1
|
García-Ferreras R, Osuna-Pérez J, Ramírez-Santiago G, Méndez-Pérez A, Acosta-Moreno AM, Del Campo L, Gómez-Sánchez MJ, Iborra M, Herrero-Fernández B, González-Granado JM, Sánchez-Madrid F, Carrasco YR, Boya P, Martínez-Martín N, Veiga E. Bacteria-instructed B cells cross-prime naïve CD8 + T cells triggering effective cytotoxic responses. EMBO Rep 2023:e56131. [PMID: 37184882 DOI: 10.15252/embr.202256131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 04/14/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023] Open
Abstract
In addition to triggering humoral responses, conventional B cells have been described in vitro to cross-present exogenous antigens activating naïve CD8+ T cells. Nevertheless, the way B cells capture these exogenous antigens and the physiological roles of B cell-mediated cross-presentation remain poorly explored. Here, we show that B cells capture bacteria by trans-phagocytosis from previously infected dendritic cells (DC) when they are in close contact. Bacterial encounter "instructs" the B cells to acquire antigen cross-presentation abilities, in a process that involves autophagy. Bacteria-instructed B cells, henceforth referred to as BacB cells, rapidly degrade phagocytosed bacteria, process bacterial antigens and cross-prime naïve CD8+ T cells which differentiate into specific cytotoxic cells that efficiently control bacterial infections. Moreover, a proof-of-concept experiment shows that BacB cells that have captured bacteria expressing tumor antigens could be useful as novel cellular immunotherapies against cancer.
Collapse
Affiliation(s)
- Raquel García-Ferreras
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Jesús Osuna-Pérez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Guillermo Ramírez-Santiago
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Almudena Méndez-Pérez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Andrés M Acosta-Moreno
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Lara Del Campo
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
- Departamento de Biología Celular, Facultad de Odontología, Universidad Complutense de Madrid, Madrid, Spain
| | - María J Gómez-Sánchez
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
- Department of Immunology, School of Medicine, Complutense University of Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Marta Iborra
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Beatriz Herrero-Fernández
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - José M González-Granado
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Sánchez-Madrid
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid, Madrid, Spain
| | - Yolanda R Carrasco
- Department of Immunology & Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Patricia Boya
- Department of Neuroscience, University of Fribourg, Fribourg, Switzerland
| | | | - Esteban Veiga
- Department of Molecular & Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| |
Collapse
|
2
|
Ibitokou SA, Gbedande K, Opata MM, Carpio VH, Marshall KM, Stephens R. Effects of Low-Level Persistent Infection on Maintenance of Immunity by CD4 T Cell Subsets and Th1 Cytokines. Infect Immun 2023; 91:e0053122. [PMID: 36920200 PMCID: PMC10016079 DOI: 10.1128/iai.00531-22] [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] [Indexed: 03/06/2023] Open
Abstract
CD4 T cells are required, along with antibodies, for complete protection from blood-stage infection with Plasmodium spp., which cause malaria. Without continuous exposure, as on emigration of people from endemic areas, protection from malaria decays. As in other persistent infections, low-level Plasmodium chabaudi infection protects the host from reinfection at 2 months postinfection, a phenomenon termed premunition. Premunition is correlated with T cell responses, rather than antibody levels. We previously showed that while both effector T cells (Teff) and memory T cells (Tmem) are present after infection, Teff protect better than Tmem. Here, we studied T cell kinetics post-infection by labeling dividing Ifng+ T cells with 5-bromo-2'-deoxyuridine (BrdU) in infected Ifng reporter mice. Large drops in specific T cell numbers and Ifng+ cells upon clearance of parasites suggest a mechanism for decay of protection. Although protection decays, CD4 Tmem persist, including a highly differentiated CD27- effector memory (Tem) subset that maintains some Ifng expression. In addition, pretreatment of chronically infected animals with neutralizing antibody to interferon gamma (IFN-γ) or with clodronate liposomes before reinfection decreases premunition, supporting a role for Th1-type immunity to reinfection. A pulse-chase experiment comparing chronically infected to treated animals showed that recently divided Ifng+ T cells, particularly IFN-γ+ TNF+ IL-2- T cells, are promoted by persistent infection. These data suggest that low-level persistent infection reduces CD4+ Tmem and multifunctional Teff survival, but promotes IFN-γ+ TNF+ IL-2- T cells and Ifng+ terminally differentiated effector T cells, and prolongs immunity.
Collapse
Affiliation(s)
- Samad A. Ibitokou
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Michael M. Opata
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Victor H. Carpio
- Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Karis M. Marshall
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
3
|
Smith MR, Gbedande K, Johnson CM, Campbell LA, Onjiko RS, Domingo ND, Opata MM. Model of severe malaria in young mice suggests unique response of CD4 T cells. Parasite Immunol 2022; 44:e12952. [PMID: 36131528 PMCID: PMC9787679 DOI: 10.1111/pim.12952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/30/2022]
Abstract
Severe malaria occurs most in young children but is poorly understood due to the absence of a developmentally-equivalent rodent model to study the pathogenesis of the disease. Though functional and quantitative deficiencies in innate response and a biased T helper 1 (Th1) response are reported in newborn pups, there is little information available about this intermediate stage of the adaptive immune system in murine neonates. To fill this gap in knowledge, we have developed a mouse model of severe malaria in young mice using 15-day old mice (pups) infected with Plasmodium chabaudi. We observe similar parasite growth pattern in pups and adults, with a 60% mortality and a decrease in the growth rate of the surviving young mice. Using a battery of behavioral assays, we observed neurological symptoms in pups that do not occur in infected wildtype adults. CD4+ T cells were activated and differentiated to an effector T cell (Teff) phenotype in both adult and pups. However, there were relatively fewer and less terminally differentiated pup CD4+ Teff than adult Teff. Interestingly, despite less activation, the pup Teff expressed higher T-bet than adults' cells. These data suggest that Th1 cells are functional in pups during Plasmodium infection but develop slowly.
