1
|
Xu Q, Li L, Zhu R. T Cell Exhaustion in Allergic Diseases and Allergen Immunotherapy: A Novel Biomarker? Curr Allergy Asthma Rep 2025; 25:18. [PMID: 40091122 DOI: 10.1007/s11882-025-01199-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW This review explores the emerging role of T cell exhaustion in allergic diseases and allergen immunotherapy (AIT). It aims to synthesize current knowledge on the mechanisms of T cell exhaustion, evaluate its potential involvement in allergic inflammation, and assess its implications as a novel biomarker for predicting and monitoring AIT efficacy. RECENT FINDINGS Recent studies highlight that T cell exhaustion, characterized by co-expression of inhibitory receptors (e.g., PD-1, CTLA-4, TIM-3), diminished cytokine production, and altered transcriptional profiles, may suppress type 2 inflammation in allergic diseases. In allergic asthma, exhausted CD4 + T cells exhibit upregulated inhibitory receptors, correlating with reduced IgE levels and airway hyperreactivity. During AIT, prolonged high-dose allergen exposure drives allergen-specific Th2 and T follicular helper (Tfh) cell exhaustion, potentially contributing to immune tolerance. Notably, clinical improvements in AIT correlate with depletion of allergen-specific Th2 cells and persistent expression of exhaustion markers (e.g., PD-1, CTLA-4) during maintenance phases. Blockade of inhibitory receptors (e.g., PD-1) enhances T cell activation, underscoring their dual regulatory role in allergy. T cell exhaustion represents a double-edged sword in allergy: it may dampen pathological inflammation in allergic diseases while serving as a mechanism for AIT-induced tolerance. The co-expression of inhibitory receptors on allergen-specific T cells emerges as a promising biomarker for AIT efficacy. Future research should clarify the transcriptional and metabolic drivers of exhaustion in allergy, validate its role across diverse allergic conditions, and optimize strategies to harness T cell exhaustion for durable immune tolerance. These insights could revolutionize therapeutic approaches and biomarker development in allergy management.
Collapse
Affiliation(s)
- Qingxiu Xu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Le Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Siedlik JA, Deckert JA, Dunbar AJ, Bhatta A, Gigliotti NM, Chan MA, Benedict SH, Bubak M, Vardiman JP, Gallagher PM. Acute high-intensity exercise enhances T cell proliferation compared to moderate-intensity exercise. Appl Physiol Nutr Metab 2025; 50:1-12. [PMID: 39947139 DOI: 10.1139/apnm-2024-0420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
Conventional belief is that high-intensity (HI) exercise inhibits immune function; however, recent work challenges this position. The purpose of this was to quantify changes in T cell proliferative capacity following either a HI or moderate-intensity (MI) exercise. Sixteen males were randomly selected to a HI or MI exercise group. Blood was obtained baseline and immediately, 1, 4, and 6 h post-exercise for analyses of CD3+ T cell proliferation (co-stimulation via phytohaemagglutinin or CD3 + CD28). The proliferative response increased in T cells in the HI group and remained significantly elevated up to 6 h post-exercise in both co-stimulation conditions. In contrast, the MI group saw no change proliferative ability following exercise. Analyses of serum stress hormones, and immunomodulatory cytokines failed to reveal any correlated variations that could clarify the T cell findings. We suggest the increase in proliferative capacity following HI exercise is indicative of an exercise-induced activation that provides for enhanced functional responses to stimuli. Moreover, this study shows that HI exercise increases T cell processes, effectively priming them for activation in response to stimuli. This study is registered with ClinicalTrials.gov (NCT06638684).
Collapse
Affiliation(s)
- Jacob A Siedlik
- Department of Biology, University of West Florida, Pensacola, FL, USA
- Florida Institute for Human and Machine Cognition, Pensacola, FL, USA
| | - Jake A Deckert
- University of Washington School of Medicine, Spokane, WA, USA
- Department of Human Physiology, Gonzaga University, Spokane, WA, USA
| | - Amanda J Dunbar
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Anuja Bhatta
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Nicole M Gigliotti
- Department of Pediatrics, Children's Mercy Hospitals & Clinics, Kansas City, MO, USA
| | - Marcia A Chan
- Department of Pediatrics, Children's Mercy Hospitals & Clinics, Kansas City, MO, USA
| | - Stephen H Benedict
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Matthew Bubak
- Department of Health, Sport, and Exercise Sciences, University of Kansas, Lawrence, KS, USA
| | - John P Vardiman
- Department of Nutrition, Kansas State University, Manhattan, KS, USA
| | - Philip M Gallagher
- Department of Health, Sport, and Exercise Sciences, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
3
|
Singh P, Bajpai P, Maheshwari D, Chawla YM, Saini K, Reddy ES, Gottimukkala K, Nayak K, Gunisetty S, Aggarwal C, Jain S, Verma C, Singla P, Soneja M, Wig N, Murali-Krishna K, Chandele A. Functional and transcriptional heterogeneity within the massively expanding HLADR +CD38 + CD8 T cell population in acute febrile dengue patients. J Virol 2023; 97:e0074623. [PMID: 37855600 PMCID: PMC10688317 DOI: 10.1128/jvi.00746-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/17/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE CD8 T cells play a crucial role in protecting against intracellular pathogens such as viruses by eliminating infected cells and releasing anti-viral cytokines such as interferon gamma (IFNγ). Consequently, there is significant interest in comprehensively characterizing CD8 T cell responses in acute dengue febrile patients. Previous studies, including our own, have demonstrated that a discrete population of CD8 T cells with HLADR+ CD38+ phenotype undergoes massive expansion during the acute febrile phase of natural dengue virus infection. Although about a third of these massively expanding HLADR+ CD38+ CD8 T cells were also CD69high when examined ex vivo, only a small fraction of them produced IFNγ upon in vitro peptide stimulation. Therefore, to better understand such functional diversity of CD8 T cells responding to dengue virus infection, it is important to know the cytokines/chemokines expressed by these peptide-stimulated HLADR+CD38+ CD8 T cells and the transcriptional profiles that distinguish the CD69+IFNγ+, CD69+IFNγ-, and CD69-IFNγ- subsets.
Collapse
Affiliation(s)
- Prabhat Singh
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Prashant Bajpai
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Deepti Maheshwari
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Yadya M. Chawla
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Keshav Saini
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Elluri Seetharami Reddy
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Kamalvishnu Gottimukkala
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Kaustuv Nayak
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sivaram Gunisetty
- Department of Pediatrics, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Charu Aggarwal
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Shweta Jain
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Chaitanya Verma
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Paras Singla
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Manish Soneja
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Naveet Wig
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Kaja Murali-Krishna
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Pediatrics, Emory University School of Medicine, Emory University, Atlanta, Georgia, USA
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
| | - Anmol Chandele
- ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
4
|
Caputo MB, Elias J, Cesar G, Alvarez MG, Laucella SA, Albareda MC. Role of the Complement System in the Modulation of T-Cell Responses in Chronic Chagas Disease. Front Cell Infect Microbiol 2022; 12:910854. [PMID: 35846776 PMCID: PMC9282465 DOI: 10.3389/fcimb.2022.910854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/26/2022] [Indexed: 01/19/2023] Open
Abstract
Chagas disease, caused by the intracellular pathogen Trypanosoma cruzi, is the parasitic disease with the greatest impact in Latin America and the most common cause of infectious myocarditis in the world. The immune system plays a central role in the control of T. cruzi infection but at the same time needs to be controlled to prevent the development of pathology in the host. It has been shown that persistent infection with T. cruzi induces exhaustion of parasite-specific T cell responses in subjects with chronic Chagas disease. The continuous inflammatory reaction due to parasite persistence in the heart also leads to necrosis and fibrosis. The complement system is a key element of the innate immune system, but recent findings have also shown that the interaction between its components and immune cell receptors might modulate several functions of the adaptive immune system. Moreover, the findings that most of immune cells can produce complement proteins and express their receptors have led to the notion that the complement system also has non canonical functions in the T cell. During human infection by T. cruzi, complement activation might play a dual role in the acute and chronic phases of Chagas disease; it is initially crucial in controlling parasitemia and might later contributes to the development of symptomatic forms of Chagas disease due to its role in T-cell regulation. Herein, we will discuss the putative role of effector complement molecules on T-cell immune exhaustion during chronic human T. cruzi infection.
Collapse
Affiliation(s)
- María Belén Caputo
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Josefina Elias
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - Gonzalo Cesar
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| | - María Gabriela Alvarez
- Chagas Section, Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - Susana Adriana Laucella
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
- Chagas Section, Hospital Interzonal General de Agudos Eva Perón, Buenos Aires, Argentina
| | - María Cecilia Albareda
- Investigation Department, Instituto Nacional de Parasitología Dr. Fatala Chaben, Buenos Aires, Argentina
| |
Collapse
|
5
|
Fenwick C, Joo V, Jacquier P, Noto A, Banga R, Perreau M, Pantaleo G. T-cell exhaustion in HIV infection. Immunol Rev 2020; 292:149-163. [PMID: 31883174 PMCID: PMC7003858 DOI: 10.1111/imr.12823] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022]
Abstract
The T‐cell response is central in the adaptive immune‐mediated elimination of pathogen‐infected and/or cancer cells. This activated T‐cell response can inflict an overwhelming degree of damage to the targeted cells, which in most instances leads to the control and elimination of foreign invaders. However, in conditions of chronic infection, persistent exposure of T cells to high levels of antigen results in a severe T‐cell dysfunctional state called exhaustion. T‐cell exhaustion leads to a suboptimal immune‐mediated control of multiple viral infections including the human immunodeficiency virus (HIV). In this review, we will discuss the role of T‐cell exhaustion in HIV disease progression, the long‐term defect of T‐cell function even in aviremic patients on antiretroviral therapy (ART), the role of exhaustion‐specific markers in maintaining a reservoir of latently infected cells, and exploiting these markers in HIV cure strategies.
Collapse
Affiliation(s)
- Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Victor Joo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Patricia Jacquier
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Riddhima Banga
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
6
|
Deng S, Sun Z, Qiao J, Liang Y, Liu L, Dong C, Shen A, Wang Y, Tang H, Fu YX, Peng H. Targeting tumors with IL-21 reshapes the tumor microenvironment by proliferating PD-1intTim-3-CD8+ T cells. JCI Insight 2020; 5:132000. [PMID: 32271164 DOI: 10.1172/jci.insight.132000] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 03/04/2020] [Indexed: 12/18/2022] Open
Abstract
The lack of sufficient functional tumor-infiltrating lymphocytes in the tumor microenvironment (TME) is one of the primary indications for the poor prognosis of patients with cancer. In this study, we developed an Erbitux-based IL-21 tumor-targeting fusion protein (Erb-IL21) to prolong the half-life and improve the antitumor efficacy of IL-21. Compared with Erb-IL2, Erb-IL21 demonstrated much lower toxicity in vivo. Mechanistically, Erb-IL21 selectively expanded functional cytotoxic T lymphocytes but not dysfunctional CD8+ T cells in the TME. We observed that the IL-21-mediated antitumor effect largely depended on the existing intratumoral CD8+ T cells, instead of newly migrated CD8+ T cells. Furthermore, Erb-IL21 overcame checkpoint blockade resistance in mice with advanced tumors. Our study reveals that Erb-IL21 can target IL-21 to tumors and maximize the antitumor potential of checkpoint blockade by expending a subset of tumor antigen-specific CD8+ T cells to achieve effective tumor control.
