Control of Innate and Adaptive Immune Responses during Infectious Diseases ppt

189 1.4K 1
Control of Innate and Adaptive Immune Responses during Infectious Diseases ppt

Đang tải... (xem toàn văn)

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

Thông tin tài liệu

Control of Innate and Adaptive Immune Responses during Infectious Diseases Julio Aliberti Editor Control of Innate and Adaptive Immune Responses during Infectious Diseases Editor Julio Aliberti Associate Professor Divisions of Molecular Immunology and Pulmonary Medicine Cincinnati Children’s Hospital Medical Center and School of Medicine University of Cincinnati Cincinnati, OH, USA julio.aliberti@cchmc.org ISBN 978-1-4614-0483-5 e-ISBN 978-1-4614-0484-2 DOI 10.1007/978-1-4614-0484-2 Springer New York Dordrecht Heidelberg London Library of Congress Control Number: 2011936972 © Springer Science+Business Media, LLC 2012 All rights reserved This work may not be translated or copied in whole or in part without the written permission of the publisher (Springer Science+Business Media, LLC, 233 Spring Street, New York, NY 10013, USA), except for brief excerpts in connection with reviews or scholarly analysis Use in connection with any form of information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed is forbidden The use in this publication of trade names, trademarks, service marks, and similar terms, even if they are not identified as such, is not to be taken as an expression of opinion as to whether or not they are subject to proprietary rights Printed on acid-free paper Springer is part of Springer Science+Business Media (www.springer.com) Preface Upon infection, pathogen and host perform a complex interaction that ultimately aims to achieve elimination of the invading microbe with the least amount of damage to host tissues and organs Interestingly, both sides of this equation co-evolved several mechanisms that mediate pathogen recognition, initiation and expansion of immune responses, neutralization of toxic elements and elimination of replicating organisms and finally healing and remodeling of damaged tissues On one side pathogens evolved mechanisms to evade recognition and killing, while on the other side, host express numerous (sometimes redundant) mechanisms of recognition and elimination of the pathogen Nonetheless, it is clear that an absolute successful strategy on the pathogen side would be lethal to both host and pathogen Therefore, several evasion mechanisms are seen among several microbes The most successful ones are not necessarily the most abundantly found within the host, but those that can achieve transmission On the other hand, hosts need a robust and extended immune response in order to expand memory cells This critical balance is where the co-evolution between host and pathogens lies This book covers several aspects of induction, control and evasion of host immune response during infectious diseases Multiple aspects are covered and each chapter focuses on one prominent infectious agent Cincinnati, OH Julio Aliberti v Contents Resolution of Inflammation During Toxoplasma gondii Infection Julio Aliberti Mechanisms of Host Protection and Pathogen Evasion of Immune Response During Tuberculosis Andre Bafica and Julio Aliberti NKT Cell Activation During (Microbial) Infection Jochen Mattner Regulation of Innate Immunity During Trypanosoma cruzi Infection Fredy Roberto Salazar Gutierrez B Cell-Mediated Regulation of Immunity During Leishmania Infection Katherine N Gibson-Corley, Christine A Petersen, and Douglas E Jones 23 39 69 85 Control of the Host Response to Histoplasma Capsulatum George S Deepe, Jr 99 Modulation of T-Cell Mediated Immunity by Cytomegalovirus 121 Chris A Benedict, Ramon Arens, Andrea Loewendorf, and Edith M Janssen T Cell Responses During Human Immunodeficiency Virus (HIV)-1 Infection 141 Claire A Chougnet and Barbara L Shacklett Index 171 vii Contributors Julio Aliberti, Ph.D Associate Professor, Divisions of Molecular Immunology and Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center and School of Medicine, University of Cincinnati, Cincinnati, OH, USA julio.aliberti@cchmc.org Ramon Arens Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA Andre Bafica, M.D., Ph.D Assistant Professor, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil andre.bafica@ufsc.br Chris A Benedict Division of Immune Regulation, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA benedict@liai.org Claire A Chougnet Division of Molecular Immunology, Cincinnati Children’s Hospital Research Foundation and Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA Claire.Chougnet@cchmc.org George S Deepe Jr, M.D Professor, Veterans Affairs Hospital, Cincinnati, OH, USA; Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH, USA george.deepe@uc.edu Katherine N Gibson-Corley Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA, USA Fredy Roberto Salazar Gutierrez, M.D., Ph.D Assistant Professor, School of Medicine, Antonio Nariño University, Bogotá, Colombia salazarfrg@gmail.com ix 164 C.A Chougnet and B.L Shacklett Kinloch-de Loes, S., B Hoen, et al (2005) “Impact of therapeutic immunization on HIV-1 viremia after discontinuation of antiretroviral therapy initiated during acute infection.” J Infect Dis 192(4): 607–17 Kinter, A., J McNally, et al (2007) “Suppression of HIV-specific T cell activity by lymph node CD25+ regulatory T cells from HIV-infected individuals.” Proc Natl Acad Sci USA 104(9): 3390–5 Kolte, L., J C Gaardbo, et al (2009) “Increased levels of regulatory T cells (Tregs) in human immunodeficiency virus-infected patients after years of highly active anti-retroviral therapy may be due to increased thymic production of naive Tregs.” Clin Exp Immunol 155(1): 44–52 Korn, T., E Bettelli, et al (2009) “IL-17 and Th17 Cells.” Annu Rev Immunol 27: 485–517 Kornfeld, C., M J Ploquin, et al (2005) “Antiinflammatory profiles during primary SIV infection in African green monkeys are associated with protection against AIDS.” J Clin Invest 115(4): 1082–91 Kosmrlj, A., E L Read, et al (2010) “Effects of thymic selection of the T-cell repertoire on HLA class I-associated control of HIV infection.” Nature 465(7296): 350–4 Koup, R A., J T Safrit, et al (1994) “Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type