Collapse
Affiliation(s)
- Margaret R. Smith
- Department of Biology, College of Arts and SciencesAppalachian State UniversityBooneNorth CarolinaUSA
- Present address:
Cancer Biology Ph.D. ProgramWake Forest College of MedicineWinston SalemNorth CarolinaUSA
| | - Komi Gbedande
- Division of Infectious Diseases, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Corey M. Johnson
- Department of Biology, College of Arts and SciencesAppalachian State UniversityBooneNorth CarolinaUSA
| | - Logan A. Campbell
- Department of Biology, College of Arts and SciencesAppalachian State UniversityBooneNorth CarolinaUSA
| | - Robert S. Onjiko
- Department of Biology, College of Arts and SciencesAppalachian State UniversityBooneNorth CarolinaUSA
| | - Nadia D. Domingo
- Division of Infectious Diseases, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTexasUSA
| | - Michael M. Opata
- Department of Biology, College of Arts and SciencesAppalachian State UniversityBooneNorth CarolinaUSA
| |
Collapse
|
4
|
Olatunde AC, Cornwall DH, Roedel M, Lamb TJ. Mouse Models for Unravelling Immunology of Blood Stage Malaria. Vaccines (Basel) 2022; 10:1525. [PMID: 36146602 PMCID: PMC9501382 DOI: 10.3390/vaccines10091525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria comprises a spectrum of disease syndromes and the immune system is a major participant in malarial disease. This is particularly true in relation to the immune responses elicited against blood stages of Plasmodium-parasites that are responsible for the pathogenesis of infection. Mouse models of malaria are commonly used to dissect the immune mechanisms underlying disease. While no single mouse model of Plasmodium infection completely recapitulates all the features of malaria in humans, collectively the existing models are invaluable for defining the events that lead to the immunopathogenesis of malaria. Here we review the different mouse models of Plasmodium infection that are available, and highlight some of the main contributions these models have made with regards to identifying immune mechanisms of parasite control and the immunopathogenesis of malaria.
Collapse
Affiliation(s)
| | | | | | - Tracey J. Lamb
- Department of Pathology, University of Utah, Emma Eccles Jones Medical Research Building, 15 N Medical Drive E, Room 1420A, Salt Lake City, UT 84112, USA
| |
Collapse
|
5
|
Bergantini L, d’Alessandro M, Otranto A, Cavallaro D, Gangi S, Fossi A, Perillo F, Luzzi L, Zanfrini E, Paladini P, Sestini P, Rottoli P, Bargagli E, Bennett D. Characterization of NKG2-A/-C, Kir and CD57 on NK Cells Stimulated with pp65 and IE-1 Antigens in Patients Awaiting Lung Transplant. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071081. [PMID: 35888169 PMCID: PMC9325149 DOI: 10.3390/life12071081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/03/2022]
Abstract
Introduction: Cytomegalovirus (CMV) is the leading opportunistic infection in lung transplant (LTx) recipients. CMV is associated with graft failure and decreased survival. Recently, new antiviral therapies have been proposed. The present study aimed to investigate NK and T cell subsets of patients awaiting LTx. We analyzed the cellular populations between reactive and non-reactive QuantiFERON (QF) CMV patients for the prediction of immunological response to infection. Methods: Seventeen pre-LTx patients and 15 healthy controls (HC) have been enrolled. QF and IFN-γ ELISA assay detections were applied. NK cell subsets and T cell and proliferation assay were detected before and after stimulation with pp-65 and IE-1 CMV antigens after stratification as QF+ and QF−. Furthermore, we quantified the serum concentrations of NK− and T-related cytokines by bead-based multiplex analysis. Results: CD56brCD16lowNKG2A+KIR+ resulted in the best discriminatory cellular subsets between pre-LTx and HC. Discrepancies emerged between serology and QF assay. Better proliferative capability emerged from patients who were QF+, in particular in CD8 and CD25-activated cells. CD56brCD16low, adaptive/memory-like NK and CD8Teff were highly increased only in QF+ patients. Conclusions: QF more than serology is useful in the detection of patients able to respond to viral infection. This study provides new insights in terms of immunological responses to CMV in pre-LTX patients, particularly in NK and T cells biology.
Collapse
Affiliation(s)
- Laura Bergantini
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Miriana d’Alessandro
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
- Respiratory Disease and Lung Transplant Unit, Department of Medical Sciences, Surgery and Neurosciences, Siena University, 53100 Siena, Italy
- Correspondence: ; Tel.: +39-0577-586713; Fax: +39-0577-280744
| | - Ambra Otranto
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Dalila Cavallaro
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Sara Gangi
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Antonella Fossi
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Felice Perillo
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Luca Luzzi
- Thoracic Surgery Unit, Cardio-Thoracic and Vascular Department, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), 53100 Siena, Italy; (L.L.); (E.Z.); (P.P.)
| | - Edoardo Zanfrini
- Thoracic Surgery Unit, Cardio-Thoracic and Vascular Department, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), 53100 Siena, Italy; (L.L.); (E.Z.); (P.P.)
| | - Piero Paladini
- Thoracic Surgery Unit, Cardio-Thoracic and Vascular Department, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), 53100 Siena, Italy; (L.L.); (E.Z.); (P.P.)
| | - Piersante Sestini
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Paola Rottoli
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - Elena Bargagli
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| | - David Bennett
- Respiratory Disease Unit, Department of Medical Sciences, University Hospital of Siena, Azienda Ospedaliera Universitaria Senese (AOUS), Viale Bracci, 53100 Siena, Italy; (L.B.); (A.O.); (D.C.); (S.G.); (A.F.); (F.P.); (P.S.); (P.R.); (E.B.); (D.B.)
| |
Collapse
|
6
|
Liu G, Qin L, Li Y, Zhao S, Shugay M, Yan Y, Ye Y, Chen Y, Huang C, Bayaer N, Adah D, Zhang H, Su Z, Chen X. Subsequent malaria enhances virus-specific T cell immunity in SIV-infected Chinese rhesus macaques. Cell Commun Signal 2022; 20:101. [PMID: 35778766 PMCID: PMC9248186 DOI: 10.1186/s12964-022-00910-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background Coinfection with HIV and Plasmodium parasites is fairly common, but the sequence of infection with these two pathogens and their impact on disease progression are poorly understood. Methods A Chinese rhesus macaque HIV and Plasmodium coinfection model was established to compare the impact of pre-existing and subsequent malaria on the progression of SIV infection. Results We found that a pre-existing malaria caused animals to produce a greater number of CD4+CCR5+ T cells for SIV replication, resulting in higher viral loads. Conversely, subsequent malaria induced a substantially larger proportion of CD4+CD28highCD95high central memory T cells and a stronger SIV-specific T cell response, maintained the repertoire diversity of SIV-specific T cell receptors, and generated new SIV-specific T cell clonotypes to trace SIV antigenic variation, resulting in improved survival of SIV-infected animals. Conclusion The complex outcomes of this study may have important implications for research on human HIV and malaria coinfection. The infection order of the two pathogens (HIV and malaria parasites) should be emphasized. Video abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00910-7.