Collapse
Affiliation(s)
- Sisi Deng
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhichen Sun
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jian Qiao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yong Liang
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Longchao Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chunbo Dong
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Aijun Shen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yang Wang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hong Tang
- Institute Pasteur of Shanghai Chinese Academy of Sciences, Shanghai, China
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hua Peng
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Avdic U, Ahl M, Öberg M, Ekdahl CT. Immune Profile in Blood Following Non-convulsive Epileptic Seizures in Rats. Front Neurol 2019; 10:701. [PMID: 31333561 PMCID: PMC6615316 DOI: 10.3389/fneur.2019.00701] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Non-convulsive status epilepticus (NCSE) is a prolonged epileptic seizure with subtle symptoms that may delay clinical diagnosis. Emerging experimental evidence shows brain pathology and epilepsy development following NCSE. New diagnostic/prognostic tools are therefore needed for earlier and better stratification of treatment. Here we examined whether NCSE initiates a peripheral immune response in blood serum from rats that experienced electrically-induced NCSE. ELISA analysis showed an acute transient increase in serum protein levels including interleukin-6 6 h post-NCSE, similar to the immune reaction in the brain. At 4 weeks post-NCSE, when 75% of rats subjected to NCSE had also developed spontaneous seizures, several immune proteins were altered. In particular, markers associated with microglia, macrophages and antigen presenting cells, such as CD68, MHCII, and galectin-3, were increased and the T-cell marker CD4 was decreased in serum compared to both non-stimulated controls and NCSE rats without spontaneous seizures, without correlation to interictal epileptiform activity. Analyses of serum following intracerebral injection of lipopolysaccharide (LPS) showed an acute increase in interleukin-6, but at 4 weeks unaltered levels of MHCII and galectin-3, an increase in CD8 and CD11b and a decrease in CD68. None of the increased serum protein levels after NCSE or LPS could be confirmed in spleen tissue. Our data identifies the possibility to detect peripheral changes in serum protein levels following NCSE, which may be related to the development of subsequent spontaneous seizures.
Collapse
Affiliation(s)
- Una Avdic
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| | - Matilda Ahl
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| | - Maria Öberg
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| | - Christine T Ekdahl
- Inflammation and Stem Cell Therapy Group, Division of Clinical Neurophysiology, Lund University, Lund, Sweden.,Department of Clinical Sciences, Epilepsy Center, Lund University, Lund, Sweden
| |
Collapse
|
8
|
Saeidi A, Zandi K, Cheok YY, Saeidi H, Wong WF, Lee CYQ, Cheong HC, Yong YK, Larsson M, Shankar EM. T-Cell Exhaustion in Chronic Infections: Reversing the State of Exhaustion and Reinvigorating Optimal Protective Immune Responses. Front Immunol 2018; 9:2569. [PMID: 30473697 PMCID: PMC6237934 DOI: 10.3389/fimmu.2018.02569] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 10/18/2018] [Indexed: 12/31/2022] Open
Abstract
T-cell exhaustion is a phenomenon of dysfunction or physical elimination of antigen-specific T cells reported in human immunodeficiency virus (HIV), hepatitis B virus (HBV), and hepatitis C virus (HCV) infections as well as cancer. Exhaustion appears to be often restricted to CD8+ T cells responses in the literature, although CD4+ T cells have also been reported to be functionally exhausted in certain chronic infections. Although our understanding of the molecular mechanisms associated with the transcriptional regulation of T-cell exhaustion is advancing, it is imperative to also explore the central mechanisms that control the altered expression patterns. Targeting metabolic dysfunctions with mitochondrion-targeted antioxidants are also expected to improve the antiviral functions of exhausted virus-specific CD8+ T cells. In addition, it is crucial to consider the contributions of mitochondrial biogenesis on T-cell exhaustion and how mitochondrial metabolism of T cells could be targeted whilst treating chronic viral infections. Here, we review the current understanding of cardinal features of T-cell exhaustion in chronic infections, and have attempted to focus on recent discoveries, potential strategies to reverse exhaustion and reinvigorate optimal protective immune responses in the host.
Collapse
Affiliation(s)
- Alireza Saeidi
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Center of Excellence for Research in AIDS, University of Malaya, Kuala Lumpur, Malaysia
| | - Keivan Zandi
- Department of Pediatrics School of Medicine Emory University, Atlanta, GA, United States
| | - Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hamidreza Saeidi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Putra Malaysia, Selangor, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chalystha Yie Qin Lee
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yean Kong Yong
- Center of Excellence for Research in AIDS, University of Malaya, Kuala Lumpur, Malaysia.,Laboratory Center, Xiamen University Malaysia, Sepang, Malaysia
| | - Marie Larsson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Esaki Muthu Shankar
- Division of Infection Biology and Medical Microbiology, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| |
Collapse
|
9
|
Luft O, Khattar R, Farrokhi K, Ferri D, Yavorska N, Zhang J, Sadozai H, Adeyi O, Chruscinski A, Levy GA, Selzner N. Inhibition of the Fibrinogen-Like Protein 2:FcγRIIB/RIII immunosuppressive pathway enhances antiviral T-cell and B-cell responses leading to clearance of lymphocytic choriomeningitis virus clone 13. Immunology 2018; 154:476-489. [PMID: 29341118 DOI: 10.1111/imm.12897] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/31/2017] [Accepted: 01/10/2018] [Indexed: 12/26/2022] Open
Abstract
Persistent viruses evade immune detection by interfering with virus-specific innate and adaptive antiviral immune responses. Fibrinogen-like protein-2 (FGL2) is a potent effector molecule of CD4+ CD25+ FoxP3+ regulatory T cells and exerts its immunosuppressive activity following ligation to its cognate receptor, FcγRIIB/RIII. The role of FGL2 in the pathogenesis of chronic viral infection caused by lymphocytic choriomeningitis virus clone-13 (LCMV cl-13) was assessed in this study. Chronically infected fgl2+/+ mice had increased plasma levels of FGL2, with reduced expression of the maturation markers, CD80, CD86 and MHC-II on macrophages and dendritic cells and impaired production of neutralizing antibody. In contrast, fgl2-/- mice or fgl2+/+ mice that had been pre-treated with antibodies to FGL2 and FcγRIIB/RIII and then infected with LCMV cl-13 developed a robust CD4+ and CD8+ antiviral T-cell response, produced high titred neutralizing antibody to LCMV and cleared LCMV. Treatment of mice with established chronic infection with antibodies to FGL2 and FcγRIIB/RIII was shown to rescue the number and functionality of virus-specific CD4+ and CD8+ T cells with reduced total and virus-specific T-cell expression of programmed cell death protein 1 leading to viral clearance. These results demonstrate an important role for FGL2 in viral immune evasion and provide a rationale to target FGL2 to treat patients with chronic viral infection.
Collapse
Affiliation(s)
- Olga Luft
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| | - Ramzi Khattar
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| | - Kaveh Farrokhi
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Dario Ferri
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Nataliya Yavorska
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| | - Jianhua Zhang
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| | - Hassan Sadozai
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| | - Oyedele Adeyi
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| | - Andrzej Chruscinski
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| | - Gary A Levy
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Nazia Selzner
- Toronto General Hospital, Multi Organ Transplant Programme, Toronto, ON, Canada
| |
Collapse
|
10
|
Ross P, Nemec PS, Kapatos A, Miller KR, Holmes JC, Suter SE, Buntzman AS, Soderblom EJ, Collins EJ, Hess PR. The canine MHC class Ia allele DLA-88*508:01 presents diverse self- and canine distemper virus-origin peptides of varying length that have a conserved binding motif. Vet Immunol Immunopathol 2018; 197:76-86. [PMID: 29475511 DOI: 10.1016/j.vetimm.2018.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/03/2018] [Accepted: 01/12/2018] [Indexed: 01/06/2023]
Abstract
Ideally, CD8+ T-cell responses against virally infected or malignant cells are defined at the level of the specific peptide and restricting MHC class I element, a determination not yet made in the dog. To advance the discovery of canine CTL epitopes, we sought to determine whether a putative classical MHC class Ia gene, Dog Leukocyte Antigen (DLA)-88, presents peptides from a viral pathogen, canine distemper virus (CDV). To investigate this possibility, DLA-88*508:01, an allele prevalent in Golden Retrievers, was expressed as a FLAG-tagged construct in canine histiocytic cells to allow affinity purification of peptide-DLA-88 complexes and subsequent elution of bound peptides. Pattern analysis of self peptide sequences, which were determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS), permitted binding preferences to be inferred. DLA-88*508:01 binds peptides that are 9-to-12 amino acids in length, with a modest preference for 9- and 11-mers. Hydrophobic residues are favored at positions 2 and 3, as are K, R or F residues at the C-terminus. Testing motif-matched and -unmatched synthetic peptides via peptide-MHC surface stabilization assay using a DLA-88*508:01-transfected, TAP-deficient RMA-S line supported these conclusions. With CDV infection, 22 viral peptides ranging from 9-to-12 residues in length were identified in DLA-88*508:01 eluates by LC-MS/MS. Combined motif analysis and surface stabilization assay data suggested that 11 of these 22 peptides, derived from CDV hemagglutinin, large polymerase, matrix, nucleocapsid, and V proteins, were processed and presented, and thus, potential targets of anti-viral CTL in DLA-88*508:01-bearing dogs. The presentation of diverse self and viral peptides indicates that DLA-88 is a classical MHC class Ia gene.
Collapse
Affiliation(s)
- Peter Ross
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA
| | - Paige S Nemec
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA
| | - Alexander Kapatos
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA
| | - Keith R Miller
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Jennifer C Holmes
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA
| | - Steven E Suter
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA
| | - Adam S Buntzman
- Department of Immunobiology, University of Arizona, Tucson, AZ, 85724, USA
| | - Erik J Soderblom
- Proteomics Core Facility, Institute for Genome Science and Policy, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Edward J Collins
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA; Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Paul R Hess
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA.
| |
Collapse
|
11
|
Tang K, Cheng L, Zhang C, Zhang Y, Zheng X, Zhang Y, Zhuang R, Jin B, Zhang F, Ma Y. Novel Identified HLA-A*0201-Restricted Hantaan Virus Glycoprotein Cytotoxic T-Cell Epitopes Could Effectively Induce Protective Responses in HLA-A2.1/K b Transgenic Mice May Associate with the Severity of Hemorrhagic Fever with Renal Syndrome. Front Immunol 2017; 8:1797. [PMID: 29312318 PMCID: PMC5732971 DOI: 10.3389/fimmu.2017.01797] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/30/2017] [Indexed: 12/15/2022] Open
Abstract
Hantaan virus (HTNV) infections can cause severe hemorrhagic fever with renal syndrome (HFRS) in humans, which is associated with high fatality rates. Cytotoxic T cell (CTL) responses contribute to virus elimination; however, to date, HLA class I allele-restricted HTNV glycoprotein (GP) epitopes recognized by CTLs have not been reported, limiting our understanding of CTL responses against HTNV infection in humans. In this study, 34 HTNV GP nine-mer epitopes that may bind to HLA-A*0201 molecules were predicted using the BIMAS and SYFPEITHI database. Seven of the epitopes were demonstrated to bind to HLA-A*0201 molecules with high affinity via the T2 cell binding assay and were successfully used to synthesize peptide/HLA-A*0201 tetramers. The results of tetramer staining showed that the frequencies of each epitope-specific CTL were higher in patients with milder HFRS, which indicated that the epitopes may induce protective CTL responses after HTNV infection. IFN-γ-enzyme-linked immunospot analysis further confirmed the immunoreactivity of epitopes by eliciting epitope-specific IFN-γ-producing CTL responses. In an HTNV challenge trial, significant inhibition of HTNV replication characterized by lower levels of antigens and RNA loads was observed in major target organs (liver, spleen, and kidneys) of HLA-A2.1/Kb transgenic mice pre-vaccinated with nonapeptides VV9 (aa8–aa16, VMASLVWPV), SL9 (aa996–aa1004, SLTECPTFL) and LL9 (aa358–aa366, LIWTGMIDL). Importantly, LL9 exhibited the best ability to induce protective CTL responses and showed a prominent effect on the kidneys, potentially preventing kidney injury after HTNV infection. Taken together, our results highlight that HTNV GP-derived HLA-A*0201-restricted epitopes could elicit protective CTL responses against the virus, and that epitope LL9 functions as an immunodominant protective epitope that may advance the design of safe and effective CTL-based HTNV peptide vaccines for humans.
Collapse
Affiliation(s)
- Kang Tang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Linfeng Cheng
- Department of Microbiology, The Fourth Military Medical University, Xi'an, China
| | - Chunmei Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Yusi Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Xuyang Zheng
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yun Zhang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Ran Zhuang
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Boquan Jin
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| | - Fanglin Zhang
- Department of Microbiology, The Fourth Military Medical University, Xi'an, China
| | - Ying Ma
- Department of Immunology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
12
|
Bliss CM, Drammeh A, Bowyer G, Sanou GS, Jagne YJ, Ouedraogo O, Edwards NJ, Tarama C, Ouedraogo N, Ouedraogo M, Njie-Jobe J, Diarra A, Afolabi MO, Tiono AB, Yaro JB, Adetifa UJ, Hodgson SH, Anagnostou NA, Roberts R, Duncan CJA, Cortese R, Viebig NK, Leroy O, Lawrie AM, Flanagan KL, Kampmann B, Imoukhuede EB, Sirima SB, Bojang K, Hill AVS, Nébié I, Ewer KJ. Viral Vector Malaria Vaccines Induce High-Level T Cell and Antibody Responses in West African Children and Infants. Mol Ther 2017; 25:547-559. [PMID: 28153101 PMCID: PMC5368405 DOI: 10.1016/j.ymthe.2016.11.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/15/2016] [Accepted: 11/15/2016] [Indexed: 12/31/2022] Open
Abstract
Heterologous prime-boosting with viral vectors encoding the pre-erythrocytic antigen thrombospondin-related adhesion protein fused to a multiple epitope string (ME-TRAP) induces CD8+ T cell-mediated immunity to malaria sporozoite challenge in European malaria-naive and Kenyan semi-immune adults. This approach has yet to be evaluated in children and infants. We assessed this vaccine strategy among 138 Gambian and Burkinabe children in four cohorts: 2- to 6-year olds in The Gambia, 5- to 17-month-olds in Burkina Faso, and 5- to 12-month-olds and 10-week-olds in The Gambia. We assessed induction of cellular immunity, taking into account the distinctive hematological status of young infants, and characterized the antibody response to vaccination. T cell responses peaked 7 days after boosting with modified vaccinia virus Ankara (MVA), with highest responses in infants aged 10 weeks at priming. Incorporating lymphocyte count into the calculation of T cell responses facilitated a more physiologically relevant comparison of cellular immunity across different age groups. Both CD8+ and CD4+ T cells secreted cytokines. Induced antibodies were up to 20-fold higher in all groups compared with Gambian and United Kingdom (UK) adults, with comparable or higher avidity. This immunization regimen elicited strong immune responses, particularly in young infants, supporting future evaluation of efficacy in this key target age group for a malaria vaccine.
Collapse
Affiliation(s)
- Carly M Bliss
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | | | - Georgina Bowyer
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Guillaume S Sanou
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | | | - Oumarou Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | - Nick J Edwards
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Casimir Tarama
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | - Nicolas Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | - Mireille Ouedraogo
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | | | - Amidou Diarra
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | | | - Alfred B Tiono
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | - Jean Baptiste Yaro
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | | | - Susanne H Hodgson
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford OX3 7LE, UK
| | - Nicholas A Anagnostou
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford OX3 7LE, UK
| | - Rachel Roberts
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford OX3 7LE, UK
| | - Christopher J A Duncan
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford OX3 7LE, UK
| | | | - Nicola K Viebig
- European Vaccine Initiative, Universitäts Klinikum Heidelberg, Voßstr. 2, 69115 Heidelberg, Germany
| | - Odile Leroy
- European Vaccine Initiative, Universitäts Klinikum Heidelberg, Voßstr. 2, 69115 Heidelberg, Germany
| | - Alison M Lawrie
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford OX3 7LE, UK
| | | | - Beate Kampmann
- Medical Research Council Unit, Fajara, The Gambia; Department of Paediatrics, Imperial College London SW7 2AZ, UK
| | - Egeruan B Imoukhuede
- Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford OX3 7LE, UK
| | - Sodiomon B Sirima
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | | | - Adrian V S Hill
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK; Centre for Clinical Vaccinology and Tropical Medicine, The Jenner Institute, Churchill Hospital, Oxford OX3 7LE, UK
| | - Issa Nébié
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, 01 BP 2208 Ouagadougou, Burkina Faso
| | - Katie J Ewer
- The Jenner Institute Laboratories, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK.
| |
Collapse
|
13
|
Li T, Wang J, Wang Y, Chen Y, Wei H, Sun R, Tian Z. Respiratory Influenza Virus Infection Induces Memory-like Liver NK Cells in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 198:1242-1252. [PMID: 28031334 DOI: 10.4049/jimmunol.1502186] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/22/2016] [Indexed: 12/18/2022]
Abstract
Although NK cells are classified as innate immune cells, recent studies have demonstrated the transformation of NK cells into long-lived memory cells that contribute to secondary immune responses in certain mouse models. However, whether NK cells mount an Ag-specific memory response to acute influenza virus infection has not yet been examined. Here, we show that, consistent with previous studies, lung NK cells play an important role in controlling viral proliferation after primary influenza virus infection. However, although lung NK cells display a memory phenotype at the late stage of infection, these cells do not protect mice against secondary influenza virus infection. Interestingly, liver NK cells from influenza virus-infected mice possess a memory phenotype and protect mice against secondary influenza virus infection. Memory-like liver NK cells display a CD49a+DX5- phenotype, and the adoptive transfer of purified liver CD49a+DX5- NK cells into naive mice followed by viral infection results in protective immunity and decreased viral titer. Moreover, we demonstrate that primary inactivated influenza virus induces memory NK cells residing in the liver of Rag1-/- mice. Collectively, these data suggest that liver CD49a+DX5- NK cells remember encountered Ag from influenza virus after primary infection and are more protective upon subsequent infection.
Collapse
Affiliation(s)
- Tingting Li
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; and
| | - Jian Wang
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; and
| | - Yanshi Wang
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; and
| | - Yongyan Chen
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; and
| | - Haiming Wei
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; and
| | - Rui Sun
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; and .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Zhigang Tian
- The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, Anhui 230027, China; and .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
14
|
Reguzova AY, Karpenko LI, Mechetina LV, Belyakov IM. Peptide-MHC multimer-based monitoring of CD8 T-cells in HIV-1 infection and AIDS vaccine development. Expert Rev Vaccines 2014; 14:69-84. [PMID: 25373312 DOI: 10.1586/14760584.2015.962520] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of MHC multimers allows precise and direct detecting and analyzing of antigen-specific T-cell populations and provides new opportunities to characterize T-cell responses in humans and animals. MHC-multimers enable us to enumerate specific T-cells targeting to viral, tumor and vaccine antigens with exceptional sensitivity and specificity. In the field of HIV/SIV immunology, this technique provides valuable information about the frequencies of HIV- and SIV-specific CD8(+) cytotoxic T lymphocytes (CTLs) in different tissues and sites of infection, AIDS progression, and pathogenesis. Peptide-MHC multimer technology remains a very sensitive tool in detecting virus-specific T -cells for evaluation of the immunogenicity of vaccines against HIV-1 in preclinical trials. Moreover, it helps to understand how immune responses are formed following vaccination in the dynamics from priming point until T-cell memory is matured. Here we review a diversity of peptide-MHC class I multimer applications for fundamental immunological studies in different aspects of HIV/SIV infection and vaccine development.
Collapse
Affiliation(s)
- Alena Y Reguzova
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Novosibirsk region, 630559, Russia
| | | | | | | |
Collapse
|
15
|
CD4+ T cells develop antiretroviral cytotoxic activity in the absence of regulatory T cells and CD8+ T cells. J Virol 2013; 87:6306-13. [PMID: 23536666 DOI: 10.1128/jvi.00432-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Conventional CD4(+) T cells play an important role in viral immunity. In most virus infections, they provide essential help for antiviral B and T cell responses. In chronic infections, including HIV infection, an expansion of regulatory T cells (Tregs) has been demonstrated, which can suppress virus-specific CD4(+) T cell responses in vitro. However, the suppressive activity of Tregs on effector CD4(+) T cells in retroviral infection is less well documented in vivo. We took advantage of a transgenic mouse in which Tregs can be selectively depleted to determine the influence of such cells on retrovirus-specific CD4(+) T cell responses during an ongoing infection. Mice were infected with Friend retrovirus (FV), and Tregs were depleted during the acute phase of the infection. In nondepleted mice, activated CD4(+) T cells produced Th1-type cytokines but did not exhibit any antiviral cytotoxicity as determined in a major histocompatibility complex (MHC) class II-restricted in vivo cytotoxic T lymphocyte (CTL) assay. Depletion of Tregs significantly increased the numbers of virus-specific CD4(+) T cells and improved their cytokine production, whereas it induced only very little CD4(+) T cell cytotoxicity. However, after dual depletion of Tregs and CD8(+) T cells, conventional CD4(+) T cells developed significant cytotoxic activity against FV epitope-labeled target cells in vivo and contributed to the control of virus replication. Thus, both Tregs and CD8(+) T cells influence the cytotoxic activity of conventional CD4(+) T cells during an acute retroviral infection.