syndrome.” J Virol 68(7): 4650–5 Kramer, R A., M D Schaber, et al (1986) “HTLV-III gag protein is processed in yeast cells by the virus pol-protease.” Science 231(4745): 1580–4 Kuritzkes, D R and B D Walker (2007) HIV-1 Pathogenesis, Clinical Manifestations and Treatment Fields Virology D Knipe and P M Howley Philadelphia, PA, Lippincott Williams & Wilkins 2: 2187–2214 Laforge, M., F Petit, et al (2007) “Commitment to apoptosis in CD4(+) T lymphocytes productively infected with human immunodeficiency virus type is initiated by lysosomal membrane permeabilization, itself induced by the isolated expression of the viral protein Nef.” J Virol 81(20): 11426–40 Lederman, M M (2001) “Immune restoration and CD4+ T-cell function with antiretroviral therapies.” AIDS 15 Suppl 2: S11-5 Legrand, F A., D F Nixon, et al (2006) “Strong HIV-1-specific T cell responses in HIV-1exposed uninfected infants and neonates revealed after regulatory T cell removal.” PLoS One 1: e102 Legrand, N., A Ploss, et al (2009) “Humanized mice for modeling human infectious disease: challenges, progress, and outlook.” Cell Host Microbe 6(1): 5–9 Leslie, A J., K J Pfafferott, et al (2004) “HIV evolution: CTL escape mutation and reversion after transmission.” Nat Med 10(3): 282–9 Letvin, N L., J R Mascola, et al (2006) “Preserved CD4+ central memory T cells and survival in vaccinated SIV-challenged monkeys.” Science 312(5779): 1530–3 Levesque, M C., M A Moody, et al (2009) “Polyclonal B cell differentiation and loss of gastrointestinal tract germinal centers in the earliest stages of HIV-1 infection.” PLoS Med 6(7): e1000107 Levy, J A (2007) HIV and the Pathogenesis of AIDS Washington, D.C., ASM Press Levy, Y., C Lacabaratz, et al (2009) “Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment.” J Clin Invest 119(4): 997–1007 Li, Q., L Duan, et al (2005) “Peak SIV replication in resting memory CD4+ T cells depletes gut lamina propria CD4+ T cells.” Nature 434(7037): 1148–52 Li, Q., P J Skinner, et al (2009) “Visualizing antigen-specific and infected cells in situ predicts outcomes in early viral infection.” Science 323(5922): 1726–9 Li, S., E J Gowans, et al (2008) “Natural regulatory T cells and persistent viral infection.” J Virol 82(1): 21–30 Lichterfeld, M., D E Kaufmann, et al (2004) “Loss of HIV-1-specific CD8+ T cell proliferation after acute HIV-1 infection and restoration by vaccine-induced HIV-1-specific CD4+ T cells.” J Exp Med 200(6): 701–12 Lifson, J D., M B Feinberg, et al (1986) “Induction of CD4-dependent cell fusion by the HTLVIII/LAV envelope glycoprotein.” Nature 323(6090): 725–8 T Cell Responses During Human Immunodeficiency Virus (HIV)-1 Infection 165 Lim, A., D Tan, et al (2007) “Proportions of circulating T cells with a regulatory cell phenotype increase with HIV-associated immune activation and remain high on antiretroviral therapy.” AIDS 21(12): 1525–34 Liu, J., N Gong, et al (2009) “Neuromodulatory activities of CD4 + CD25+ regulatory T cells in a murine model of HIV-1-associated neurodegeneration.” J Immunol 182(6): 3855–65 Liu, J., B F Keele, et al (2010) “Low-dose mucosal simian immunodeficiency virus infection restricts early replication kinetics and transmitted virus variants in rhesus monkeys.” J Virol 84(19): 10406–12 Lopez-Herrera, A., Y Liu, et al (2005) “HIV-1 interaction with human mannose receptor (hMR) induces production of matrix metalloproteinase (MMP-2) through hMR-mediated intracellular signaling in astrocytes.” Biochim Biophys Acta 1741(1–2): 55–64 Macal, M., S Sankaran, et al (2008) “Effective CD4+ T-cell restoration in gut-associated lymphoid tissue of HIV-infected patients is associated with enhanced Th17 cells and polyfunctional HIV-specific T-cell responses.” Mucosal Immunol 1(6): 475–88 Maecker, H T (2009) “Multiparameter flow cytometry monitoring of T cell responses.” Methods Mol Biol 485: 375–91 Maggi, E., M Mazzetti, et al (1994) “Ability of HIV to promote a TH1 to TH0 shift and to replicate preferentially in TH2 and TH0 cells.” Science 265(5169): 244–8 Makedonas, G and M R Betts (2011) “Living in a house of cards: re-evaluating CD8+ T-cell immune correlates against HIV.” Immunol Rev 239(1): 109–24 Malhotra, U., S Holte, et al (2001) “Role for HLA class II molecules in HIV-1 suppression and cellular immunity following antiretroviral treatment.” J Clin Invest 107(4): 505–17 Malim, M H., J Hauber, et al (1989) “The HIV-1 rev trans-activator acts through a structured target sequence to activate nuclear export of unspliced viral mRNA.” Nature 338(6212): 254–7 Manches, O., D Munn, et al (2008) “HIV-activated human plasmacytoid DCs induce Tregs through an indoleamine 2,3-dioxygenase-dependent mechanism.” J Clin Invest 118(10): 3431–9 Martin, M P., Y Qi, et al (2007) “Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1.” Nat Genet 39(6): 733–40 Mattapallil, J J., D C Douek, et al (2005) “Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection.” Nature 434(7037): 1093–7 McCune, J M., M B Hanley, et al (2000) “Factors influencing T-cell turnover in HIV-1-seropositive patients.” J Clin Invest 105(5): R1-8 McMichael, A J and C A O’Callaghan (1998) “A new look at T cells.” J Exp Med 187(9): 1367–71 Mehandru, S., M A Poles, et al (2004) “Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract.” J Exp Med 200(6): 761–70 Mellors, J W., A Munoz, et al (1997) “Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection.” Ann Intern Med 126(12): 946–54 Mellors, J W., C R Rinaldo, Jr., et al (1996) “Prognosis in HIV-1 infection predicted by the quantity of virus in plasma [see comments] [published erratum appears in Science 1997 Jan 3;275(5296):14].” Science 272(5265): 1167–70 Michel, N., C Goffinet, et al (2009) “Human cyclin T1 expression ameliorates a T-cell-specific transcriptional limitation for HIV in transgenic rats, but is not sufficient for a spreading infection of prototypic R5 HIV-1 strains ex vivo.” Retrovirology 6: Miedema, F., L Meyaard, et al (1994) “Changing virus-host interactions in the course of HIV-1 infection.” Immunol Rev 140: 35–72 Migueles, S A., A C Laborico, et al (2002) “HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors.” Nat Immunol 3(11): 1061–8 Migueles, S A., C M Osborne, et al (2008) “Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control.” Immunity 