Collapse
Affiliation(s)
- Guangjie Liu
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Graduate School, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.,The Fist Affiliated Hospital of Shenzhen University, Shenzhen, China.,Shenzhen Institute of Geriatrics, Shenzhen, China
| | - Li Qin
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,CAS Lamvac Biotech Co., Ltd, Guangzhou, China
| | - Youjia Li
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,The Fist Affiliated Hospital of Shenzhen University, Shenzhen, China.,Integrated Chinese and Western Medicine Postdoctoral Research Station, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,Shenzhen Institute of Geriatrics, Shenzhen, China
| | - Siting Zhao
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,CAS Lamvac Biotech Co., Ltd, Guangzhou, China
| | - Mikhail Shugay
- Genomics of Adaptive Immunity Laboratory, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences (RAS), Moscow, Russia
| | - Yongxiang Yan
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yijian Ye
- Laboratory of Immunobiology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yue Chen
- Laboratory of Immunobiology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Cuizhu Huang
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Nashun Bayaer
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dickson Adah
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Zhong Su
- Laboratory of Immunobiology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| | - Xiaoping Chen
- Laboratory of Pathogen Biology, State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,CAS Lamvac Biotech Co., Ltd, Guangzhou, China.
| |
Collapse
|
7
|
Schulte S, Heide J, Ackermann C, Peine S, Ramharter M, Mackroth MS, Woost R, Jacobs T, Schulze zur Wiesch J. Deciphering the Plasmodium falciparum malaria-specific CD4+ T-cell response: ex vivo detection of high frequencies of PD-1+TIGIT+ EXP1-specific CD4+ T cells using a novel HLA-DR11-restricted MHC class II tetramer. Clin Exp Immunol 2021; 207:227-236. [PMID: 35020841 PMCID: PMC8982981 DOI: 10.1093/cei/uxab027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 02/03/2023] Open
Abstract
Relatively little is known about the ex vivo frequency and phenotype of the Plasmodium falciparum-specific CD4+ T-cell response in humans. The exported protein 1 (EXP1) is expressed by plasmodia at both, the liver stage and blood stage, of infection making it a potential target for CD4+ and CD8+ effector T cells. Here, a fluorochrome-labelled HLA-DRB1∗11:01-restriced MHC class II tetramer derived from the P. falciparum EXP1 (aa62-74) was established for ex vivo tetramer analysis and magnetic bead enrichment in 10 patients with acute malaria. EXP1-specific CD4+ T cells were detectable in 9 out of 10 (90%) malaria patients expressing the HLA-DRB1∗11 molecule with an average ex vivo frequency of 0.11% (0-0.22%) of total CD4+ T cells. The phenotype of EXP1-specific CD4+ T cells was further assessed using co-staining with activation (CD38, HLA-DR, CD26), differentiation (CD45RO, CCR7, KLRG1, CD127), senescence (CD57), and co-inhibitory (PD-1, TIGIT, LAG-3, TIM-3) markers as well as the ectonucleotidases CD39 and CD73. EXP1-specific tetramer+ CD4+ T cells had a distinct phenotype compared to bulk CD4+ T cells and displayed a highly activated effector memory phenotype with elevated levels of co-inhibitory receptors and activation markers: EXP1-specific CD4+ T cells universally expressed the co-inhibitory receptors PD-1 and TIGIT as well as the activation marker CD38 and showed elevated frequencies of CD39. These results demonstrate that MHC class II tetramer enrichment is a sensitive approach to investigate ex vivo antigen-specific CD4+ T cells in malaria patients that will aid further analysis of the role of CD4+ T cells during malaria.
Collapse
Affiliation(s)
- Sophia Schulte
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janna Heide
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Christin Ackermann
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Ramharter
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany,Department of Tropical Medicine, Bernhard-Nocht-Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - Maria Sophia Mackroth
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany,Department of Tropical Medicine, Bernhard-Nocht-Institute for Tropical Medicine (BNITM), Hamburg, Germany,Protozoa Immunology, Bernhard-Nocht-Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - Robin Woost
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Thomas Jacobs
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany,Protozoa Immunology, Bernhard-Nocht-Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | - Julian Schulze zur Wiesch
- Infectious Diseases Unit, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany,Correspondence: Julian Schulze zur Wiesch, Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| |
Collapse
|
8
|
Acquired clinical immunity to malaria in non-human primates co-infected with Schistosoma and Plasmodium parasites. Infect Immun 2021; 90:e0046421. [PMID: 34871040 DOI: 10.1128/iai.00464-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background. Naturally acquired immunity to malaria develops over several years and can be compromised by concomitant infections. This study explored the influence of chronic schistosomiasis on clinical outcome and immunity to repeated malaria infection. Methods. Two groups of baboons (n=8 each), were infected with Schistosoma mansoni cercariae to establish chronic infections. One of the two groups was treated with Praziquantel to eliminate schistosome infection. The two groups plus a new malaria control group (n=8), were inoculated three times with Plasmodium knowlesi parasites at one-month intervals. Clinical data, IgG, IgG1, memory T-cells and monocyte levels were recorded. Results. We observed after three P. knowlesi infections; i) reduced clinical symptoms in all groups with each subsequent infection, ii) increase IgG and IgG1in the malaria control (Pk-only) group iii) increased IgG and IgG1, CD14+ and CD14-CD16+ in the Schistosoma treated (Schisto/PZQ+Pk) group and iv) significantly lower IgG and IgG1 levels compared to Pk-only, reduced CD4+CD45RO+ and increased CD14-CD16+ cells in the co-infected (Schisto+Pk) group. Conclusion. Chronic S. mansoni does not compromise establishment of clinical immunity after multiple malaria infections with non-classical monocytes seeming to play a role. Failure to develop robust antibody and memory T-cells may have a long-term impact on acquired immunity to malaria infection.
Collapse
|
9
|
Smith NL, Nahrendorf W, Sutherland C, Mooney JP, Thompson J, Spence PJ, Cowan GJM. A Conserved TCRβ Signature Dominates a Highly Polyclonal T-Cell Expansion During the Acute Phase of a Murine Malaria Infection. Front Immunol 2020; 11:587756. [PMID: 33329568 PMCID: PMC7719809 DOI: 10.3389/fimmu.2020.587756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/27/2020] [Indexed: 01/31/2023] Open
Abstract
CD4+ αβ T-cells are key mediators of the immune response to a first Plasmodium infection, undergoing extensive activation and splenic expansion during the acute phase of an infection. However, the clonality and clonal composition of this expansion has not previously been described. Using a comparative infection model, we sequenced the splenic CD4+ T-cell receptor repertoires generated over the time-course of a Plasmodium chabaudi infection. We show through repeat replicate experiments, single-cell RNA-seq, and analyses of independent RNA-seq data, that following a first infection - within a highly polyclonal expansion - T-effector repertoires are consistently dominated by TRBV3 gene usage. Clustering by sequence similarity, we find the same dominant clonal signature is expanded across replicates in the acute phase of an infection, revealing a conserved pathogen-specific T-cell response that is consistently a hallmark of a first infection, but not expanded upon re-challenge. Determining the host or parasite factors driving this conserved response may uncover novel immune targets for malaria therapeutic purposes.
Collapse
Affiliation(s)
- Natasha L. Smith
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
10
|
Transcriptome dynamics of CD4 + T cells during malaria maps gradual transit from effector to memory. Nat Immunol 2020; 21:1597-1610. [PMID: 33046889 DOI: 10.1038/s41590-020-0800-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023]
Abstract
The dynamics of CD4+ T cell memory development remain to be examined at genome scale. In malaria-endemic regions, antimalarial chemoprevention protects long after its cessation and associates with effects on CD4+ T cells. We applied single-cell RNA sequencing and computational modelling to track memory development during Plasmodium infection and treatment. In the absence of central memory precursors, two trajectories developed as T helper 1 (TH1) and follicular helper T (TFH) transcriptomes contracted and partially coalesced over three weeks. Progeny of single clones populated TH1 and TFH trajectories, and fate-mapping suggested that there was minimal lineage plasticity. Relationships between TFH and central memory were revealed, with antimalarials modulating these responses and boosting TH1 recall. Finally, single-cell epigenomics confirmed that heterogeneity among effectors was partially reset in memory. Thus, the effector-to-memory transition in CD4+ T cells is gradual during malaria and is modulated by antiparasitic drugs. Graphical user interfaces are presented for examining gene-expression dynamics and gene-gene correlations ( http://haquelab.mdhs.unimelb.edu.au/cd4_memory/ ).
Collapse
|
11
|
Arroyo EN, Pepper M. B cells are sufficient to prime the dominant CD4+ Tfh response to Plasmodium infection. J Exp Med 2020; 217:jem.20190849. [PMID: 31748243 PMCID: PMC7041722 DOI: 10.1084/jem.20190849] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/19/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Arroyo and Pepper demonstrate that interactions with B cells, not dendritic cells, are required for the priming of the CD4+ T cell response during Plasmodium infection. This results in a Tfh-biased response as reported by others in both mice and humans. CD4+ T follicular helper (Tfh) cells dominate the acute response to a blood-stage Plasmodium infection and provide signals to direct B cell differentiation and protective antibody expression. We studied antigen-specific CD4+ Tfh cells responding to Plasmodium infection in order to understand the generation and maintenance of the Tfh response. We discovered that a dominant, phenotypically stable, CXCR5+ Tfh population emerges within the first 4 d of infection and results in a CXCR5+ CCR7+ Tfh/central memory T cell response that persists well after parasite clearance. We also found that CD4+ T cell priming by B cells was both necessary and sufficient to generate this Tfh-dominant response, whereas priming by conventional dendritic cells was dispensable. This study provides important insights into the development of CD4+ Tfh cells during Plasmodium infection and highlights the heterogeneity of antigen-presenting cells involved in CD4+ T cell priming.
Collapse
Affiliation(s)
- E Nicole Arroyo
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
12
|
Carpio VH, Aussenac F, Puebla-Clark L, Wilson KD, Villarino AV, Dent AL, Stephens R. T Helper Plasticity Is Orchestrated by STAT3, Bcl6, and Blimp-1 Balancing Pathology and Protection in Malaria. iScience 2020; 23:101310. [PMID: 32634740 PMCID: PMC7339051 DOI: 10.1016/j.isci.2020.101310] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/20/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022] Open
Abstract
Hybrid Th1/Tfh cells (IFN-γ+IL-21+CXCR5+) predominate in response to several persistent infections. In Plasmodium chabaudi infection, IFN-γ+ T cells control parasitemia, whereas antibody and IL-21+Bcl6+ T cells effect final clearance, suggesting an evolutionary driver for the hybrid population. We found that CD4-intrinsic Bcl6, Blimp-1, and STAT3 coordinately regulate expression of the Th1 master regulator T-bet, supporting plasticity of CD4 T cells. Bcl6 and Blimp-1 regulate CXCR5 levels, and T-bet, IL-27Rα, and STAT3 modulate cytokines in hybrid Th1/Tfh cells. Infected mice with STAT3 knockout (KO) T cells produced less antibody and more Th1-like IFN-γ+IL-21−CXCR5lo effector and memory cells and were protected from re-infection. Conversely, T-bet KO mice had reduced Th1-bias upon re-infection and prolonged secondary parasitemia. Therefore, each feature of the CD4 T cell population phenotype is uniquely regulated in this persistent infection, and the cytokine profile of memory T cells can be modified to enhance the effectiveness of the secondary response. Plasmodium infection induces a CXCR5+IFN-γ+IL-21+ hybrid Th1/Tfh cell subset STAT3/WSX-1, T-bet, Bcl6, and Blimp-1 regulate different aspects of Th1/Tfh phenotype T cell-intrinsic STAT3 regulates degree of Th1 commitment of hybrid Th1/Tfh Shifting the plastic response toward Th1-like cells promotes resistance from reinfection
Collapse
Affiliation(s)
- Victor H Carpio
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | - Florentin Aussenac
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | - Lucinda Puebla-Clark
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | - Kyle D Wilson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-0435, USA
| | - Alejandro V Villarino
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Metabolic, and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-1674, USA
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Robin Stephens
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-0435, USA; Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX 77555-0435, USA.