Collapse
|
16
|
Liu Y, McNevin JP, Holte S, McElrath MJ, Mullins JI. Dynamics of viral evolution and CTL responses in HIV-1 infection. PLoS One 2011; 6:e15639. [PMID: 21283794 PMCID: PMC3024315 DOI: 10.1371/journal.pone.0015639] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 11/18/2010] [Indexed: 02/06/2023] Open
Abstract
Improved understanding of the dynamics of host immune responses and viral evolution is critical for effective HIV-1 vaccine design. We comprehensively analyzed Cytotoxic T-lymphocyte (CTL)-viral epitope dynamics in an antiretroviral therapy-naïve subject over the first four years of HIV-1 infection. We found that CTL responses developed sequentially and required constant antigenic stimulation for maintenance. CTL responses exerting strong selective pressure emerged early and led to rapid escape, proliferated rapidly and were predominant during acute/early infection. Although CTL responses to a few persistent epitopes developed over the first two months of infection, they proliferated slowly. As CTL epitopes were replaced by mutational variants, the corresponding responses immediately declined, most rapidly in the cases of strongly selected epitopes. CTL recognition of epitope variants, via cross-reactivity and de novo responses, was common throughout the period of study. Our data demonstrate that HIV-specific CTL responses, especially in the critical acute/early stage, were focused on regions that are prone to escape. Failure of CTL responses to strongly target functional or structurally critical regions of the virus, as well as the sequential cascade of CTL responses, followed closely by viral escape and decline of the corresponding responses, likely contribute to a lack of sustainable viral suppression. Focusing early and rapidly proliferating CTL on persistent epitopes may be essential for durable viral control in HIV-1 infection.
Collapse
Affiliation(s)
- Yi Liu
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - John P. McNevin
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Sarah Holte
- Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - James I. Mullins
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
17
|
Asakura S, Hashimoto D, Takashima S, Sugiyama H, Maeda Y, Akashi K, Tanimoto M, Teshima T. Alloantigen expression on non-hematopoietic cells reduces graft-versus-leukemia effects in mice. J Clin Invest 2010; 120:2370-8. [PMID: 20530875 DOI: 10.1172/jci39165] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 04/07/2010] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is used effectively to treat a number of hematological malignancies. Its beneficial effects rely on donor-derived T cell-targeted leukemic cells, the so-called graft-versus-leukemia (GVL) effect. Induction of GVL is usually associated with concomitant development of graft-versus-host disease (GVHD), a major complication of allogeneic HSCT. The T cells that mediate GVL and GVHD are activated by alloantigen presented on host antigen-presenting cells of hematopoietic origin, and it is not well understood how alloantigen expression on non-hematopoietic cells affects GVL activity. Here we show, in mouse models of MHC-matched, minor histocompatibility antigen-mismatched bone marrow transplantation, that alloantigen expression on host epithelium drives donor T cells into apoptosis and dysfunction during GVHD, resulting in a loss of GVL activity. During GVHD, programmed death-1 (PD-1) and PD ligand-1 (PD-L1), molecules implicated in inducing T cell exhaustion, were upregulated on activated T cells and the target tissue, respectively, suggesting that the T cell defects driven by host epithelial alloantigen expression might be mediated by the PD-1/PD-L1 pathway. Consistent with this, blockade of PD-1/PD-L1 interactions partially restored T cell effector functions and improved GVL. These results elucidate a previously unrecognized significance of alloantigen expression on non-hematopoietic cells in GVL and suggest that separation of GVL from GVHD for more effective HSCT may be possible in human patients.
Collapse
Affiliation(s)
- Shoji Asakura
- Biopathological Science, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Yi JS, Cox MA, Zajac AJ. T-cell exhaustion: characteristics, causes and conversion. Immunology 2010; 129:474-81. [PMID: 20201977 PMCID: PMC2842494 DOI: 10.1111/j.1365-2567.2010.03255.x] [Citation(s) in RCA: 468] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 01/23/2010] [Accepted: 01/25/2010] [Indexed: 12/11/2022] Open
Abstract
T-cell exhaustion is characterized by the stepwise and progressive loss of T-cell functions and can culminate in the physical deletion of the responding cells. Exhaustion is well-defined during chronic lymphocytic choriomeningitis virus infection and commonly develops under conditions of antigen-persistence, which occur following many chronic infections that are of significant public health concern including hepatitis B virus, hepatitis C virus and human immunodeficiency virus infections, as well as during tumour outgrowth. Exhaustion is not a uniformly disabled setting as a gradation of phenotypic and functional defects can manifest, and these cells are distinct from prototypic effector, memory and also anergic T cells. We are gaining insights into the extrinsic and intrinsic factors that determine the severity of exhaustion. These include the duration and magnitude of antigenic activation, availability of CD4 T-cell help, the levels of stimulatory and suppressive cytokines, as well as the expression of activatory and inhibitory receptors. More information is now becoming available regarding the molecular mechanisms that attenuate the responsiveness of exhausted T cells. As the parameters that dictate exhaustion are more thoroughly defined, this is fostering the development of methods that prevent and rejuvenate functionally inferior responses. In this article we discuss our current understanding of the properties of exhausted T cells and the mechanisms that promote and maintain this state.
Collapse
Affiliation(s)
- John S Yi
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
19
|
Bertoletti A, Tan AT, Gehring AJ. HBV-Specific Adaptive Immunity. Viruses 2009; 1:91-103. [PMID: 21994540 PMCID: PMC3185487 DOI: 10.3390/v1020091] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2009] [Revised: 07/08/2009] [Accepted: 07/16/2009] [Indexed: 02/08/2023] Open
Abstract
The successful control of HBV infection requires an efficient expansion of distinct elements of the adaptive immune system (B cells, helper and cytotoxic T cells) that, due to the hepatotropic nature of HBV, need to operate in the liver parenchyma. In this respect, we will discuss broad features of HBV immunity in patients with resolved or chronic HBV infection and analyze how the liver environment can directly modulate HBV-immunity.
Collapse
Affiliation(s)
- Antonio Bertoletti
- Laboratory of Hepatic Viral Diseases, Singapore Institute for Clinical Sciences, Agency of Science Technology and Research (A*Star), 30 Medical Drive, 117609, Singapore; E-Mails: ;
- Singapore Immunology Network, Agency of Science Technology and Research (A*Star), Singapore
- Program Emerging Viral Diseases Unit, Duke-NUS Graduate Medical School, Singapore
- Author to whom correspondence should be addressed; E-mail: ; Tel.: +65 64070091; Fax: +65 67766837
| | - Anthony T. Tan
- Laboratory of Hepatic Viral Diseases, Singapore Institute for Clinical Sciences, Agency of Science Technology and Research (A*Star), 30 Medical Drive, 117609, Singapore; E-Mails: ;
| | - Adam J. Gehring
- Laboratory of Hepatic Viral Diseases, Singapore Institute for Clinical Sciences, Agency of Science Technology and Research (A*Star), 30 Medical Drive, 117609, Singapore; E-Mails: ;
| |
Collapse
|
20
|
Impact of epitope escape on PD-1 expression and CD8 T-cell exhaustion during chronic infection. J Virol 2009; 83:4386-94. [PMID: 19211743 DOI: 10.1128/jvi.02524-08] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
During some persistent viral infections, virus-specific T-cell responses wane due to the antigen-specific deletion or functional inactivation (i.e., exhaustion) of responding CD8 T cells. T-cell exhaustion often correlates with high viral load and is associated with the expression of the inhibitory receptor PD-1. In other infections, functional T cells are observed despite high levels of pathogen persistence. The reasons for these different T-cell fates during chronic viral infections are not clear. Here, we tracked the fate of virus-specific CD8 T cells in lymphocytic choriomeningitis virus (LCMV)-infected mice during viral clearance, the persistence of wild-type virus, or the selection and persistence of a viral variant that abrogates the presentation of a single epitope. Viral clearance results in PD-1(lo) functional virus-specific CD8 T cells, while the persistence of wild-type LCMV results in high PD-1 levels and T-cell exhaustion. However, following the emergence of a GP35V-->A variant virus that abrogates the presentation of the GP33 epitope, GP33-specific CD8 T cells remained functional, continued to show low levels of PD-1, and reexpressed CD127, a marker of memory T-cell differentiation. In the same animals and under identical environmental conditions, CD8 T cells recognizing nonmutated viral epitopes became physically deleted or were PD-1(hi) and nonfunctional. Thus, the upregulation of PD-1 and the functional inactivation of virus-specific T cells during chronic viral infection is dependent upon continued epitope recognition. These data suggest that optimal strategies for vaccination should induce high-magnitude broadly specific T-cell responses that prevent cytotoxic T-lymphocyte escape and highlight the need to evaluate the function of vaccine-induced T cells in the context of antigens presented during virus persistence.
Collapse
|
21
|
Casado JG, DelaRosa O, Pawelec G, Peralbo E, Duran E, Barahona F, Solana R, Tarazona R. Correlation of effector function with phenotype and cell division after in vitro differentiation of naive MART-1-specific CD8+ T cells. Int Immunol 2008; 21:53-62. [DOI: 10.1093/intimm/dxn123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
22
|
Calbo S, Delagrèverie H, Arnoult C, Authier FJ, Tron F, Boyer O. Functional tolerance of CD8+ T cells induced by muscle-specific antigen expression. THE JOURNAL OF IMMUNOLOGY 2008; 181:408-17. [PMID: 18566407 DOI: 10.4049/jimmunol.181.1.408] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Skeletal muscles account for more than 30% of the human body, yet mechanisms of immunological tolerance to this tissue remain mainly unexplored. To investigate the mechanisms of tolerance to muscle-specific proteins, we generated transgenic mice expressing the neo-autoantigen OVA exclusively in skeletal muscle (SM-OVA mice). SM-OVA mice were bred with OT-I or OT-II mice that possess a transgenic TCR specific for OVA peptides presented by MHC class I or class II, respectively. Tolerance to OVA did not involve clonal deletion, anergy or an increased regulatory T cell compartment. Rather, CD4+ T cell tolerance resulted from a mechanism of ignorance revealed by their response following OVA immunization. In marked contrast, CD8+ T cells exhibited a loss of OVA-specific cytotoxic activity associated with up-regulation of the immunoregulatory programmed death-1 molecule. Adoptive transfer experiments further showed that OVA expression in skeletal muscle was required to maintain this functional tolerance. These results establish a novel asymmetric model of immunological tolerance to muscle autoantigens involving Ag ignorance for CD4+ T cells, whereas muscle autoantigens recognized by CD8+ T cells results in blockade of their cytotoxic function. These observations may be helpful for understanding the breakage of tolerance in autoimmune muscle diseases.