29(6): 1009–21 Mikovits, J A., D D Taub, et al (1998) “Similar levels of human immunodeficiency virus type replication in human TH1 and TH2 clones.” J Virol 72(6): 5231–8 Mildvan, D., U Mathur, et al (1982) “Opportunistic infections and immune deficiency in homosexual men.” Ann Intern Med 96(6 Pt 1): 700–4 166 C.A Chougnet and B.L Shacklett Milush, J M., J D Reeves, et al (2007) “Virally induced CD4+ T cell depletion is not sufficient to induce AIDS in a natural host.” J immunol 179(5): 3047–56 Miura, T., M A Brockman, et al (2009) “HLA-associated alterations in replication capacity of chimeric NL4-3 viruses carrying gag-protease from elite controllers of human immunodeficiency virus type 1.” J Virol 83(1): 140–9 Miura, T., M A Brockman, et al (2009) “HLA-B57/B*5801 human immunodeficiency virus type elite controllers select for rare gag variants associated with reduced viral replication capacity and strong cytotoxic T-lymphocyte [corrected] recognition.” J Virol 83(6): 2743–55 Montes, M., C Sanchez, et al (2011) “Normalization of FoxP3(+) regulatory T cells in response to effective antiretroviral therapy.” J Infect Dis 203(4): 496–9 Moonis, M., B Lee, et al (2001) “CCR5 and CXCR4 expression correlated with X4 and R5 HIV-1 infection yet not sustained replication in Th1 and Th2 cells.” AIDS 15(15): 1941–9 Moreno-Fernandez, M E., C Rueda, et al (2011) “Regulatory T cells control HIV replication in activated T cells through a cAMP-dependent mechanism.” Blood 117(20): 5372–80 Moreno-Fernandez, M E., W Zapata, et al (2009) “Human Regulatory T Cells Are Targets for Human Immunodeficiency Virus (HIV) Infection, and Their Susceptibility Differs Depending on the HIV Type Strain.” J Virol 83(24): 12925–12933 Ndhlovu, L C., C P Loo, et al (2008) “FOXP3 expressing CD127lo CD4+ T cells inversely correlate with CD38+ CD8+ T cell activation levels in primary HIV-1 infection.” J Leukoc Biol 83(2): 254–62 Neil, S and P Bieniasz (2009) “Human immunodeficiency virus, restriction factors, and interferon.” J Interferon Cytokine Res 29(9): 569–80 Nilsson, J., A Boasso, et al (2006) “HIV-1-driven regulatory T-cell accumulation in lymphoid tissues is associated with disease progression in HIV/AIDS.” Blood 108(12): 3808–17 Nyakeriga, A M., C J Fichtenbaum, et al (2009) “Engagement of the CD4 receptor affects the redistribution of Lck to the immunological synapse in primary T cells: implications for T-cell activation during human immunodeficiency virus type infection.” J Virol 83(3): 1193–200 Oberlin, E., A Amara, et al (1996) “The CXC chemokine SDF-1 is the ligand for LESTR/fusin and prevents infection by T-cell-line-adapted HIV-1.” Nature 382(6594): 833–5 Ogg, G S., X Jin, et al (1999) “Decay kinetics of human immunodeficiency virus-specific effector cytotoxic T lymphocytes after combination antiretroviral therapy.” J Virol 73(1): 797–800 Ortiz, G M., M Wellons, et al (2001) “Structured antiretroviral treatment interruptions in chronically HIV-1-infected subjects.” Proc Natl Acad Sci USA 98(23): 13288–93 Oyaizu, N., Y Adachi, et al (1997) “Monocytes express Fas ligand upon CD4 cross-linking and induce CD4+ T cells apoptosis: a possible mechanism of bystander cell death in HIV infection.” J immunol 158(5): 2456–63 Palmer, B E., E Boritz, et al (2004) “Effects of sustained HIV-1 plasma viremia on HIV-1 Gagspecific CD4+ T cell maturation and function.” J immunol 172(5): 3337–47 Pandrea, I., G Silvestri, et al (2009) “AIDS in african nonhuman primate hosts of SIVs: a new paradigm of SIV infection.” Curr HIV Res 7(1): 57–72 Pandrea, I V., R Gautam, et al (2007) “Acute loss of intestinal CD4+ T cells is not predictive of simian immunodeficiency virus virulence.” J immunol 179(5): 3035–46 Pelak, K., D B Goldstein, et al (2010) “Host determinants of HIV-1 control in African Americans.” J Infect Dis 201(8): 1141–9 Pereyra, F., X Jia, et al (2010) “The major genetic determinants of HIV-1 control affect HLA class I peptide presentation.” Science 330(6010): 1551–7 Petrovas, C., J P Casazza, et al (2006) “PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection.” J Exp Med 203(10): 2281–92 Qi, Y., M P Martin, et al (2006) “KIR/HLA pleiotropism: protection against both HIV and opportunistic infections.” PLoS Pathog 2(8): e79 Qin, S., Y Sui, et al (2008) “Chemokine and cytokine mediated loss of regulatory T cells in lymph nodes during pathogenic simian immunodeficiency virus infection.” J immunol 180(8): 5530–6 T Cell Responses During Human Immunodeficiency Virus (HIV)-1 Infection 167 Raffatellu, M., R L Santos, et al (2008) “Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency promotes Salmonella dissemination from the gut.” Nat Med 14(4): 421–8 Reynolds, M R., E Rakasz, et al (2005) “CD8+ T-lymphocyte response to major immunodominant epitopes after vaginal exposure to simian immunodeficiency virus: too late and too little.” J Virol 79(14): 9228–35 Reynolds, M R., A M Weiler, et al (2010) “Macaques vaccinated with simian immunodeficiency virus SIVmac239Delta nef delay acquisition and control replication after repeated lowdose heterologous SIV challenge.” J Virol 84(18): 9190–9 Rinaldo, C R (2009) “Dendritic cell-based human immunodeficiency virus vaccine.” J Intern Med 265(1): 138–58 Rinaldo, C R., Jr., J M Liebmann, et al (1999) “Prolonged suppression of human immunodeficiency virus type (HIV-1) viremia in persons with advanced disease results in enhancement of CD4 T cell reactivity to microbial antigens but not to HIV-1 antigens.” J Infect Dis 179(2): 329–36 Robbins, G K., J G Spritzler, et al (2009) “Incomplete reconstitution of T cell subsets on combination antiretroviral therapy in the AIDS Clinical Trials Group protocol 384.” Clin Infect Dis 48(3): 350–61 Rosenberg, E S., M Altfeld, et al (2000) “Immune control of HIV-1 after early treatment of acute infection.” Nature 407: 523–6 Sattentau, Q J and R A Weiss (1988) “The CD4 antigen: physiological ligand and HIV receptor.” Cell 52(5): 631–3 Schacker, T W., C Reilly, et al (2005) “Amount of lymphatic tissue fibrosis in HIV infection predicts magnitude of HAART-associated change in peripheral CD4 cell count.” AIDS 19(18): 2169–71 Schmitz, J E., M J Kuroda, et al (1999) “Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes.” Science 283(5403): 857–60 Schneidewind, A., M