPRNKVTVTGGEVTLSCEQTNSHNYMYWYRQDTGHGLRL IHYSYGAGNLQIGDVPDGYEATRTTQEDFFLLLELASPSQTSLYFCASS EAGDTEVFFGDGTRLTVVEDLDNVTPPEVSLFEPSDAEIANKDKATLV CLARGFFPDHVELSWWVNGEEVHSGVCTDPQAYKESEYSYALSSRLR VSATFWHNPDNHFRCQVQFHGLSEEDEWPEGSPKPVTQNISAEAWGR ALPETGGLNDIFEAQKIEWHE Predicted p.I.: 4.50 Molecular weight: 28.91 kDa Rop7c1β (+5) MEAAVTQSPRNKVTVTGGNVTLSCRQTNSHNYMYWYRQDTGHGLR LIHYSYGAGNLQIGDVPKGYKATRTTQEDFFLLLELASPSQTSLYFCAS SEAGDTEVFFGKGTRLTVVEDLRNVTPPKVSLFEPSKAEIANKQKATL VCLARGFFPDHVELSWWVNGKEVHSGVCTDPQAYKESKYSYALSSR 145   LRVSATFWHNPRNHFRCQVQFHGLSRKDKWPRGSPKPVTQNISAEAW GRALPETGGLNDIFEAQKIEWHE Predicted p.I.: 8.89 Molecular weight: 29.11 kDa Rop7c1β (+9) MEAAVTQSPRNKVTVTGGRVTLSCRQTNSHNYMYWYRQDTGHGLR LIHYSYGAGNLQIGDVPKGYKATRTTQEDFFLLLRLASPSQTSLYFCAS SEAGDTEVFFGKGTRLTVVEDLRNVTPPKVSLFEPSKAEIANKKKATL VCLARGFFPDHVELSWWVNGKEVHSGVCTDPQAYKESKYSYALSSR LRVSATFWHNPRNHFRCQVQFHGLSRKDKWPRGSPKPVTQNISAEAW GRALPETGGLNDIFEAQKIEWHE Predicted p.I.: 9.33 Molecular weight: 29.18 kDa Molecular Weights and p.I.s of refolded supercharged TCRs TCR Rop7c1α 1β (-11) Rop7c1α 1β (-25) Rop7c1α (-16) 1β (-11) Rop7c1α 1β (+5) Molecular Weight (kDa) 50.64 50.55 50.63 50.74 146   p.I 5.42 4.73 4.74 8.23 ... stages of the parasite Toxoplasma gondii Toxoplasma exists as oocysts, the sexual stage of the parasite, in the guts of cats They can then infect humans or animals that come into close contact with... causing death to the patient receiving the therapy 75 In addition, it is important to choose the appropriate subsets of T cells to transfer given that there are different types of T cell subsets such... selection of cells for transfer to maximize the full therapeutic potential of this approach 20   1.6 TCR therapeutics: Soluble TCRs Another arm of T cell therapeutics would be soluble TCRs They

Ngày đăng: 09/09/2015, 11:31

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan