1
|
Ye C, Clements SA, Gu W, Geurts AM, Mathews CE, Serreze DV, Chen YG, Driver JP. Deletion of Vβ3 +CD4 + T cells by endogenous mouse mammary tumor virus 3 prevents type 1 diabetes induction by autoreactive CD8 + T cells. Proc Natl Acad Sci U S A 2023; 120:e2312039120. [PMID: 38015847 PMCID: PMC10710095 DOI: 10.1073/pnas.2312039120] [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: 07/18/2023] [Accepted: 09/23/2023] [Indexed: 11/30/2023] Open
Abstract
In both humans and NOD mice, type 1 diabetes (T1D) develops from the autoimmune destruction of pancreatic beta cells by T cells. Interactions between both helper CD4+ and cytotoxic CD8+ T cells are essential for T1D development in NOD mice. Previous work has indicated that pathogenic T cells arise from deleterious interactions between relatively common genes which regulate aspects of T cell activation/effector function (Ctla4, Tnfrsf9, Il2/Il21), peptide presentation (H2-A g7, B2m), and T cell receptor (TCR) signaling (Ptpn22). Here, we used a combination of subcongenic mapping and a CRISPR/Cas9 screen to identify the NOD-encoded mammary tumor virus (Mtv)3 provirus as a genetic element affecting CD4+/CD8+ T cell interactions through an additional mechanism, altering the TCR repertoire. Mtv3 encodes a superantigen (SAg) that deletes the majority of Vβ3+ thymocytes in NOD mice. Ablating Mtv3 and restoring Vβ3+ T cells has no effect on spontaneous T1D development in NOD mice. However, transferring Mtv3 to C57BL/6 (B6) mice congenic for the NOD H2 g7 MHC haplotype (B6.H2 g7) completely blocks their normal susceptibility to T1D mediated by transferred CD8+ T cells transgenically expressing AI4 or NY8.3 TCRs. The entire genetic effect is manifested by Vβ3+CD4+ T cells, which unless deleted by Mtv3, accumulate in insulitic lesions triggering in B6 background mice the pathogenic activation of diabetogenic CD8+ T cells. Our findings provide evidence that endogenous Mtv SAgs can influence autoimmune responses. Furthermore, since most common mouse strains have gaps in their TCR Vβ repertoire due to Mtvs, it raises questions about the role of Mtvs in other mouse models designed to reflect human immune disorders.
Collapse
Affiliation(s)
- Cheng Ye
- Department of Animal Sciences, University of Florida, Gainesville, FL32611
| | - Sadie A. Clements
- Division of Animal Sciences, University of Missouri, Columbia, MO65201
| | - Weihong Gu
- Division of Animal Sciences, University of Missouri, Columbia, MO65201
| | - Aron M. Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI53226
| | - Clayton E. Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL32610
| | | | - Yi-Guang Chen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI53226
| | - John P. Driver
- Division of Animal Sciences, University of Missouri, Columbia, MO65201
| |
Collapse
|
2
|
An opinion on the debatable function of brain resident immune protein, T-cell receptor beta subunit in the central nervous system. IBRO Neurosci Rep 2022; 13:235-242. [PMID: 36590097 PMCID: PMC9795316 DOI: 10.1016/j.ibneur.2022.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/02/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years scientific research has established that the nervous and immune systems have shared molecular signaling components. Proteins native to immune cells, which are also found in the brain, have neuronal functions in the nervous system where they affect synaptic plasticity, axonal regeneration, neurogenesis, and neurotransmission. Certain native immune molecules like major histocompatibility complex I (MHC-I), paired immunoglobulin receptor B (PirB), toll-like receptor (TLR), cluster of differentiation-3 zeta (CD3ζ), CD4 co-receptor, and T-cell receptor beta (TCR-β) expression in neurons have been extensively documented. In this review, we provide our opinion and discussed the possible roles of T-cell receptor beta subunits in modulating the function of neurons in the central nervous system. Based on the previous findings of Syken and Shatz., 2003; Nishiyori et al., 2004; Rodriguez et., 1993 and Komal et., 2014; we discuss whether restrictive expression of TCR-β subunits in selected brain regions could be involved in the pathology of neurological disorders and whether their aberrant enhancement in expression may be considered as a suitable biomarker for aging or neurodegenerative diseases like Huntington's disease (HD).
Collapse
|
3
|
A TCRβ Repertoire Signature Can Predict Experimental Cerebral Malaria. PLoS One 2016; 11:e0147871. [PMID: 26844551 PMCID: PMC4742225 DOI: 10.1371/journal.pone.0147871] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/04/2015] [Indexed: 11/19/2022] Open
Abstract
Cerebral Malaria (CM) is associated with a pathogenic T cell response. Mice infected by P. berghei ANKA clone 1.49 (PbA) developing CM (CM+) present an altered PBL TCR repertoire, partly due to recurrently expanded T cell clones, as compared to non-infected and CM- infected mice. To analyse the relationship between repertoire alteration and CM, we performed a kinetic analysis of the TRBV repertoire during the course of the infection until CM-related death in PbA-infected mice. The repertoires of PBL, splenocytes and brain lymphocytes were compared between infected and non-infected mice using a high-throughput CDR3 spectratyping method. We observed a modification of the whole TCR repertoire in the spleen and blood of infected mice, from the fifth and the sixth day post-infection, respectively, while only three TRBV were significantly perturbed in the brain of infected mice. Using multivariate analysis and statistical modelling, we identified a unique TCRβ signature discriminating CM+ from CTR mice, enriched during the course of the infection in the spleen and the blood and predicting CM onset. These results highlight a dynamic modification and compartmentalization of the TCR diversity during the course of PbA infection, and provide a novel method to identify disease-associated TCRβ signature as diagnostic and prognostic biomarkers.