| |
Collapse
|
13
|
Abstract
Immunity to malaria has been linked to the availability and function of helper CD4+ T cells, cytotoxic CD8+ T cells and γδ T cells that can respond to both the asymptomatic liver stage and the symptomatic blood stage of Plasmodium sp. infection. These T cell responses are also thought to be modulated by regulatory T cells. However, the precise mechanisms governing the development and function of Plasmodium-specific T cells and their capacity to form tissue-resident and long-lived memory populations are less well understood. The field has arrived at a point where the push for vaccines that exploit T cell-mediated immunity to malaria has made it imperative to define and reconcile the mechanisms that regulate the development and functions of Plasmodium-specific T cells. Here, we review our current understanding of the mechanisms by which T cell subsets orchestrate host resistance to Plasmodium infection on the basis of observational and mechanistic studies in humans, non-human primates and rodent models. We also examine the potential of new experimental strategies and human infection systems to inform a new generation of approaches to harness T cell responses against malaria.
Collapse
|
14
|
Gbedande K, Carpio VH, Stephens R. Using two phases of the CD4 T cell response to blood-stage murine malaria to understand regulation of systemic immunity and placental pathology in Plasmodium falciparum infection. Immunol Rev 2020; 293:88-114. [PMID: 31903675 PMCID: PMC7540220 DOI: 10.1111/imr.12835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Plasmodium falciparum infection and malaria remain a risk for millions of children and pregnant women. Here, we seek to integrate knowledge of mouse and human T helper cell (Th) responses to blood-stage Plasmodium infection to understand their contribution to protection and pathology. Although there is no complete Th subset differentiation, the adaptive response occurs in two phases in non-lethal rodent Plasmodium infection, coordinated by Th cells. In short, cellular immune responses limit the peak of parasitemia during the first phase; in the second phase, humoral immunity from T cell-dependent germinal centers is critical for complete clearance of rapidly changing parasite. A strong IFN-γ response kills parasite, but an excess of TNF compared with regulatory cytokines (IL-10, TGF-β) can cause immunopathology. This common pathway for pathology is associated with anemia, cerebral malaria, and placental malaria. These two phases can be used to both understand how the host responds to rapidly growing parasite and how it attempts to control immunopathology and variation. This dual nature of T cell immunity to Plasmodium is discussed, with particular reference to the protective nature of the continuous generation of effector T cells, and the unique contribution of effector memory T cells.
Collapse
Affiliation(s)
- Komi Gbedande
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Victor H Carpio
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Robin Stephens
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
15
|
Abstract
A single exposure to many viral and bacterial pathogens typically induces life-long immunity, however, the development of the protective immunity to Plasmodium parasites is strikingly less efficient and achieves only partial protection, with adults residing in endemic areas often experiencing asymptomatic infections. Although naturally acquired immunity to malaria requires both cell-mediated and humoral immune responses, antibodies govern the control of malarial disease caused by the blood-stage form of the parasites. A large body of epidemiological evidence described that antibodies to Plasmodium antigens are inefficiently generated and rapidly lost without continued parasite exposure, suggesting that malaria is accompanied by defects in the development of immunological B cell memory. This topic has been of focus of recent studies of malaria infection in humans and mice. This review examines the main findings to date on the processes that modulate the acquisition of memory B cell responses to malaria, and highlights the importance of closing outstanding gaps of knowledge in the field for the rational design of next generation therapeutics against malaria.
Collapse
Affiliation(s)
- Ann Ly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Diana S Hansen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
16
|
Cheng Q, Liu J, Pei Y, Zhang Y, Zhou D, Pan W, Zhang J. Neddylation contributes to CD4+ T cell-mediated protective immunity against blood-stage Plasmodium infection. PLoS Pathog 2018; 14:e1007440. [PMID: 30462731 PMCID: PMC6249024 DOI: 10.1371/journal.ppat.1007440] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 10/29/2018] [Indexed: 01/30/2023] Open
Abstract
CD4+ T cells play predominant roles in protective immunity against blood-stage Plasmodium infection, both for IFN-γ-dependent effector mechanisms and providing B cell helper signals. Neddylation, an ubiquitination-like process triggered by covalent conjugation of NEDD8 to specific targets, has emerged as a potential regulator of T cell activities to TCR engagement. However, its contribution to T cell-mediated immunity to blood-stage malaria remains unclear. Here using an experimental model induced by Plasmodium yoelii 17XNL, and conditional knockout mice with T cell-specific deficiency of crucial components of neddylation pathway, we demonstrate activation of neddylation in T cells during blood-stage Plasmodium infection is essential for parasite control and host survival. Mechanistically, we show that apart from promoting CD4+ T cell activation, proliferation, and development of protective T helper 1 (Th1) cell response as suggested previously, neddylation is also required for supporting CD4+ T cell survival, mainly through B-cell lymphoma-2 (Bcl-2) mediated suppression of the mitochondria-dependent apoptosis. Furthermore, we provide evidence that neddylation contributes to follicular helper T (Tfh) cell differentiation, probably via augmenting the ubiquitin ligase Itch activity and proteasomal degradation of FoxO1, thereby facilitating germinal center (GC) formation and parasite-specific antibody production. This study identifies neddylation as a positive regulator of anti-Plasmodium immunity and provides insight into an involvement of such pathway in host resistance to infectious diseases. Malaria, which is caused by the intracellular parasite Plasmodium, remains a major infectious disease with significant morbidity and mortality annually. Better understanding of the molecular mechanisms involved in protective immunity against the pathogenic blood-stage Plasmodium will facilitate development of anti-malarial drugs and vaccines. Neddylation has recently been identified as a potential regulator of T cell function. Here, we directly addressed the effects of neddylation on T cell responses and the outcome of blood-stage P. yoelii 17XNL malaria. We show that activation of neddylation in T cells is essential for IFN-γ-mediated proinflammatory response and generation of parasite-specific antibodies, thus contributing to full resolution of the infection. This is primarily associated with the reported beneficial effects of neddylation on CD4+ T cell activities, including activation, proliferation, and differentiation into T helper 1 (Th1) cells. Additionally, we establish a novel role of neddylation in parasite-responsive CD4+ T cell survival and follicular helper T (Tfh) cell differentiation. Therefore, we provide evidence that neddylation may represent a novel mechanism in orchestrating optimum CD4+ T cell effector response and subsequent humoral immunity to blood-stage Plasmodium infection.