Collapse
Affiliation(s)
- Sébastien Calbo
- Institut National de la Santé et de la Recherche Médicale, Unité 905, University of Rouen, and Department of Immunology, Rouen University Hospital, Rouen, France.
| | | | | | | | | | | |
Collapse
|
23
|
Ha SJ, West EE, Araki K, Smith KA, Ahmed R. Manipulating both the inhibitory and stimulatory immune system towards the success of therapeutic vaccination against chronic viral infections. Immunol Rev 2008; 223:317-33. [PMID: 18613845 DOI: 10.1111/j.1600-065x.2008.00638.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
SUMMARY One potentially promising strategy to control chronic infections such as human immunodeficiency virus, hepatitis B virus, and hepatitis C virus is therapeutic vaccination, which aims to reduce persisting virus by stimulating a patient's own antiviral immune responses. However, this approach has fallen short of expectations, because antiviral T cells generated during chronic infections often become functionally exhausted and thus do not respond properly to therapeutic vaccination. Therefore, it is necessary to develop a therapeutic vaccine strategy to more effectively boost endogenous T-cell responses to control persistent viral infections. Studies to elucidate the cause of impaired T-cell function have pointed to sustained inhibitory receptor signaling through T-cell expression of programmed death 1 (PD-1). Recently, another inhibitory molecule, cytotoxic T lymphocyte antigen 4 (CTLA-4), and also an immunosuppressive cytokine, interleukin 10 (IL-10), have been reported to be potential factors of establishing immune suppression and viral persistence. Blocking these negative signaling pathways could restore the host immune system, enabling it to respond to further stimulation. Indeed, combining therapeutic vaccination along with the blockade of inhibitory signals could synergistically enhance functional CD8(+) T-cell responses and improve viral control in chronically infected mice, providing a promising strategy for the treatment of chronic viral infections. Furthermore, not only the ablation of negative signals but also the addition of stimulatory signals, such as interleukin 2 (IL-2), might prove to be a potentially promising strategy to augment the efficacy of therapeutic vaccination against chronic viral infections.
Collapse
Affiliation(s)
- Sang-Jun Ha
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
24
|
Khanolkar A, Badovinac VP, Harty JT. CD8 T cell memory development: CD4 T cell help is appreciated. Immunol Res 2008; 39:94-104. [PMID: 17917058 DOI: 10.1007/s12026-007-0081-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/25/2022]
Abstract
An important goal of vaccination strategies is to elicit long term, effective immunity. Therefore it is imperative to define the parameters that regulate the development and preservation of the numbers and functional quality of cells that confer this property to the host. CD8 T cells are a key component of the host adaptive immune response that helps eradicate invading viruses and other cell-associated pathogens. Once the primary infection is controlled, the CD8 T cells transition from being effector cells into memory cells that act as sentinels of the immune system capable of rapidly purging the host of recurrent infections by the same pathogen. The factors that regulate and orchestrate this transition from effector CD8 T cells into functionally robust memory CD8 T cells are poorly understood. In recent years it has been determined that CD4 T cells play a vital role in the survival and functional responsiveness of memory CD8 T cells. However, the mechanism(s) of this interaction are still unclear.
Collapse
Affiliation(s)
- Aaruni Khanolkar
- Department of Microbiology, University of Iowa, 3-512 Bowen Science Building, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
25
|
Rallón NI, Soriano V, Benito JM. [Adaptive cell immune response against the hepatitis C virus infection]. Med Clin (Barc) 2007; 129:469-76. [PMID: 17953913 DOI: 10.1157/13111005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Hepatitis C virus (HCV) infects around 175 million people worldwide and is one of the leading causes of chronic liver disease. Less than one third of patients infected with HCV are able to spontaneously clear the virus during acute infection, while most patients evolve to chronic infection. Control of viral replication has been associated to the cellular component of the host immune response. It is not fully understood what distinguish a successful cellular immune response. An integral interpretation of the numerous experimental findings may allow a better understanding of the immune mechanisms involved in the inability of the immune system to successfully control chronic HCV infection.
Collapse
Affiliation(s)
- Norma Ibón Rallón
- Laboratorio de Biología Molecular, Servicio de Enfermedades Infecciosas, Hospital Carlos III, Madrid, España
| | | | | |
Collapse
|
26
|
Kemball CC, Szomolanyi-Tsuda E, Lukacher AE. Allogeneic differences in the dependence on CD4+ T-cell help for virus-specific CD8+ T-cell differentiation. J Virol 2007; 81:13743-53. [PMID: 17913814 PMCID: PMC2168883 DOI: 10.1128/jvi.01778-07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD4(+) T-cell help enables antiviral CD8(+) T cells to differentiate into fully competent memory cells and sustains CD8(+) T-cell-mediated immunity during persistent virus infection. We recently reported that mice of C57BL/6 and C3H strains differ in their dependence on CD28 and CD40L costimulation for long-term control of infection by polyoma virus, a persistent mouse pathogen. In this study, we asked whether mice of these inbred strains also vary in their requirement for CD4(+) T-cell help for generating and maintaining polyoma virus-specific CD8(+) T cells. CD4(+) T-cell-depleted C57BL/6 mice mounted a robust antiviral CD8(+) T-cell response during acute infection, whereas unhelped CD8(+) T-cell effectors in C3H mice were functionally impaired during acute infection and failed to expand upon antigenic challenge during persistent infection. Using (C57BL/6 x C3H)F(1) mice, we found that the dispensability for CD4(+) T-cell help for the H-2(b)-restricted polyoma virus-specific CD8(+) T-cell response during acute infection extends to the H-2(k)-restricted antiviral CD8(+) T cells. Our findings demonstrate that dependence on CD4(+) T-cell help for antiviral CD8(+) T-cell effector differentiation can vary among allogeneic strains of inbred mice.
Collapse
Affiliation(s)
- Christopher C Kemball
- Department of Pathology, Emory University School of Medicine, Woodruff Memorial Research Building, Room 7307, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
27
|
Tuuminen T, Kekäläinen E, Mäkelä S, Ala-Houhala I, Ennis FA, Hedman K, Mustonen J, Vaheri A, Arstila TP. Human CD8+ T cell memory generation in Puumala hantavirus infection occurs after the acute phase and is associated with boosting of EBV-specific CD8+ memory T cells. THE JOURNAL OF IMMUNOLOGY 2007; 179:1988-95. [PMID: 17641066 DOI: 10.4049/jimmunol.179.3.1988] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The induction and maintenance of T cell memory is incompletely understood, especially in humans. We have studied the T cell response and the generation of memory during acute infection by the Puumala virus (PUUV), a hantavirus endemic to Europe. It causes a self-limiting infection with no viral persistence, manifesting as hemorrhagic fever with renal syndrome. HLA tetramer staining of PBMC showed that the CD8(+) T cell response peaked at the onset of the clinical disease and decreased within the next 3 wk. Expression of activation markers on the tetramer-positive T cells was also highest during the acute phase, suggesting that the peak population consisted largely of effector cells. Despite the presence of tetramer-positive T cells expressing cytoplasmic IFN-gamma, PUUV-specific cells producing IFN-gamma in vitro were rare during the acute phase. Their frequency, as well as the expression of IL-7R alpha mRNA and surface protein, increased during a follow-up period of 6 wk and probably reflected the induction of memory T cells. Simultaneously with the PUUV-specific response, we also noted in seven of nine patients an increase in EBV-specific T cells and the transient presence of EBV DNA in three patients, indicative of viral reactivation. Our results show that in a natural human infection CD8(+) memory T cells are rare during the peak response, gradually emerging during the first weeks of convalescence. They also suggest that the boosting of unrelated memory T cells may be a common occurrence in human viral infections, which may have significant implications for the homeostasis of the memory T cell compartment.
Collapse
|
28
|
Gehring AJ, Sun D, Kennedy PTF, Nolte-'t Hoen E, Lim SG, Wasser S, Selden C, Maini MK, Davis DM, Nassal M, Bertoletti A. The level of viral antigen presented by hepatocytes influences CD8 T-cell function. J Virol 2007; 81:2940-9. [PMID: 17202217 PMCID: PMC1866017 DOI: 10.1128/jvi.02415-06] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
CD8 T cells exert their antiviral function through cytokines and lysis of infected cells. Because hepatocytes are susceptible to noncytolytic mechanisms of viral clearance, CD8 T-cell antiviral efficiency against hepatotropic viruses has been linked to their capacity to produce gamma interferon (IFN-gamma) and tumor necrosis factor alpha (TNF-alpha). On the other hand, intrahepatic cytokine production triggers the recruitment of mononuclear cells, which sustain acute and chronic liver damage. Using virus-specific CD8 T cells and human hepatocytes, we analyzed the modulation of virus-specific CD8 T-cell function after recognition peptide-pulsed or virally infected hepatocytes. We observed that hepatocyte antigen presentation was generally inefficient, and the quantity of viral antigen strongly influenced CD8 T-cell antiviral function. High levels of hepatitis B virus production induced robust IFN-gamma and TNF-alpha production in virus-specific CD8 T cells, while limiting amounts of viral antigen, both in hepatocyte-like cells and naturally infected human hepatocytes, preferentially stimulated CD8 T-cell degranulation. Our data document a mechanism where virus-specific CD8 T-cell function is influenced by the quantity of virus produced within hepatocytes.
Collapse
Affiliation(s)
- Adam J Gehring
- UCL Institute of Hepatology, Royal Free and University College Medical School, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
van Leeuwen EMM, ten Berge IJM, van Lier RAW. Induction and maintenance of CD8+ T cells specific for persistent viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 590:121-37. [PMID: 17191382 DOI: 10.1007/978-0-387-34814-8_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Ester M M van Leeuwen
- Dept. of Experimental Immunology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | | | | |
Collapse
|
30
|
Mescher MF, Curtsinger JM, Agarwal P, Casey KA, Gerner M, Hammerbeck CD, Popescu F, Xiao Z. Signals required for programming effector and memory development by CD8+ T cells. Immunol Rev 2006; 211:81-92. [PMID: 16824119 DOI: 10.1111/j.0105-2896.2006.00382.x] [Citation(s) in RCA: 462] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stimulation of naïve CD8+ T cells with antigen and costimulation results in proliferation and weak clonal expansion, but the cells fail to develop effector functions and are tolerant long term. Initiation of the program leading to the strong expansion and development of effector functions and memory requires a third signal that can be provided by interleukin-12 (IL-12) or interferon-alpha (IFN-alpha). CD4+ T cells condition dendritic cells (DCs) to effectively present antigen to CD8+ T cells, and this conditioning involves, at least in part, CD40-dependent upregulation of the production of these signal 3 cytokines by the DCs. Upon being fully activated, the cytotoxic T lymphocytes develop activation-induced non-responsiveness (AINR), a form of split anergy characterized by an inability to produce IL-2 to support continued expansion. If antigen remains present, IL-2 provided by CD4+ T cells can reverse AINR to allow further expansion of the effector population and conversion to responsive memory cells following antigen clearance. If IL-2 or potentially other proliferative signals are not available, persistent antigen holds cells in the AINR state and prevents the development of a responsive memory population. Thus, in addition to antigen and costimulation, CD8+ T cells require cytokine signals at distinct stages of the response to be programmed for optimal generation of effector and memory populations.