A Brockman, et al (2008) “Structural and functional constraints limit options for cytotoxic T-lymphocyte escape in the immunodominant HLA-B27-restricted epitope in human immunodeficiency virus type capsid.” J Virol 82(11): 5594–605 Schneidewind, A., M A Brockman, et al (2007) “Escape from the dominant HLA-B27-restricted cytotoxic T-lymphocyte response in Gag is associated with a dramatic reduction in human immunodeficiency virus type replication.” J Virol 81(22): 12382–93 Schweneker, M., D Favre, et al (2008) “HIV-induced changes in T cell signaling pathways.” J Immunol 180(10): 6490–500 Selby, M J., E S Bain, et al (1989) “Structure, sequence, and position of the stem-loop in tar determine transcriptional elongation by tat through the HIV-1 long terminal repeat.” Genes Dev 3(4): 547–58 Shearer, G M (1998) “HIV-induced immunopathogenesis.” Immunity 9: 587–93 Shearer, G M., E Roilides, et al (1991) “CD4+ T helper cell function is actively suppressed in HIV infection.” Int Conf Aids 7(1) Sodora, D L., J S Allan, et al (2009) “Toward an AIDS vaccine: lessons from natural simian immunodeficiency virus infections of African nonhuman primate hosts.” Nat Med 15(8): 861–5 Sodroski, J., W C Goh, et al (1986) “A second post-transcriptional trans-activator gene required for HTLV-III replication.” Nature 321(6068): 412–7 Spiegel, H M., E DeFalcon, et al (1999) “Changes in frequency of HIV-1-specific cytotoxic T cell precursors and circulating effectors after combination antiretroviral therapy in children.” J Infect Dis 180(2): 359–68 Stamatatos, L., L Morris, et al (2009) “Neutralizing antibodies generated during natural HIV-1 infection: good news for an HIV-1 vaccine?” Nat Med 15(8): 866–70 Starcich, B., L Ratner, et al (1985) “Characterization of long terminal repeat sequences of HTLVIII.” Science 227(4686): 538–40 168 C.A Chougnet and B.L Shacklett Sterne, J A., M May, et al (2009) “Timing of initiation of antiretroviral therapy in AIDS-free HIV-1-infected patients: a collaborative analysis of 18 HIV cohort studies.” Lancet 373(9672): 1352–63 Strebel, K., D Daugherty, et al (1987) “The HIV ‘A’ (sor) gene product is essential for virus infectivity.” Nature 328(6132): 728–30 Strebel, K., T Klimkait, et al (1988) “A novel gene of HIV-1, vpu, and its 16-kilodalton product.” Science 241(4870): 1221–3 Strebel, K., J Luban, et al (2009) “Human cellular restriction factors that target HIV-1 replication.” BMC Med 7: 48 Streeck, H., Z L Brumme, et al (2008) “Antigen load and viral sequence diversification determine the functional profile of HIV-1-specific CD8+ T cells.” PLoS Med 5(5): e100 Streeck, H., J S Jolin, et al (2009) “Human immunodeficiency virus type 1-specific CD8+ T-cell responses during primary infection are major determinants of the viral set point and loss of CD4+ T cells.” J Virol 83(15): 7641–8 Streeck, H., M Lichterfeld, et al (2007) “Recognition of a defined region within p24 gag by CD8+ T cells during primary human immunodeficiency virus type infection in individuals expressing protective HLA class I alleles.” J Virol 81(14): 7725–31 Sun, Y., J E Schmitz, et al (2005) “Dysfunction of simian immunodeficiency virus/simian human immunodeficiency virus-induced IL-2 expression by central memory CD4+ T lymphocytes.” J immunol 174(8): 4753–60 Tamma, S M., N Chirmule, et al (1997) “Signals transduced through the CD4 molecule interfere with TCR/CD3-mediated ras activation leading to T cell anergy/apoptosis.” Clin Immunol Immunopathol 85(2): 195–201 Tanaka, Y., Y Koyanagi, et al (1997) “Productive and lytic infection of human CD4+ type helper T cells with macrophage-tropic human immunodeficiency virus type 1.” J Virol 71(1): 465–70 Teleshova, N., I Frank, et al (2003) “Immunodeficiency virus exploitation of dendritic cells in the early steps of infection.” J Leukoc Biol 74(5): 683–90 Tervo, H M., C Goffinet, et al (2008) “Mouse T-cells restrict replication of human immunodeficiency virus at the level of integration.” Retrovirology 5: 58 Terwilliger, E., J G Sodroski, et al (1986) “Effects of mutations within the orf open reading frame region of human T-cell lymphotropic virus type III (HTLV-III/LAV) on replication and cytopathogenicity.” J Virol 60(2): 754–60 Thorborn, G., L Pomeroy, et al (2010) “Increased sensitivity of CD4+ T-effector cells to CD4 + CD25+ Treg suppression compensates for reduced Treg number in asymptomatic HIV-1 infection.” PLoS ONE 5(2): e9254 Thoulouze, M I., N Sol-Foulon, et al (2006) “Human immunodeficiency virus type-1 infection impairs the formation of the immunological synapse.” Immunity 24(5): 547–61 Trautmann, L., L Janbazian, et al (2006) “Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction.” Nat Med 12(10): 1198–202 Van Duyne, R., C Pedati, et al (2009) “The utilization of humanized mouse models for the study of human retroviral infections.” Retrovirology 6: 76 Veazey, R S., M DeMaria, et al (1998) “Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection.” Science 280(5362): 427–31 Velilla, P A., M T Shata, et al (2008) “Effect of Low-Dose IL-2 Immunotherapy on Frequency and Phenotype of Regulatory T cells and NK Cells in HIV/HCV-coinfected Patients.” AIDS Res Hum Retroviruses 24(1): 52–61 Velu, V., K Titanji, et al (2009) “Enhancing SIV-specific immunity in vivo by PD-1 blockade.” Nature 458(7235): 206–10 Veronese, F D., A L DeVico, et al (1985) “Characterization of gp41 as the transmembrane protein coded by the HTLV-III/LAV envelope gene.” Science 229(4720): 1402–5 Vetter, M L., M E Johnson, et al (2009) “Differences in APOBEC3G expression in CD4+ T helper lymphocyte subtypes modulate HIV-1 infectivity.” PLoS Pathog 5(2): e1000292 T Cell Responses During Human Immunodeficiency Virus (HIV)-1 Infection 169 Vingert, B., S Perez-Patrigeon, et al (2010) “HIV controller CD4+ T cells respond to minimal amounts of Gag antigen due to high TCR avidity.” PLoS Pathog 6(2): e1000780 Weiss, L., V Donkova-Petrini, et al (2004) “Human immunodeficiency virus-driven expansion of CD4 + CD25+ Regulatory T cells Which Suppress HIV-specific CD4 T-cell Responses in HIVinfected Patients.” Blood 104: 3249–56 Weiss, L., F A Letimier, et al (2010) “In vivo expansion of naive and activated CD4 + CD25 + FOXP3+ regulatory T cell populations in interleukin-2-treated HIV patients.” Proc Natl