Collapse
Affiliation(s)
- Qianqian Cheng
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, China
- * E-mail: (QC); (JZ)
| | - Jian Liu
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Yujun Pei
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Yaolin Zhang
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, China
| | - Dawang Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Fujian, China
| | - Weiqing Pan
- Department of Tropical Infectious Diseases, Second Military Medical University, Shanghai, China
| | - Jiyan Zhang
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, China
- * E-mail: (QC); (JZ)
| |
Collapse
|
17
|
Zander RA, Vijay R, Pack AD, Guthmiller JJ, Graham AC, Lindner SE, Vaughan AM, Kappe SHI, Butler NS. Th1-like Plasmodium-Specific Memory CD4 + T Cells Support Humoral Immunity. Cell Rep 2018; 21:1839-1852. [PMID: 29141217 DOI: 10.1016/j.celrep.2017.10.077] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 09/15/2017] [Accepted: 10/20/2017] [Indexed: 01/13/2023] Open
Abstract
Effector T cells exhibiting features of either T helper 1 (Th1) or T follicular helper (Tfh) populations are essential to control experimental Plasmodium infection and are believed to be critical for resistance to clinical malaria. To determine whether Plasmodium-specific Th1- and Tfh-like effector cells generate memory populations that contribute to protection, we developed transgenic parasites that enable high-resolution study of anti-malarial memory CD4 T cells in experimental models. We found that populations of both Th1- and Tfh-like Plasmodium-specific memory CD4 T cells persist. Unexpectedly, Th1-like memory cells exhibit phenotypic and functional features of Tfh cells during recall and provide potent B cell help and protection following transfer, characteristics that are enhanced following ligation of the T cell co-stimulatory receptor OX40. Our findings delineate critical functional attributes of Plasmodium-specific memory CD4 T cells and identify a host-specific factor that can be targeted to improve resolution of acute malaria and provide durable, long-term protection against Plasmodium parasite re-exposure.
Collapse
Affiliation(s)
- Ryan A Zander
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Rahul Vijay
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Angela D Pack
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Jenna J Guthmiller
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Amy C Graham
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Scott E Lindner
- Center for Malaria Research, Penn State University, University Park, PA 16802, USA; Department of Biochemistry and Molecular Biology, Penn State University, University Park, PA 16802, USA; Center for Infectious Disease Research, Seattle, WA 98109, USA
| | | | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Seattle, WA 98109, USA
| | - Noah S Butler
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Graduate Program in Biosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
18
|
Dhume K, McKinstry KK. Early programming and late-acting checkpoints governing the development of CD4 T-cell memory. Immunology 2018; 155:53-62. [PMID: 29701246 DOI: 10.1111/imm.12942] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/17/2018] [Accepted: 04/18/2018] [Indexed: 12/25/2022] Open
Abstract
CD4 T cells contribute to protection against pathogens through numerous mechanisms. Incorporating the goal of memory CD4 T-cell generation into vaccine strategies therefore offers a powerful approach to improve their efficacy, especially in situations where humoral responses alone cannot confer long-term immunity. These threats include viruses such as influenza that mutate coat proteins to avoid neutralizing antibodies, but that are targeted by T cells that recognize more conserved protein epitopes shared by different strains. A major barrier in the design of such vaccines is that the mechanisms controlling the efficiency with which memory cells form remain incompletely understood. Here, we discuss recent insights into fate decisions controlling memory generation. We focus on the importance of three general cues: interleukin-2, antigen and co-stimulatory interactions. It is increasingly clear that these signals have a powerful influence on the capacity of CD4 T cells to form memory during two distinct phases of the immune response. First, through 'programming' that occurs during initial priming, and second, through 'checkpoints' that operate later during the effector stage. These findings indicate that novel vaccine strategies must seek to optimize cognate interactions, during which interleukin-2-, antigen- and co-stimulation-dependent signals are tightly linked, well beyond initial antigen encounter to induce robust memory CD4 T cells.
Collapse
Affiliation(s)
- Kunal Dhume
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Karl Kai McKinstry
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
19
|
Opata MM, Ibitokou SA, Carpio VH, Marshall KM, Dillon BE, Carl JC, Wilson KD, Arcari CM, Stephens R. Protection by and maintenance of CD4 effector memory and effector T cell subsets in persistent malaria infection. PLoS Pathog 2018; 14:e1006960. [PMID: 29630679 PMCID: PMC5908200 DOI: 10.1371/journal.ppat.1006960] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/19/2018] [Accepted: 03/04/2018] [Indexed: 01/12/2023] Open
Abstract
Protection at the peak of Plasmodium chabaudi blood-stage malaria infection is provided by CD4 T cells. We have shown that an increase in Th1 cells also correlates with protection during the persistent phase of malaria; however, it is unclear how these T cells are maintained. Persistent malaria infection promotes protection and generates both effector T cells (Teff), and effector memory T cells (Tem). We have previously defined new CD4 Teff (IL-7Rα-) subsets from Early (TeffEarly, CD62LhiCD27+) to Late (TeffLate, CD62LloCD27-) activation states. Here, we tested these effector and memory T cell subsets for their ability to survive and protect in vivo. We found that both polyclonal and P. chabaudi Merozoite Surface Protein-1 (MSP-1)-specific B5 TCR transgenic Tem survive better than Teff. Surprisingly, as Tem are associated with antigen persistence, Tem survive well even after clearance of infection. As previously shown during T cell contraction, TeffEarly, which can generate Tem, also survive better than other Teff subsets in uninfected recipients. Two other Tem survival mechanisms identified here are that low-level chronic infection promotes Tem both by driving their proliferation, and by programming production of Tem from Tcm. Protective CD4 T cell phenotypes have not been precisely determined in malaria, or other persistent infections. Therefore, we tested purified memory (Tmem) and Teff subsets in protection from peak pathology and parasitemia in immunocompromised recipient mice. Strikingly, among Tmem (IL-7Rαhi) subsets, only TemLate (CD62LloCD27-) reduced peak parasitemia (19%), though the dominant memory subset is TemEarly, which is not protective. In contrast, all Teff subsets reduced peak parasitemia by more than half, and mature Teff can generate Tem, though less. In summary, we have elucidated four mechanisms of Tem maintenance, and identified two long-lived T cell subsets (TemLate, TeffEarly) that may represent correlates of protection or a target for longer-lived vaccine-induced protection against malaria blood-stages. Malaria causes significant mortality but current vaccine candidates have poor efficacy and duration, as does natural immunity to malaria. T helper cells (CD4+) are essential to protection from malaria, but it is unknown what kinds of T cells would be both protective and long-lasting. Here, we explored the mechanisms of survival used by memory T cells in malaria, and their ability to protect immunodeficient animals from malaria. We identified four mechanisms by which memory T cells are maintained in chronic infection. We also showed that highly activated effector T cells protect better than memory T cells in general, however, effector T cells have a shorter lifespan suggesting a mechanism for short-lived immunity. In total, we identified two protective T cell subsets that are long-lived. Unfortunately, the memory T cell subset that protects, is not the predominant memory T cell population generated by natural infection, suggesting a mechanism for the poor immunity seen in malaria. Our work suggests that vaccines that induce these two T cell subsets may improve on current immunity from malaria infection and disease.