Collapse
Affiliation(s)
- Matthew F Mescher
- Center for Immunology and Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
McCoy L, Tsunoda I, Fujinami RS. Multiple sclerosis and virus induced immune responses: autoimmunity can be primed by molecular mimicry and augmented by bystander activation. Autoimmunity 2006; 39:9-19. [PMID: 16455578 DOI: 10.1080/08916930500484799] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polymicrobial infections have been associated with plausible immune mediated diseases, including multiple sclerosis (MS). Virus infection can prime autoimmune T cells specific for central nervous system (CNS) antigens, if virus has molecular mimicry with CNS proteins. On the other hand, infection of irrelevant viruses will induce two types of cytokine responses. Infection with a virus such as lymphocytic choriomeningitis virus (LCMV), can induce interferon (IFN)-alpha/beta production and suppress autoimmunity, while infection with a virus, such as murine cytomegalovirus (MCMV), can activate natural killer (NK), NKT and dendritic cells, resulting in interleukin (IL)-12 and IFN-gamma production. These cytokines can cause bystander activation of autoreactive T cells. We established an animal model, where mice infected with vaccinia virus encoding myelin protein can mount autoimmune responses. However, the mice develop clinical disease only after irrelevant immune activation either with complete Freund's adjuvant or MCMV infection. In this review, we propose that a combination of two mechanisms, molecular mimicry and bystander activation, induced by virus infection, can lead to CNS demyelinating diseases, including MS. Viral proteins having molecular mimicry with self-proteins in the CNS can prime genetically susceptible individuals. Once this priming has occurred, an immunologic challenge could result in disease through bystander activation by cytokines.
Collapse
Affiliation(s)
- Lori McCoy
- University of Utah School of Medicine, Department of Neurology, 30 North 1900 East, Room 3R330, Salt Lake City, UT 84132-2305, USA
| | | | | |
Collapse
|
32
|
Bonilla WV, Geuking MB, Aichele P, Ludewig B, Hengartner H, Zinkernagel RM. Microchimerism maintains deletion of the donor cell-specific CD8+ T cell repertoire. J Clin Invest 2006; 116:156-62. [PMID: 16395404 PMCID: PMC1323267 DOI: 10.1172/jci26565] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2005] [Accepted: 10/25/2005] [Indexed: 01/16/2023] Open
Abstract
Rare cases of stable allograft acceptance after discontinuation of immunosuppression are often accompanied by macrochimerism (> 1% donor cells in blood) or microchimerism (< 1% donor cells in blood). Here, we have investigated whether persistence of donor cells is the cause or the consequence of long-lasting CTL unresponsiveness. We found that engraftment of splenocytes bearing a single foreign MHC class I-restricted epitope resulted in lifelong donor cell microchimerism and specific CTL unresponsiveness. This status was reversed in a strictly time- and thymus-dependent fashion when the engrafted cells were experimentally removed. The results presented herein show that microchimerism actively maintains CTL unresponsiveness toward a minor histocompatibility antigen by deleting the specific repertoire and thus excluding dominant, T cell extrinsic mechanisms of CTL unresponsiveness independent of systemically persisting donor cell antigen.
Collapse
Affiliation(s)
- Weldy V Bonilla
- Institute of Experimental Immunology, Department of Pathology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
33
|
Miller NE, Bonczyk JR, Nakayama Y, Suresh M. Role of thymic output in regulating CD8 T-cell homeostasis during acute and chronic viral infection. J Virol 2005; 79:9419-29. [PMID: 16014905 PMCID: PMC1181607 DOI: 10.1128/jvi.79.15.9419-9429.2005] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although it is well documented that CD8 T cells play a critical role in controlling chronic viral infections, the mechanisms underlying the regulation of CD8 T-cell responses are not well understood. Using the mouse model of an acute and chronic lymphocytic choriomeningitis virus (LCMV) infection, we have examined the relative importance of peripheral T cells and thymic emigrants in the elicitation and maintenance of CD8 T-cell responses. Virus-specific CD8 T-cell responses were compared between mice that were either sham thymectomized or thymectomized (Thx) at approximately 6 weeks of age. In an acute LCMV infection, thymic deficiency did not affect either the primary expansion of CD8 T cells or the proliferative renewal and maintenance of virus-specific lymphoid and nonlymphoid memory CD8 T cells. Following a chronic LCMV infection, in Thx mice, although the initial expansion of CD8 T cells was normal, the contraction phase of the CD8 T-cell response was exaggerated, which led to a transient but striking CD8 T-cell deficit on day 30 postinfection. However, the virus-specific CD8 T-cell response in Thx mice rebounded quickly and was maintained at normal levels thereafter, which indicated that the peripheral T-cell repertoire is quite robust and capable of sustaining an effective CD8 T-cell response in the absence of thymic output during a chronic LCMV infection. Taken together, these findings should further our understanding of the regulation of CD8 T-cell homeostasis in acute and chronic viral infections and might have implications in the development of immunotherapy.
Collapse
Affiliation(s)
- Nicole E Miller
- Department of Pathobiological Sciences, University of Wisconsin--Madison, 53706, USA
| | | | | | | |
Collapse
|
34
|
Kobayashi K, Ishii M, Shiina M, Ueno Y, Kondo Y, Kanno A, Miyazaki Y, Yamamoto T, Kobayashi T, Niitsuma H, Kikumoto Y, Takizawa H, Shimosegawa T. Interferon-gamma is produced by CD8 T cells in response to HLA-A24-restricted hepatitis C virus epitopes after sustained virus loss. Clin Exp Immunol 2005; 141:81-88. [PMID: 15958073 PMCID: PMC1809409 DOI: 10.1111/j.1365-2249.2005.02018.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2005] [Indexed: 11/29/2022] Open
Abstract
Differences in cytotoxic T lymphocyte activity in hepatitis C virus infection may account for the outcome of interferon monotherapy. To investigate this hypothesis, we analysed the response of peripheral CD8(+) T cells that recognized epitopes presented by HLA-A*2402. We synthesized HLA/beta2-microglobulin/peptide complexes using two epitopes. Production of interferon-gamma by CD8(+) T cells in response to plastic-bound monomeric HLA/peptide complex was observed frequently in sustained virus responders (SVR) (n = 13) against all the peptides, NS31296-1304 (the percentage of responding patients, 61.5%) and core 129-137 (53.8%), while no interferon-gamma production was observed in non-responders (NR) (n = 13) for any of the peptides. Tetramer-staining showed the presence of CD8(+) T cells specific for all the peptides except NS31296-1304 in two SVR at the end of interferon monotherapy, although hardly any such cells were found in four NR. Specific killing was observed against peptides NS31296-1304 (3/4) and core 129-137 (1/4) in sustained responders but none in non-responders. These results suggest that the responses of cytotoxic T lymphocytes (CTLs) were induced during interferon therapy in these patients and that interferon-gamma production by CD8(+) T lymphocytes against HCV NS31296-1304 and core 129-137 are well maintained in patients with SVR compared with those with NR. These findings emphasize the importance of the CD8(+) T cell response in controlling HCV infection.
Collapse
Affiliation(s)
- K Kobayashi
- Tohoku University School of Health Sciences and Comprehensive Research and Education Center for Planning of Drug Development and Clinical Evaluation, Sendai, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhou S, Kurt-Jones EA, Mandell L, Cerny A, Chan M, Golenbock DT, Finberg RW. MyD88 is critical for the development of innate and adaptive immunity during acute lymphocytic choriomeningitis virus infection. Eur J Immunol 2005; 35:822-30. [PMID: 15724245 DOI: 10.1002/eji.200425730] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We investigated the roles of Toll-like receptor 2 (TLR2) and myeloid differentiation factor 88 (MyD88) in the course of a lymphocytic choriomeningitis virus (LCMV) infection and revealed the following: (i) studies of transfected cells and murine peritoneal macrophages demonstrated that TLR2 and MyD88 are essential for the initial pro-inflammatory cytokine response (human IL-8, mouse IL-6) to LCMV; (ii) TLR2 knockout (KO) mice and MyD88 KO mice challenged with LCMV produced less IL-6 and monocyte chemotactic protein-1 in the serum than wild-type mice; (iii) in contrast to inflammatory cytokines, the production of type 1 IFN (IFN-alpha) in response to LCMV was MyD88 independent; (iv) MyD88 plays an essential role in antiviral CD8(+) T cell responses, CD8(+) T cells in MyD88 KO mice were defective in their expression of intracellular antiviral cytokines; and (v) the failure of MyD88 KO mice to activate CD8(+) T cells was accompanied by persistent viral infection in MyD88 KO mice. We demonstrate that TLR-mediated responses are important in the innate immune response to LCMV and that MyD88 is essential for the control of the LCMV infection and the maturation/activation of virus-specific CD8(+) T cells.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Antigens, Differentiation/genetics
- Antigens, Differentiation/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Cytokines/immunology
- Cytokines/metabolism
- Humans
- Immunity, Innate
- Lymphocyte Activation/immunology
- Lymphocytic Choriomeningitis/immunology
- Lymphocytic choriomeningitis virus/immunology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/virology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Knockout
- Myeloid Differentiation Factor 88
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Toll-Like Receptor 2
- Toll-Like Receptors
- Transfection
Collapse
Affiliation(s)
- Shenghua Zhou
- Department of Medicine, University of Massachusetts Medical Center, Worcester, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Gray PM, Arimilli S, Palmer EM, Parks GD, Alexander-Miller MA. Altered function in CD8+ T cells following paramyxovirus infection of the respiratory tract. J Virol 2005; 79:3339-49. [PMID: 15731228 PMCID: PMC1075682 DOI: 10.1128/jvi.79.6.3339-3349.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
For many respiratory pathogens, CD8+ T cells have been shown to play a critical role in clearance. However, there are still many unanswered questions with regard to the factors that promote the most efficacious immune response and the potential for immunoregulation of effector cells at the local site of infection. We have used infection of the respiratory tract with the model paramyxovirus simian virus 5 (SV5) to study CD8+ T-cell responses in the lung. For the present study, we report that over time a population of nonresponsive, virus-specific CD8+ T cells emerged in the lung, culminating in a lack of function in approximately 85% of cells specific for the immunodominant epitope from the viral matrix (M) protein by day 40 postinfection. Concurrent with the induction of nonresponsiveness, virus-specific cells that retained function at later times postinfection exhibited an increased requirement for CD8 engagement. This change was coupled with a nearly complete loss of functional phosphoprotein-specific cells, a response previously shown to be almost exclusively CD8 independent. These studies add to the growing evidence for immune dysregulation following viral infection of the respiratory tract.