Acad Sci USA 107(23): 10632–7 Wherry, E J and R Ahmed (2004) “Memory CD8 T-cell differentiation during viral infection.” J Virol 78(11): 5535–45 Wherry, E J., J N Blattman, et al (2003) “Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment.” J Virol 77(8): 4911–27 Wherry, E J., S J Ha, et al (2007) “Molecular signature of CD8+ T cell exhaustion during chronic viral infection.” Immunity 27(4): 670–84 Wu, L and V N KewalRamani (2006) “Dendritic-cell interactions with HIV: infection and viral dissemination.” Nat Rev Immunol 6(11): 859–68 Xiao, P., O Usami, et al (2008) “Characterization of a CD4-independent clinical HIV-1 that can efficiently infect human hepatocytes through chemokine (C-X-C motif) receptor 4.” AIDS 22(14): 1749–57 Zhang, R., C Fichtenbaum, et al (2004) “CD40 Ligand dysregulation in HIV infection: HIV gp120 inhibits signaling cascades upstream of CD40 Ligand transcription.” J Immunol 172: 2678–2686 Zhang, Z., T Schuler, et al (1999) “Sexual transmission and propagation of SIV and HIV in resting and activated CD4+ T cells.” Science 286(5443): 1353–7 Zolopa, A R., D S Berger, et al (2010) “Activity of elvitegravir, a once-daily integrase inhibitor, against resistant HIV Type 1: results of a phase 2, randomized, controlled, dose-ranging clinical trial.” J Infect Dis 201(6): 814–22 Zwick, M B and D R Burton (2007) “HIV-1 neutralization: mechanisms and relevance to vaccine design.” Curr HIV Res 5(6): 608–24 Index A Aliberti, J., 1, 23–34 Anti-retroviral therapy and T cell responses HAART AIDS and death, 155 CD4 count levels and healthy volunteers, 155–156 CD4+ T cells amelioration, 156 diagnosis, HIV infection, 155 immunological defects and interruption, 156 limiting viral replication, 156 Treg cell activation and frequency, 156 immune based therapies blocking anti-CTLA–4, 157 goal and IL–2 effects, 157 PD–1/PD-L1, 157 vaccines, 157–158 Arens, R., 121 B Bacillus Calmette Guerin (BCG), 24 Bacterial and non-bacterial infection, iNKt cells bystander indirect activation description, 50–51 microbial infections, dual recognition, 51 Salmonella and TCR signaling, 51 direct recognition, microbial GSLs Agelas mauritianus, 52–53 alphaglucuronylceramides, 52 description, 52 lyme disease, 53 Sphingomonas/Novosphingobium, 52 gram-negative bacteria, 50 parasitic, helminth and fungal infections Candida albicans and Aspergillus fumigatus, 54 control parasitic replication, 54 Plasmodium, 54 role and anti-parasite responses, 54 viral infections detection, viral pathogens, 53 Hepatitis B virus (HBV) model, 53–54 immune defense strategy, 54 iNKT role, 53 lethal outcome, Epstein-Barr virus, 53 role and SAP-Fyn signaling, 53 Bafica, A., 23–34 B and T lymphocyte attenuator (BTLA), 133 B cell-mediated regulation, Leishmania antibodies, 86 arm, immune system, 85–86 CMI, 85 humoral immunity, 86 IL–1 and IL–6, 88 intercellular pathogen clinical infection, 90–91 description, 89 murine models, 92–93 Th1 vs Th2, 89–90 intracellular pathogen antibodies role, 86–87 CMI effects, 86 immune control, Ehrlichia chaffeensis, 87 proinflammator vs anti-inflammatory, 87–88 Th1 response, Fc R, 86 macropages activation and antibodies J Aliberti (ed.), Control of Innate and Adaptive Immune Responses during Infectious Diseases, DOI 10.1007/978-1-4614-0484-2, © Springer Science+Business Media, LLC 2012 171 172 B cell-mediated regulation, Leishmania (cont.) functions, 88–89 NADPH oxidase, 89 nitric oxide (NO) production, 88 superoxide production, 89 Benedict, C.A., 121 BTLA See B and T lymphocyte attenuator Buzoni-Gatel, D., 11 C CD1 system and NKT cells description, 41 group II (CD1d) description, 42 in mouse, 42 restricted diverse type II iNKT cells, 43–44 restricted type I iNKT cells, 42–43 group III (CD1e), 44 group I (CD1a, b, c), mycobacterium tuberculosis, 41 mediated presentation, GSLs, 49 MHC gene complex, 41 self-GSL antigens endogenous presentation, 49 -hexosaminidase B, 50 isoglobotrihexosylceramide (iGb3), 49–50 regulation, 50 self-lipid antigens, 50 species distribution birds, 44 existence, 44 human and guinea pigs, 44–45 isoforms, 44 mycobacteria-infection, 45 CD4+ T cells and HIV affecting subsets regulatory T cells, 149–150 Th17 cells, 149 Th1/Th2 cells, 148–149 AIDS and depletion disease cell loss, morbidity and mortality, 145 cross-linking, 146–147 cytopathic effects, virus, 145–146 GI loss, 145 IFNs effects, 147 increases “bystander” cells, 146 “natural” vs “non-natural” hosts, 145 primary target cells, 146 immune dysregulation abnormalities, 147 Index chronic “tickling”, 148 defects, 147heterodimeric integrin 7, 148 in vitro CD4+ T cells, 147 CD8+ T cells responses, HIV acute/early infection, 154 function and dysfunction, chronic infection ageing, immune system, 155 characterization, 154 contribution, 155 immune exhaustion and LCMV, 154 immune control, 151 lessons and HIV controllers antiretroviral therapy, 152 GWAS, 152 HLA-B*35 alleles and AIDS, 153 human HLA-B*5701, Mamu-B*17, 153 in silico model, 153 MHC class I and II alleles, 152 natural killer (NK) cell function, 152–153 Cell mediated immunity (CMI), 85–87 Chemokine receptors CCR2 binds CCL2, 111, 113 inability, CCL2 and CCL7, 112 L-arginine, fungal elimination, 111, 112 CCR5 IL–17A neutralization, 113 role, T and Th17 cells, 113, 114 Chougnet, C.A., 141 CMI See Cell mediated immunity (CMI) CMV See Cytomegalovirus Culture-derived tachyzoites (STag), 4, 13 Cytomegalovirus (CMV) adaptive T cell responses costimulation, 127–128 MHC-TCR interactions, 126–127 apoptosis pathways, 134 CD8+ and CD4+ T cells HCMV, 125 immunomodulatory effects, DCs, 126 MCMV replication, 124–125 memory inflation, 125–126 DCs, 122–124 epidemiology and pathology HCMV infection, 122 risk factor, 122 herpesviruses, 121 immunosuppressive machinations, 134 modification, T cells B7 costimulation, 130–131 cytokines, 133–134 HVEM/BTLA interactions, 133 Index MHC expressions, 128–130 PD–1/PDL–1 pathway, 132–133 soluble positive cosignals, 131–132 NK and NKT cells, 124 D Debbabi, H., 11 Deepe, G.S Jr., 99 Dendritic cells (DCs) CMV cytokines, 122–123, 131–132 HCMV infection, 126 immune modulatory gene products, 123–124 MCMV, 128, 132 MHC molecules, 126 negative net signal, 128, 129 NK cells, 124 PD-L1, 132 positive net signal, 128, 129 cross-presentation pathway, DC, 104 cytomegalovirus (CMV), 