Collapse
Affiliation(s)
- Michael M. Opata
- Departments of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Samad A. Ibitokou
- Departments of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Victor H. Carpio
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Karis M. Marshall
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Brian E. Dillon
- Departments of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Jordan C. Carl
- Departments of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Kyle D. Wilson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
| | - Christine M. Arcari
- Department of Preventive Medicine & Community Health, University of Texas Medical Branch Galveston, TX, United States of America
| | - Robin Stephens
- Departments of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States of America
- * E-mail:
| |
Collapse
|
20
|
Zander RA, Vijay R, Pack AD, Guthmiller JJ, Graham AC, Lindner SE, Vaughan AM, Kappe SHI, Butler NS. Th1-like Plasmodium-Specific Memory CD4 + T Cells Support Humoral Immunity. Cell Rep 2018; 23:1230-1237. [PMID: 29694898 DOI: 10.1016/j.celrep.2018.04.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
21
|
Orme IM, Henao-Tamayo MI. Trying to See the Forest through the Trees: Deciphering the Nature of Memory Immunity to Mycobacterium tuberculosis. Front Immunol 2018; 9:461. [PMID: 29568298 PMCID: PMC5852080 DOI: 10.3389/fimmu.2018.00461] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/21/2018] [Indexed: 01/18/2023] Open
Abstract
The purpose of vaccination against tuberculosis and other diseases is to establish a heightened state of acquired specific resistance in which the memory immune response is capable of mediating an accelerated and magnified expression of protection to the pathogen when this is encountered at a later time. In the earliest studies in mice infected with Mycobacterium tuberculosis, memory immunity and the cells that express this were definable both in terms of kinetics of emergence, and soon thereafter by the levels of expression of markers including CD44, CD62L, and the chemokine receptor CCR7, allowing the identification of effector memory and central memory T cell subsets. Despite these initial advances in knowledge, more recent information has not revealed more clarity, but instead, has created a morass of complications—complications that, if not resolved, could harm correct vaccine design. Here, we discuss two central issues. The first is that we have always assumed that memory is induced in the same way, and consists of the same T cells, regardless of whether that immunity is generated by BCG vaccination, or by exposure to M. tuberculosis followed by effective chemotherapy. This assumption is almost certainly incorrect. Second, a myriad of additional memory subsets have now been described, such as resident, stem cell-like, tissue specific, among others, but as yet we know nothing about the relative importance of each, or whether if a new vaccine needs to induce all of these, or just some, to be fully effective.
Collapse
Affiliation(s)
- Ian M Orme
- Mycobacteria Research Laboratories, Colorado State University, Fort Collins, CO, United States
| | - Marcela I Henao-Tamayo
- Mycobacteria Research Laboratories, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
22
|
Quan L, Lan X, Meng Y, Guo X, Guo Y, Zhao L, Chen X, Liu A. BTLA marks a less cytotoxic T-cell subset in diffuse large B-cell lymphoma with high expression of checkpoints. Exp Hematol 2018; 60:47-56.e1. [PMID: 29353075 DOI: 10.1016/j.exphem.2018.01.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 01/03/2018] [Accepted: 01/09/2018] [Indexed: 01/07/2023]
Abstract
Immunotherapy results in lymphoma have been encouraging. Preclinical and clinical trials have proven checkpoint blockade, such as PD-1 antibody, as an effective treatment for lymphoma, including diffuse large B-cell lymphoma (DLBCL). Combination of checkpoint blockades has emerged as a new way to treat lymphoma; however, the status of checkpoint expression and their function in DLBCL have not been fully elucidated yet. In this study, we examined the expression of BTLA, PD-1, TIM-3, LIGHT, and LAG-3 in tumor microenvironmental T cells of DLBCL using flow cytometry and compared the cytotoxicity and differentiation status of BTLA+ and BTLA- T-cells. We further characterized the relationship of STAT3 phosphorylation (p-STAT3) with BTLA expression. Our results suggest that BTLA+ T cells highly express other checkpoint molecules, including PD-1, TIM-3, LIGHT, and LAG-3. Moreover, high expression of BTLA is correlated with advanced stage of DLBCL. BTLA+ T cells have a less-differentiated phenotype, lower cytolytic function, and higher potential to proliferate compared with BTLA- T cells. Taken together, our data provide the first evidence that increased BTLA predicts poor prognosis in patients with DLBCL, and blockade of BTLA with other checkpoints may potentially represent a new strategy for immunotherapy of DLBCL.