Collapse
Affiliation(s)
- Peter M Gray
- Department of Microbiology & Immunology, Room 5108, Gray Building, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
37
|
Meraldi V, Romero JF, Kensil C, Corradin G. A strong CD8+ T cell response is elicited using the synthetic polypeptide from the C-terminus of the circumsporozoite protein of Plasmodium berghei together with the adjuvant QS-21: quantitative and phenotypic comparison with the vaccine model of irradiated sporozoites. Vaccine 2005; 23:2801-12. [PMID: 15780728 DOI: 10.1016/j.vaccine.2004.10.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2004] [Revised: 09/06/2004] [Accepted: 10/25/2004] [Indexed: 10/26/2022]
Abstract
Stable protective immunity can be achieved against malaria by the injection of radiation-attenuated sporozoites (gamma-spz) and is mediated by IFN-gamma producing CD8+ T cells targeting the pre-erythrocytic stages. An efficient malaria vaccine should mimic this immunity. We compared the immune response specific for the circumsporozoite protein (CSP) of Plasmodium berghei (P. berghei), an important target of this protective response, elicited in mice immunized with the long synthetic polypeptide (LSP) PbCS 242-310, representing the C-terminus of the CSP of P. berghei, with the adjuvant QS-21 or injected with gamma-spz. The ex vivo evaluation of the CD8+ T cell response by IFN-gamma ELISPOT assay revealed that the injection of LSP with QS-21 induced, compared to gamma-spz, a similar frequency of peptide-specific lymphocytes in the spleen but a eight-fold increase in the draining lymph-nodes. A very high frequency of CD8+ T cells, specific for the sequence PbCS 245-253, a H-2Kd-restricted CTL epitope, was obtained in the liver and spleen of mice immunized with the two regimens. Even though the frequency of H-2Kd PbCS 245-253 multimer+, CD8+ T cells was higher in gamma-spz immunized mice, the frequency of IFN-gamma producing CD8+ T cells was comparable. The phenotype of the CD8+ T cell responses was characterized with the help H-2Kd PbCS 245-253 multimer and most of the CSP-specific CD8+ T cells represented an intermediate subset between effector and central memory with CD44(high), CD45RB(high), CD62L(low) and CD122(high). The number of memory CD8+ T cells decreased after the last LSP immunization but could be boosted to higher level with live spz. The unique combination of LSP PbCS 242-310 and the adjuvant QS-21 induced an immune response that was comparable in terms of quality to the one generated with gamma-spz. This confirmed the potential of LSP as malaria vaccine candidates as well as for the study of the repertoire of targets of protective immunity in the gamma-spz vaccine model.
Collapse
Affiliation(s)
- Valentin Meraldi
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | | | | | | |
Collapse
|
38
|
Abstract
Polyomaviruses establish persistent infection in a variety of hosts, including humans, where they pose an oncogenic threat under conditions of depressed immune function. Control of persistent infection by these DNA tumor viruses requires continuous immunosurveillance by functionally competent antiviral CD8+ T cells. Repetitive antigen encounter by these T cells, however, often leads to their deletion or inactivation. Elucidation of the in vivo mechanisms that sustain antigen-specific CD8+ T-cell effector activity in the face of persistent antigen is essential for devising immunotherapeutic strategies against viral oncogenesis.
Collapse
Affiliation(s)
- Aron E Lukacher
- Department of Pathology, Woodruff Memorial Research Building, Rm. 7307, Emory University School of Medicine, 1639 Pierce Drive, Atlanta, GA 30322, USA.
| |
Collapse
|
39
|
Missale G, Cariani E, Ferrari C. Role of viral and host factors in HCV persistence: which lesson for therapeutic and preventive strategies? Dig Liver Dis 2004; 36:703-11. [PMID: 15570998 DOI: 10.1016/j.dld.2004.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Several lines of evidence support the view that hepatitis C virus is not directly cytopathic for infected host cells and that the immune response plays a central role in the pathogenesis of liver damage. Innate and adaptive immune responses are induced in most individuals infected with hepatitis C virus but are insufficient to eliminate the virus. The mechanisms responsible for this failure are largely unknown but the kinetics of hepatitis C virus replication relative to the priming of the adaptive responses may exert a profound influence on the balance between virus and host. Immediately after hepatitis C virus infection, the virus replicates efficiently, inducing the production of type I interferons. However, the rapid increase in viral replication seems to be ignored by the adaptive immune response, and after a short interval from exposure, viral load can reach levels comparable to those of patients with established persistent infection. The CD8-mediated response shows functional defects, with impaired production of interferon-gamma, low perforin content, decreased capacity of expansion and lysis of target cells. Late appearance and functional defects of T cells in hepatitis C virus infection might be the result of the rapid increase of the viral load that could create the conditions for exhaustion of the adaptive response or reflect an insufficient function of the innate immune response. This possibility is suggested by in vitro studies showing that hepatitis C virus gene products can interfere with the anti-viral activity of type I interferons and natural killer cells as well as with the maturation of dendritic cells. While T-cell defects are reversed in a minority of infected individuals who succeed in controlling the infection, the T-cell impairment becomes progressively more profound as infection progresses to chronicity. In this situation, therapeutic restoration of adaptive responses may represent a rational strategy to obtain resolution of infection and to complement available therapies. The peculiar kinetics of hepatitis C virus replication and T-cell induction soon after infection may have important implications also for the design of protective vaccines since memory responses may not be able to precede the early peak of viral replication. Therefore, vaccines against hepatitis C virus may be unable to prevent infection but may rather be effective in facilitating a self-limited evolution of infection.
Collapse
Affiliation(s)
- G Missale
- Division of Infectious Diseases and Hepatology, University of Parma, via Gramsci 14, 43100 Parma, Italy
| | | | | |
Collapse
|
40
|
Kondo Y, Asabe S, Kobayashi K, Shiina M, Niitsuma H, Ueno Y, Kobayashi T, Shimosegawa T. Recovery of functional cytotoxic T lymphocytes during lamivudine therapy by acquiring multi-specificity. J Med Virol 2004; 74:425-433. [PMID: 15368520 DOI: 10.1002/jmv.20194] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
To characterize cytotoxic T lymphocytes (CTLs) that appeared in circulation during lamivudine therapy, we analyzed HBV-specific CTLs using HLA-A24 tetramer and HBcAg-specific Th1 cells in patients receiving lamivudine therapy. Six patients (HLA-A24(+)) with chronic hepatitis B, six patients (HLA-A24(-)) with chronic hepatitis B, and six patients (HLA-A24(+)) with chronic hepatitis C were studied. In addition to known CTL epitopes (C117 and P756), three epitopes were confirmed as CTL epitopes (C23, S89, S226) by chromium release assay and by staining intracellular perforin. CTLs specific for P756 were most frequently found at pre-treatment. During lamivudine therapy, increase in the frequencies of HLA-tetramer(+) cells was found for C117, S89, and S226. Recovery of CTLs was observed earlier in patients with HBeAg(-)/anti-HBe(+) compared with those with HBeAg(+)/anti-HBe(-). HBcAg-specific Th1 cells did not increase significantly up to 8 weeks. T cell lines from patients with chronic hepatitis B had a lower level of proliferation (0- to 24.9-fold expansion by in vitro stimulation) and a higher ability to produce interferon-gamma (0-84% except for S89), while perforin-positive cells showed low frequencies (0-50% except for S89). In conclusion, these results suggest that lamivudine therapy induces mainly CTLs that were less frequent before the therapy. Since recovered CTLs maintained the ability to produce interferon-gamma in response to peptides, these CTLs apparently contribute to the efficacy of lamivudine therapy in patients with hepatitis B.
Collapse
Affiliation(s)
- Yasuteru Kondo
- Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Homann D, McGavern DB, Oldstone MBA. Visualizing the viral burden: phenotypic and functional alterations of T cells and APCs during persistent infection. THE JOURNAL OF IMMUNOLOGY 2004; 172:6239-50. [PMID: 15128812 DOI: 10.4049/jimmunol.172.10.6239] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Persistent viral infections continue to present major public health problems. Failure to achieve virus control confronts the immune system with a chronic viral burden that may involve immune cells themselves and directly compromise the functionality of effector lymphocytes and APCs. In this study we use the lymphocytic choriomeningitis virus system for persistent viral infection of its natural murine host and use analytical techniques for direct ex vivo visualization of virus-infected immune cells. We report that virtually all cells of the immune system can be infected, but the distribution of the viral burden is differentially allocated to lymphocyte and APC subsets of defined phenotypes. Importantly, the profile of immune cell infection found in the blood is broadly representative for the pattern of cellular infection in most organs and is independent of the presence of Abs or complement. By direct comparison of virus-infected and uninfected cell subsets, we demonstrate that lymphocytic choriomeningitis virus-infected T cells show preferential activation, skewed cytokine profiles, and increased apoptosis. In contrast, increased activation of APCs is generalized and independent of the presence of viral Ag. Our data indicate that specific patterns of immune cell infection are associated with distinct forms of immunostimulatory and immunosuppressive alterations that may provide insights into autoimmune processes associated with infectious disease and offer clues for therapeutic interventions aimed at restoration of complete immunity.
Collapse
Affiliation(s)
- Dirk Homann
- Department of Neuropharmacology, Division of Virology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | |
Collapse
|
42
|
Affiliation(s)
- Michael J Bevan
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, 1959 NE Pacific Street, Seattle, Washington 98195-7650 USA.
| |
Collapse
|
43
|
Fuller MJ, Khanolkar A, Tebo AE, Zajac AJ. Maintenance, loss, and resurgence of T cell responses during acute, protracted, and chronic viral infections. THE JOURNAL OF IMMUNOLOGY 2004; 172:4204-14. [PMID: 15034033 DOI: 10.4049/jimmunol.172.7.4204] [Citation(s) in RCA: 182] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The acute phase of many viral infections is associated with the induction of a pronounced CD8 T cell response which plays a principle role in clearing the infection. By contrast, certain infections are not as readily controlled. In this study, we have used the well-defined system of lymphocytic choriomeningitis virus (LCMV) infection of mice to determine quantitative and qualitative changes in virus-specific CD8 T cell responses that rapidly resolve acute infections, more slowly control protracted infections, or fail to clear chronic infections. Acute LCMV infection elicits potent, functional, multi-epitope-specific CD8 T cell responses. Virus-specific CD8 T cells also expand, albeit to a lesser extent, during protracted LCMV infection. Under these conditions, there is a progressive diminution in the capacity to produce IL-2, TNF-alpha, and IFN-gamma. Changes in cytotoxic activities are also detectable but differ depending upon the specificity of the responding cells. As the infection is slowly resolved, a resurgence of cytokine production by virus-specific CD8 T cells is observed. CD4-deficient mice cannot control infection with certain strains of LCMV, but do mount multi-epitope-specific CD8 T cell responses that also lose effector capabilities; however, they are not maintained indefinitely in an unresponsive state as these cells become deleted over time. Overall, our findings suggest that constant high viral loads result in the progressive diminution of T cell effector functions and subsequent physical loss of the responding cells, whereas if the viral load is brought under control a partial restoration of CD8 T cell functions can occur.