122–124 Histoplasma capsulatum bind and ingest, 103, 104 cross-presentation pathway, 104 population, 101 yeast cells, 103, 104 immunomodulatory effects, 126 intracellular lifestyle, 104 microbial recognition, neutrophils, 104 paralysis, 13 production, DC, 131–132 G Gibson-Corley, K.N., 85–93 Glycosphingolipid (GSL) bacterial and non-bacterial infection Agelas mauritianus, 52–53 alphaglucuronylceramides, 52 description, 52 lyme disease, 53 Sphingomonas/Novosphingobium, 52 CD1 system and NKT cells endogenous presentation, 49 -hexosaminidase B, 50 isoglobotrihexosylceramide (iGb3), 49–50 mediated presentation, GSLs, 49 regulation, 50 self-lipids reportoire, 50 iNKT cell activation GSL antigens and structures, 40 173 in humans, 56 self-GSL antigens, 49–50 Gram-negative, LPS-positive bacteria, 50–53 Granuloma formation and bacilli tuberculosis chronic maintenance, 28 description, 27–28 fibrous capsule, 28 IFN- , 29 IL–10, 30 lipoxins, 29–30 TGF- , 30 TNF, 28–29 Gutoerrez, F.R.S., 69–81 H HAART See Highly Active Anti-Retroviral Therapy (HAART) HCMV See Human cytomegalovirus Hepatitis B virus (HBV) model, 53–54 Herpesvirus entry mediator (HVEM), 133 Highly Active Anti-Retroviral Therapy (HAART), 143, 155–156 Histoplasma capsulatum characterization, inflammatory response M , 101 neutrophils, 100–101 T and B cells, 101 chemokines and chemokine receptors CCR2, 111–113 CCR5, 113 cytokines GM-CSF, 108 IL–1, 108 IL–4, 110 IL–10, 111 IL–17 and Th17, 109 IL–12/IFN- axis, 107–108 TNF- , 109–110 description, 99–100 granuloma intracellular pathogens, 101 T cells, 102 intracellular lifestyle DC, 104 M , 102–103 neutrophils, 103–104 leukotrienes, 113–114 lymphoid cell, infection control B cells, 107 T cells, 104–107 TCR, 105–106 174 Human cytomegalovirus (HCMV) CD4 and CD8 T cells, 125 glycoprotein products, 129 IL–10, 133–134 NK cells, 124 Human immunodeficiency virus (HIV)–1 anti-retroviral therapy effect HAART, 155–156 search immune based therapies, 157–158 CD4+ T cells depletion disease, 145–147 immune dysregulation, 147–148 regulatory T cells, 149–150 Th17 cells, 149 Th1/Th2 cells, 148–149 CD8+ T-cells function and dysfunction, chronic infection, 154–155 immune control, 151 lessons and controllers, 152–153 responses, acute/early infection, 154 cell-cell interaction, 158 discovery, 158 experimental models limitation, heterologous viruses, 144 rodents, 143 SIV and Rhesus macaques (RM), 143–144 immune system, humans and honhumans, 158 life cycle CD4 T cells, 142 gag, pol and env genes, 142 HAART, 143 mRNA encoding, 143 replication, 143 Tat and Rev, transport RNAs, 142–143 T cell activation, 158 transmission cis-/trans-infection, 144 clade B virus, 144 draining lumph nodes, 144 mucosal exposure, 144 phenotypic analysis, 144 route and mucosal infections, 145 virus dose and human transmission, 144–145 HVEM See Herpesvirus entry mediator I IL See Interleukin Immunopathology mechanism, Toxoplasma gondii Index IL–27 and suppression, 12–13 IL–22 role, 11–12 inflammation redundancy and control, 14–15 interleukin–10 central role, 11 mediators and antigen processing, modulatory activities, 10 neutralization, 10 pathogens and poxviruses, 9–10 pro-inflammatory responses, 10 TGF and IL–35, lipoxin A4 control, 14 “DC paralysis”, 13 injection, STAg, 13 Mycobacterium tuberculosis, 16–17 pathogen evasion, 15–16 production, 14 receptors and evidence role, 13 resolution phase cardinal signs, controlling and promoting, homeostasis, omega–3 PUFA/fish oils, 8–9 tissue injury, cause and consequences, TGFdescription, 10 macrophage deactivator, 11 mucosal host/pathogen interaction, 11 role, 12 iNKT cells, humans CD1d-restricted, 55 infections, Novosphingobium/ Sphingomonas spp osocomial and septic shock, 56 xenobiotic-metabolizing properties, 55–56 PBC association GSL recognition role, 56 pathogenesis, 56–57 PDC-E2 homologues, 57 primary biliary cirrhosis (PBC), 55 V 24 iNKT cells, 55 Innate immunity regulation, Trypanosoma cruzi cardiomyopathy, 70–71 cell migration, 79 Chagas’ disease, 69 characterization, 70 cytokines, 78 description, 69 development, cardiac damages, 71 diagnosis, 70 Index GIPL and Tc52 activation, 73 intracellular parasite, 73 production, TGF- and IL–10 and IL.12p70, 73 TLR2 activation, 72 TLR2-/-and MyD88-/-mice, 73 GIPL recognition, 73–74 glycoinositolphospholipids (GIPL), 71 in human beings, 70 interaction and evolution, 71 life cycle, 70 mechanisms, innate immune system, 72 MMP, 80 nitric oxide (NO) APC and T cells and induce apoptosis, 75 deficiency impact, 76 donors and control, 75 production and role, 75 response mechanisms, 75 versatile immune mediators, 75 NLR participation, 74–75 phagocytic cells intracellular replication, 78 intracellular signaling, 77–78 macrophages activation, 76 mechanisms, 78 parasite killing, 77 TLR, 71 TLR9 involvement, 74 Interleukin (IL) cmvIL–10, 133–134 HCMV, IL–10, 133–134 Histoplasma capsulatum IL–1, 108 IL–4, 110 IL–10, 111 IL–17 and Th17, 109 IL–12/IFN- axis, 107–108 IL–1 receptor, 108 IL–17A neutralization, 113 IL–12 induction, inflammation resolution (see Toxoplasma gondii infection) immune based therapies, IL–2 effects, 157 Leishmania, IL and 6, 88 prevent immunopathology mechanism IL–22, 11–12 IL–27, 12–13 Toxoplasma gondii IL–10, IL–27 and suppression, 12–13 IL–12 induction (see Toxoplasma gondii and inflammation resolution) IL–22 role, 11–12 175 Trypanosoma cruzi IL–10, 73 IL.12p70, 73 tuberculosis, IL–10, 30 Invariant Natural Killer T (iNKT) cell activation, microbial infection bacterial infection bystander indirect activation, 50–51 cognate recognition, GSL antigens, 52–53 CD1 presentation, 49 CD1 system division, 41 formation, 41 group I (CD1a, b, c), 41 group II (CD1d), 42–44 group III, (CD1e), 44 species distribution, 44–45 function antibody production, 48 anti-microbial activity, 48 autoreactivity, 46 bacteria and virus detection, 45 circuits and molecular mechanisms, 46 deficient CD1d0/J 180 mice, 45 interactions, 47–48 mechanisms, 48 natural and immune rejection, 45 systemic administration, 46–47 Th1 or Th2 cytokines, 45 trans-activation, 47 type I diabetes, 45–46 GSL antigens and structures, 40 in human correlation, 55 Novovosphingobium/Sphingomonas spp., 55–56 primary biliary cirrhosis, 56–57 MHC class I, 40 mouse model infection, 57–58 non-bacterial infection