Collapse
Affiliation(s)
- Lina Quan
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Xiuwen Lan
- Gastroenterological Department, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Yuanyuan Meng
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Xiuchen Guo
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Yiwei Guo
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Lina Zhao
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Xue Chen
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Aichun Liu
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.
| |
Collapse
|
23
|
Ibitokou SA, Dillon BE, Sinha M, Szczesny B, Delgadillo A, Reda Abdelrahman D, Szabo C, Abu-Elheiga L, Porter C, Tuvdendorj D, Stephens R. Early Inhibition of Fatty Acid Synthesis Reduces Generation of Memory Precursor Effector T Cells in Chronic Infection. THE JOURNAL OF IMMUNOLOGY 2017; 200:643-656. [PMID: 29237780 DOI: 10.4049/jimmunol.1602110] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 11/09/2017] [Indexed: 01/13/2023]
Abstract
Understanding the mechanisms of CD4 memory T cell (Tmem) differentiation in malaria is critical for vaccine development. However, the metabolic regulation of CD4 Tmem differentiation is not clear, particularly in persistent infections. In this study, we investigated the role of fatty acid synthesis (FAS) in Tmem development in Plasmodium chabaudi chronic mouse malaria infection. We show that T cell-specific deletion and early pharmaceutical inhibition of acetyl CoA carboxylase 1, the rate limiting step of FAS, inhibit generation of early memory precursor effector T cells (MPEC). To compare the role of FAS during early differentiation or survival of Tmem in chronic infection, a specific inhibitor of acetyl CoA carboxylase 1, 5-(tetradecyloxy)-2-furoic acid, was administered at different times postinfection. Strikingly, the number of Tmem was only reduced when FAS was inhibited during T cell priming and not during the Tmem survival phase. FAS inhibition during priming increased effector T cell (Teff) proliferation and strongly decreased peak parasitemia, which is consistent with improved Teff function. Conversely, MPEC were decreased, in a T cell-intrinsic manner, upon early FAS inhibition in chronic, but not acute, infection. Early cure of infection also increased mitochondrial volume in Tmem compared with Teff, supporting previous reports in acute infection. We demonstrate that the MPEC-specific effect was due to the higher fatty acid content and synthesis in MPEC compared with terminally differentiated Teff. In conclusion, FAS in CD4 T cells regulates the early divergence of Tmem from Teff in chronic infection.
Collapse
Affiliation(s)
- Samad A Ibitokou
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Brian E Dillon
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Mala Sinha
- Biomedical Informatics, Institute for Translational Science, University of Texas Medical Branch, Galveston, TX 77555
| | - Bartosz Szczesny
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555
| | | | | | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555
| | - Lutfi Abu-Elheiga
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Craig Porter
- Shriners Hospital for Children, Galveston, TX 77550
| | - Demidmaa Tuvdendorj
- Division of Endocrinology, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Robin Stephens
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; .,Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
24
|
Ford T, Wenden C, Mbekeani A, Dally L, Cox JH, Morin M, Winstone N, Hill AVS, Gilmour J, Ewer KJ. Cryopreservation-related loss of antigen-specific IFNγ producing CD4 + T-cells can skew immunogenicity data in vaccine trials: Lessons from a malaria vaccine trial substudy. Vaccine 2017; 35:1898-1906. [PMID: 28285985 PMCID: PMC5387668 DOI: 10.1016/j.vaccine.2017.02.038] [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: 09/16/2016] [Revised: 01/31/2017] [Accepted: 02/18/2017] [Indexed: 12/19/2022]
Abstract
Ex vivo functional immunoassays such as ELISpot and intracellular cytokine staining (ICS) by flow cytometry are crucial tools in vaccine development both in the identification of novel immunogenic targets and in the immunological assessment of samples from clinical trials. Cryopreservation and subsequent thawing of PBMCs via validated processes has become a mainstay of clinical trials due to processing restrictions inherent in the disparate location and capacity of trial centres, and also in the need to standardize biological assays at central testing facilities. Logistical and financial requirement to batch process samples from multiple study timepoints are also key. We used ELISpot and ICS assays to assess antigen-specific immunogenicity in blood samples taken from subjects enrolled in a phase II malaria heterologous prime-boost vaccine trial and showed that the freeze thaw process can result in a 3–5-fold reduction of malaria antigen-specific IFNγ-producing CD3+CD4+ effector populations from PBMC samples taken post vaccination. We have also demonstrated that peptide responsive CD8+ T cells are relatively unaffected, as well as CD4+ T cell populations that do not produce IFNγ. These findings contribute to a growing body of data that could be consolidated and synthesised as guidelines for clinical trials with the aim of increasing the efficiency of vaccine development pipelines.
Collapse
Affiliation(s)
- Tom Ford
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK.
| | - Claire Wenden
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Alison Mbekeani
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Len Dally
- EMMES Corporation, Rockville, MD, USA
| | - Josephine H Cox
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK
| | | | - Nicola Winstone
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Adrian V S Hill
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford, UK
| | - Jill Gilmour
- IAVI-HIL, Human Immunology Laboratory, International AIDS Vaccine Initiative, London, UK; Department of Medicine, Imperial College, London, UK
| | - Katie J Ewer
- Centre for Clinical Vaccinology and Tropical Medicine and the Jenner Institute Laboratories, University of Oxford, UK
| |
Collapse
|
25
|
Chronic Plasmodium chabaudi Infection Generates CD4 Memory T Cells with Increased T Cell Receptor Sensitivity but Poor Secondary Expansion and Increased Apoptosis. Infect Immun 2017; 85:IAI.00744-16. [PMID: 28031266 DOI: 10.1128/iai.00744-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/20/2016] [Indexed: 01/13/2023] Open
Abstract
Exposure to blood-stage malaria infection is often persistent, leading to generation of CD4 effector and effector memory T cells that contribute to protection. We showed previously that chronic exposure to blood-stage Plasmodium chabaudi offers the best protection from parasitemia and pathology in reinfection cases, correlating with an increase in Th1 cells. Although much is known about the features of resting or exhausted memory T cells (Tmem), little is known about the functional capacities of chronically stimulated but protective T cells. To determine the functional capacity of CD4 T cells generated by chronic infection upon reexposure to parasite, we compared their responses to known features of classical Tmem. The numbers of cytokine-producing T cells increased following infection in the polyclonal populations, suggesting an increase in pathogen-specific T cells. Malaria antigen-specific B5 T cell receptor (TCR) transgenic (Tg) T cells from chronic infection proliferated on reinfection and were highly sensitive to TCR stimulation without costimulation, as shown for Tmem in acute stimulations. However, B5 Tmem did not accumulate more than naive B5 T cells in vivo or in vitro and became apoptotic. Failure to accumulate was partly the result of chronic stimulation, since eliminating persistent parasites before reinfection slightly increased the accumulation of B5 Tg T cells upon reinfection. The levels of specific gamma interferon-positive, interleukin-10-positive T cells, which protect animals from pathology, increased after malaria infection. These data demonstrate that although chronic infection generates a protective T cell population with increased TCR sensitivity and cytokine production, they do not reexpand upon reexposure due to increased apoptosis.
Collapse
|