Collapse
Affiliation(s)
- Michael J Fuller
- Department of Microbiology, University of Alabama, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|
44
|
Zhang JB, Chen SY, Yang ZQ, Li TR, Chen A, Wu YZ. Comprehensive analysis of the quantity of epitope-specific cytotoxic T lymphocytes in chronic viral hepatitis B infection. Shijie Huaren Xiaohua Zazhi 2004; 12:1069-1072. [DOI: 10.11569/wcjd.v12.i5.1069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the function state of epitope-specific cytotoxic T lymphocytes (CTLs) in chronic hepatitis B infection
METHODS: The study was performed to quantify the HBV specific CTL directly in vitro by HLA-A2 tetrameric complexes for core 18-27 (Tc 18-27), envelope 183-191 (Te 183-191), envelope 335-343 (Te 335-343), and polymerase 575-583 (Tp 575-583) in active chronic hepatitis patients, and then the correlation of HBV epitope-specific CTL between serum HBV DNA loads or alanine aminotransmerase (ALT) levels were analyzed by multiple regression analysis.
RESULTS: It was found that there were multiple CTLs responses in active chronic hepatitis patients. The frequency of Tc18-27 response was higher than the other three epitope-specific CTLs. No significant correlation was found either between the frequency of HBV specific CD8+ T cells and the viral load, or the frequency of HBV specific CD8+ T cells and the levels of alanine transaminase.
CONCLUSION: The frequencies of HBV-specific T cells are not determinant of immune-mediated protection in HBV infection and the existence of epitope-specific HBV CTLs is not directly correlated to hepatocytic injury.
Collapse
|
45
|
Zhou S, Ou R, Huang L, Price GE, Moskophidis D. Differential tissue-specific regulation of antiviral CD8+ T-cell immune responses during chronic viral infection. J Virol 2004; 78:3578-600. [PMID: 15016881 PMCID: PMC371037 DOI: 10.1128/jvi.78.7.3578-3600.2004] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The hallmarks of the immune response to viral infections are the expansion of antigen-specific CD8(+) cytotoxic T lymphocytes (CTLs) after they encounter antigen-presenting cells in the lymphoid tissues and their subsequent redistribution to nonlymphoid tissues to deal with the pathogen. Control mechanisms exist within CTL activation pathways to prevent inappropriate CTL responses against disseminating infections with a broad distribution of pathogen in host tissues. This is demonstrated during overwhelming infection with the noncytolytic murine lymphocytic choriomeningitis virus, in which clonal exhaustion (anergy and/or deletion) of CTLs prevents immune-mediated pathology but allows persistence of the virus. The mechanism by which the immune system determines whether or not to mount a full response to such infections is unknown. Here we present data showing that the initial encounter of specific CTLs with infected cells in lymphoid tissues is critical for this decision. Whether the course of the viral infection is acute or persistent for life primarily depends on the degree and kinetics of CTL exhaustion in infected lymphoid tissues. Virus-driven CTL expansion in lymphoid tissues resulted in the migration of large quantities of CTLs to nonlymphoid tissues, where they persisted at stable levels. Surprisingly, although virus-specific CTLs were rapidly clonally exhausted in lymphoid tissues under conditions of chronic infection, a substantial number of them migrated to nonlymphoid tissues, where they retained an effector phenotype for a long time. However, these cells were unable to control the infection and progressively lost their antiviral capacities (cytotoxicity and cytokine secretion) in a hierarchical manner before their eventual physical elimination. These results illustrate the differential tissue-specific regulation of antiviral T-cell responses during chronic infections and may help us to understand the dynamic relationship between antigen and T-cell populations in many persistent infections in humans.
Collapse
Affiliation(s)
- Shenghua Zhou
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | | | |
Collapse
|
46
|
Khanolkar A, Fuller MJ, Zajac AJ. CD4 T Cell-Dependent CD8 T Cell Maturation. THE JOURNAL OF IMMUNOLOGY 2004; 172:2834-44. [PMID: 14978084 DOI: 10.4049/jimmunol.172.5.2834] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have investigated the contribution of CD4 T cells to the optimal priming of functionally robust memory CD8 T cell subsets. Intranasal infection of CD4 T cell-deficient (CD4(-/-)) mice with lymphocytic choriomeningitis virus resulted in the elaboration of virus-specific CD8 T cell responses that cleared the infection. However, by comparison with normal mice, the virus-specific CD8 T cells in CD4(-/-) mice were quantitatively and qualitatively different. In normal mice, lymphocytic choriomeningitis virus-specific memory CD8 T cells are CD44(high), many are CD122(high), and a majority of these cells regain expression of CD62L overtime. These cells produce IFN-gamma and TNF-alpha, and a subset also produces IL-2. In the absence of CD4 T cell help, a distinct subset of memory CD8 T cells develops that remains CD62L(low) up to 1 year after infection and exhibits a CD44(int)CD122(low) phenotype. These cells are qualitatively different from their counterparts in normal hosts, as their capacity to produce TNF-alpha and IL-2 is diminished. In addition, although CD4-independent CD8 T cells can contain the infection following secondary viral challenge, their ability to expand is impaired. These findings suggest that CD4 T cell responses not only contribute to the optimal priming of CD8 T cells in chronically infected hosts, but are also critical for the phenotypic and functional maturation of CD8 T cell responses to Ags that are more rapidly cleared. Moreover, these data imply that the development of CD62L(high) central memory CD8 T cells is arrested in the absence of CD4 T cell help.
Collapse
Affiliation(s)
- Aaruni Khanolkar
- Department of Microbiology, University of Alabama, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
47
|
Blais ME, Gérard G, Martinic MM, Roy-Proulx G, Zinkernagel RM, Perreault C. Do thymically and strictly extrathymically developing T cells generate similar immune responses? Blood 2003; 103:3102-10. [PMID: 15070691 DOI: 10.1182/blood-2003-09-3311] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
If present in sufficient numbers, could extrathymic T cells substitute for thymus-derived T cells? To address this issue, we studied extrathymic T cells that develop in athymic mice under the influence of oncostatin M (OM). In this model, extensive T-cell development is probably due to amplification of a minor pathway of T-cell differentiation taking place only in the lymph nodes. Extrathymic CD4 T cells expanded poorly and were deficient in providing B-cell help after infection with vesicular stomatitis virus (VSV) and lymphocytic choriomeningitis virus (LCMV). Compared with classic T cells, stimulated extrathymic CD8 T cells produced copious amounts of interferon gamma (IFN-gamma), and their expansion was precocious but of limited amplitude because of a high apoptosis rate. Consequently, although extrathymic cytotoxic T lymphocytes (CTLs) responded to LCMV infection, as evidenced by the expansion of GP33-41 tetramer-positive CD8 T cells, they were unable to eradicate the virus. Our data indicate that the site of development impinges on T-cell quality and function and that extrathymic T cells functionally cannot substitute for classical thymic T cells.
Collapse
Affiliation(s)
- Marie-Eve Blais
- Guy-Bernier Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Stamou P, de Jersey J, Carmignac D, Mamalaki C, Kioussis D, Stockinger B. Chronic exposure to low levels of antigen in the periphery causes reversible functional impairment correlating with changes in CD5 levels in monoclonal CD8 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:1278-84. [PMID: 12874216 DOI: 10.4049/jimmunol.171.3.1278] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This study describes a double-transgenic model in which monoclonal CD8 F5 T cells are chronically exposed to self Ag (nucleoprotein) in the periphery, but are not affected during thymic development. Chronic exposure of CD8 T cells to their cognate Ag rendered them unable to proliferate or produce cytokines in response to antigenic stimulation in vitro. However, the cells still retained some killer function in vivo and continuously eliminated APC expressing high levels of Ag. In addition, when crossed with mice expressing Ag in the anterior pituitary gland (triple-transgenic mice), F5 T cells migrated to this site and killed growth hormone producing somatotrophs. The anergic state was reversible upon transfer into Ag-free recipients, resulting in full recovery of in vitro responsiveness to Ag. Anergic CD8 T cells express higher levels of CD5, a negative regulator of T cell signaling, whereas after transfer and residence in Ag-free hosts, CD5 levels returned to normal. This suggests that up-regulation of negative T cell regulators in peripheral T cells exposed to chronic stimulation by Ag may prevent full functionality and thus avoid overt autoreactivity.
Collapse
Affiliation(s)
- Panagiota Stamou
- Division of Molecular Immunology, The National Institute for Medical Research, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
49
|
Chakrabarti S, Milligan DW, Pillay D, Mackinnon S, Holder K, Kaur N, McDonald D, Fegan CD, Waldmann H, Hale G, Rickinson A, Steven N. Reconstitution of the Epstein-Barr virus-specific cytotoxic T-lymphocyte response following T-cell-depleted myeloablative and nonmyeloablative allogeneic stem cell transplantation. Blood 2003; 102:839-42. [PMID: 12869487 DOI: 10.1182/blood.v102.3.839] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The recovery of circulating antigen-specific T-cell immunity to Epstein-Barr virus (EBV) was determined in ELIspot assays following allogeneic myeloablative or nonmyeloablative stem cell transplantation (MST/NST). In 8 of 12 MST patients receiving an alemtuzumab-treated graft, the frequency of the EBV-specific reactivities was similar to or greater than that seen in the healthy controls. A response was detectable in 3 of 6 and 6 of 9 patients by 3 and 6 months, respectively, and in all patients by one year following MST. In contrast, only 1 of 9 (95% confidence interval [CI], 0-2.8) patients made a detectable EBV-specific response by 6 months following NST conditioned with fludarabine, melphalan, and alemtuzumab. Responses were detected in 7 of 10 patients by 1 year after NST. Parallel surveillance demonstrated that other virus infections occurred more frequently and earlier after transplantation in NST patients. The use of alemtuzumab in vivo in the nonmyeloablative conditioning might have resulted in the delay in EBV-specific T-cell recovery and increased virus infections.
Collapse
MESH Headings
- Alemtuzumab
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antibodies, Neoplasm/adverse effects
- Antibodies, Neoplasm/therapeutic use
- Antigens, Viral/blood
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Epstein-Barr Virus Infections/chemically induced
- Epstein-Barr Virus Infections/etiology
- Epstein-Barr Virus Infections/immunology
- Graft vs Host Disease/drug therapy
- Graft vs Host Disease/prevention & control
- Hematologic Neoplasms/therapy
- Hematopoietic Stem Cell Transplantation/adverse effects
- Hematopoietic Stem Cell Transplantation/methods
- Herpesvirus 4, Human/immunology
- Humans
- Lymphocyte Depletion
- T-Lymphocytes
- T-Lymphocytes, Cytotoxic/immunology
- Transplantation Conditioning/methods
- Transplantation, Homologous
Collapse
Affiliation(s)
- Suparno Chakrabarti
- Department of Haematology, Birmingham Heartlands Hospital, Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The innate immune system has a role not only in protecting the host during the initial period of virus infection, but also in shaping the nature of the adaptive immune response. In this review, we follow the kinetics of the virologic and immunologic events occurring from the time of hepatitis B virus (HBV) and hepatitis C virus (HCV) infection. We primarily discuss how the early events after infection might influence the development of the adaptive immune response in these 2 important viral infections and how new strategies for more efficient preventive and therapeutic vaccines can be derived from this knowledge.
Collapse
Affiliation(s)
- Antonio Bertoletti
- Institute of Hepatology, University College London, Royal Free and University College of London Medical School, London, UK.
| | | |
Collapse
|