parasitic, helminth and fungal infections, 54 viral infections, 53–54 role, 40 self-GSL antigens, 49–50 TCR, cell population, 40 V124 TCR, human, 40 V114 TCR, mice, 40 J Janssen, E.M., 121 Jones, D.E., 85–93 176 K Koch, R., 23 L Lipoxins M tuberculosis in humans, ALOX5, 29–30 5-LO-dependent, 29 risk, 30 role, 29 Toxoplasma gondii control, 14 “DC paralysis”, 13 injection, STAg, 13 mycobacterium tuberculosis, 16–17 vs Mycobacterium tuberculosis, 17 pathogen evasion, 15–16 production, 14 receptors and evidence role, 13 Loewendorf, A., 121 M Macrophages (M ), Histoplasma capsulatum CD11/CD18 adhesin receptors, 102, 103 CD8+ cytotoxic T cells, 106 cross-presentation pathway, DC, 104 granuloma, 101 growth inhibition, 103 IL–4, 110 neutrophils, 103 yeast cells, 102, 103 Major histocompatibility complex (MHC) CD1 system and NKT cells, 41 CD8+ T cells responses, HIV, 152 class I expression antigen-presentation, 128–129 glycoprotein products, 129 “immunoevasion” genes, 129–130 class II expression antigen presentation, 130 proteolytic degradation process, 130 CmvIL–10, 133–134 cytomegalovirus (CMV), 126–130 dendritic cells, 126 iNKT cell activation, 40 MCMV, 128–129 TCR interactions cross-presentation, 127 exogenous and endogenous pathway, 126–127 Matrix Metalloproteinases (MMP), 80 Index Mattner, J., 31–58 MCMV See Murine cytomegalovirus MHC See Major histocompatibility complex Mixed lymphocyte reactions (MLR), 128 M See Macrophages MLR See Mixed lymphocyte reactions Mouse model infection, 57–58 Murine cytomegalovirus (MCMV) CD8 and CD4 T cells, 124–125 m138 gene, 131 MHC class I pathway, 128–129 m157 protein, 124 N NADPH oxidase, 87, 89 Natural killer (NK) cells absence, 27 accumulation, 27 CMV DCs, 124 MCMV m157 protein, 124 depletion, 27 role, 26 Natural vs non-natural hosts, 145 Neutorphils Histoplasma capsulatum DC, 104 human defensins, 103–104 IL–4, 110 yeast cells, M , 103 tuberculosis acute pulmonary tuberculosis, 24 definition, 24 depletion, 24 mouse strains, 25 potential role, 25 protection, 24–25 Nitric oxide (NO) Leishmania, 88 Trypanosoma cruzi deficiency impact, 76 donors and control, 75 production and role, 75 response mechanisms, 75 versatile immune mediators, 75 NK cells See Natural killer cells P PDL–1 See Programmed death ligand Petersen, C.A., 85–93 Programmed death ligand (PDL–1), 132–133 Proinflammator vs anti-inflammatory, 87–88 Index S Shacklett, B.L., 141 Superoxide production, 89 T T cell receptor (TCR) cell population V124 TCR, human, 40 V114 TCR, mice, 40 Histoplasma capsulatum CD3+, 105 protective immunity, 105–106 pulmonary infection, 105 reactivation histoplasmosis, 106 interactions cross-presentation, 127 exogenous and endogenous pathway, 126–127 lymphoid cell, infection control, 105–106 T cells B7 costimulation CD28 and CTLA–4, 130–131 MCMV m138 gene, 131 positive cosignaling pathways, 131 costimulation B7 family, 127 positive and negative net signals, 128, 129 TNF receptor and soluble mediators, 127 cytokines cmvIL–10 and IDO, 133–134 HCMV, 134 production, DC, 131–132 Histoplasma capsulatum CCR5, 113, 114 CD4+ and CD8+ cells, 104–105 granuloma, 102 IL–1 receptor, 108 PD ligands, 106 promote immunity, 106 receptors, CD3+, 105 regulatory, 107 TNF- production, 105, 109 HVEM/BTLA interactions, 133 MHC expressions, 128–130 MHC-TCR interactions cross-presentation, 127 exogenous and endogenous pathway, 126–127 MLR, 128 PD–1/PDL–1 pathway B7-CD28 pathway, 132–133 177 cell cycle arrest, 132 negative cosignaling, 132 TCR See T cell receptor Th1 vs Th2, 89–90 TNF- See Tumor necrosis factor-alpha Toll-like receptors (TLRs) microbial recognition, 5–6 signaling, 51 Trypanosoma cruzi TLR, 71 TLR2 activation, 72 TLR2-/-and MyD88-/-mice, 73 TLR9 involvement, 74 Toxoplasma gondii and inflammation resolution cysts and bradyzoites, description, felines, cat, in human, IFN- , Th1 cells and microbicidal activity activation factors and parasite strains, components, immune responses mechanisms, investigation, 18 life cycle, microbial recognition and IL–12 induction biochemical signaling, CCR5 role, complexity and protection, cyclophillin–18, cytoplasmic protein profillin, hypothesis, dendritic cells, IFN- , immune response and pathogen, IRF–8, mice, macrophages, neutrophils and DCs, p38 MAP kinases, 6–7 TLRs, 5–6 transcription factors, use STAg, 4–5 natural conditions infection, parasite replication, prevent immunopathology mechanism endogenous LXA4, 15–17 IL–22, 11–12 IL–27, 12–13 inflammation redundancy and control, 14–15 interleukin–10, 9–10 lipoxin A4, 13–14 resolution phase, 8–9 TGF-B, 10–11 protozoan apicomplexa parasite, survival, oocysts, 2, 178 Toxoplasma gondii and inflammation resolution (cont.) symptoms development and risk, tachyzoites replication and “dripping” effect, transmission, Toxoplasma gondii vs Mycobacterium tuberculosis, 17 Tuberculosis, host protection and pathogen evasion BCG and treatment, 24 BCG vaccine, 33–34 disease reactivation AIDS and TNF, 32 drug and treatment, 32 effects, 33 epidemiology, 32 HIV, 32–33 granuloma formation and bacilli (see Granuloma formation and bacilli) history, 23 immune response cell wall components, 31 host cell signalling, 32 mycobacterial dormancy, 31–32 phagosome-lysosome fusion, 31 Index infection and innate immunity dendritic cells, 26 natural killer cells, 26–27 neutorphils, 24–25 regulatory T cells, 27 T cells, 25–26 risk, 24 WHO, 23–24 Tumor necrosis factor-alpha (TNF- ), Histoplasma capsulatum primary and secondary infection, 109 T cells, 106 TNF receptors, 109–110 V Vaccines BCG, 33 HIV, 33 Viral infections detection, viral pathogens, 53 Hepatitis B virus (HBV) model, 53–54 immune defense strategy, 54 iNKT role, 53 lethal outcome, Epstein-Barr virus, 53 role and SAP-Fyn signaling, 53 .. .Control of Innate and Adaptive Immune Responses during Infectious Diseases Julio Aliberti Editor Control of Innate and Adaptive Immune Responses during Infectious Diseases Editor... Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil e-mail: andre.bafica@ufsc.br J Aliberti (ed.), Control of Innate and Adaptive Immune Responses during Infectious. .. Aliberti (ed.), Control of Innate and Adaptive Immune Responses during Infectious Diseases, DOI 10.1007/978-1-4614-0484-2_1, © Springer Science+Business Media, LLC 2012 J Aliberti Ingestion of oocysts

Ngày đăng: 05/03/2014, 23:20

Từ khóa liên quan

Mục lục

  • Cover

  • Frontmatter

    • Preface

    • Contents

    • Contributors

    • Chapter 1: Resolution of Inflammation During Toxoplasma gondii Infection

      • 1.1 Introduction

      • 1.2 Experimental T. gondii Infection

        • 1.2.1 Microbial Recognition and IL-12 Induction

        • 1.2.2 IFN-γ, Th1 Cells and Microbicidal Activity

        • 1.3 Pro-resolution Strategies as a Mechanism to Prevent Immunopathology

          • 1.3.1 Resolution Phase of the Inflammatory Response

          • 1.3.2 Interleukin-10

          • 1.3.3 TGF-β

          • 1.3.4 IL-22

          • 1.3.5 IL-27

          • 1.3.6 Lipoxin A 4

          • 1.3.7 Redundancy and Control of Inflammation

          • 1.3.8 Induction of Endogenous LXA 4 as an Pathogen Evasion Pathway

          • 1.4 Conclusions

          • References

          • Chapter 2: Mechanisms of Host Protection and Pathogen Evasion of Immune Response During Tuberculosis

            • 2.1 Introduction

            • 2.2 Infection and Innate Immunity

              • 2.2.1 Neutrophils

              • 2.2.2 T Cells

Tài liệu cùng người dùng

Tài liệu liên quan