Collapse
|
4
|
Hernandez-Valladares M, Rihet P, Iraqi FA. Host susceptibility to malaria in human and mice: compatible approaches to identify potential resistant genes. Physiol Genomics 2014; 46:1-16. [DOI: 10.1152/physiolgenomics.00044.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
There is growing evidence for human genetic factors controlling the outcome of malaria infection, while molecular basis of this genetic control is still poorly understood. Case-control and family-based studies have been carried out to identify genes underlying host susceptibility to malarial infection. Parasitemia and mild malaria have been genetically linked to human chromosomes 5q31-q33 and 6p21.3, and several immune genes located within those regions have been associated with malaria-related phenotypes. Association and linkage studies of resistance to malaria are not easy to carry out in human populations, because of the difficulty in surveying a significant number of families. Murine models have proven to be an excellent genetic tool for studying host response to malaria; their use allowed mapping 14 resistance loci, eight of them controlling parasitic levels and six controlling cerebral malaria. Once quantitative trait loci or genes have been identified, the human ortholog may then be identified. Comparative mapping studies showed that a couple of human and mouse might share similar genetically controlled mechanisms of resistance. In this way, char8, which controls parasitemia, was mapped on chromosome 11; char8 corresponds to human chromosome 5q31-q33 and contains immune genes, such as Il3, Il4, Il5, Il12b, Il13, Irf1, and Csf2. Nevertheless, part of the genetic factors controlling malaria traits might differ in both hosts because of specific host-pathogen interactions. Finally, novel genetic tools including animal models were recently developed and will offer new opportunities for identifying genetic factors underlying host phenotypic response to malaria, which will help in better therapeutic strategies including vaccine and drug development.
Collapse
Affiliation(s)
| | - Pascal Rihet
- UMR1090 TAGC, INSERM, Marseille, France
- Aix-Marseille University, Marseille, France; and
| | - Fuad A. Iraqi
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| |
Collapse
|
5
|
Holt MP, Shevach EM, Punkosdy GA. Endogenous mouse mammary tumor viruses (mtv): new roles for an old virus in cancer, infection, and immunity. Front Oncol 2013; 3:287. [PMID: 24324930 PMCID: PMC3840357 DOI: 10.3389/fonc.2013.00287] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 11/10/2013] [Indexed: 12/31/2022] Open
Abstract
Mouse Mammary Tumor Viruses are beta-retroviruses that exist in both exogenous (MMTV) and endogenous (Mtv) forms. Exogenous MMTV is transmitted via the milk of lactating animals and is capable of inducing mammary gland tumors later in life. MMTV has provided a number of critical models for studying both viral infection as well as human breast cancer. In addition to the horizontally transmitted MMTV, most inbred mouse strains contain permanently integrated Mtv proviruses within their genome that are remnants of MMTV infection and vertically transmitted. Historically, Mtv have been appreciated for their role in shaping the T cell repertoire during thymic development via negative selection. In addition, more recent work has demonstrated a larger role for Mtv in modulating host immune responses due to its peripheral expression. The influence of Mtv on host response has been observed during experimental murine models of Polyomavirus- and ESb-induced lymphoma as well as Leishmania major and Plasmodium berghei ANKA infection. Decreased susceptibility to bacterial pathogens and virus-induced tumors has been observed among mice lacking all Mtv. We have also demonstrated a role for Mtv Sag in the expansion of regulatory T cells following chronic viral infection. The aim of this review is to summarize the latest research in the field regarding peripheral expression of Mtv with a particular focus on their role and influence on the immune system, infectious disease outcome, and potential involvement in tumor formation.
Collapse
Affiliation(s)
- Michael P Holt
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD , USA
| | | | | |
Collapse
|
6
|
Rénia L, Potter SM, Mauduit M, Rosa DS, Kayibanda M, Deschemin JC, Snounou G, Grüner AC. Pathogenic T cells in cerebral malaria. Int J Parasitol 2006; 36:547-54. [PMID: 16600241 DOI: 10.1016/j.ijpara.2006.02.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2005] [Revised: 02/01/2006] [Accepted: 02/10/2006] [Indexed: 11/24/2022]
Abstract
Malaria remains a major global health problem and cerebral malaria (CM) is one of the most serious complications of this disease. Recent years have seen important advances in our understanding of the pathogenesis of cerebral malaria. Parasite sequestration, a hallmark of this syndrome, is thought to be solely responsible for the pathological process. However, this phenomenon cannot explain all aspects of the pathogenesis of CM. The use of an animal model, Plasmodium berghei ANKA in mice, has allowed the identification of specific pathological components of CM. Although multiple pathways may lead to CM, an important role for CD8+ T cells has been clarified. Other cells, including platelets, and mediators such as cytokines also have an important role. In this review we have focused on the role of T cells, and discuss what remains to be studied to understand the pathways by which these cells mediate CM.
Collapse
Affiliation(s)
- Laurent Rénia
- Department of Immunology, Institut Cochin, INSERM U567, CNRS UMR 8104, Université René Descartes, Hôpital Cochin, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Bakir HY, Tomiyama-Miyaji C, Watanabe H, Nagura T, Kawamura T, Sekikawa H, Abo T. Reasons why DBA/2 mice are resistant to malarial infection: expansion of CD3int B220+ gammadelta T cells with double-negative CD4- CD8- phenotype in the liver. Immunology 2006; 117:127-35. [PMID: 16423048 PMCID: PMC1782202 DOI: 10.1111/j.1365-2567.2005.02273.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
DBA/2 (H-2(d)) mice are known to be more resistant than C57BL/6 (B6, H-2(b)) mice to the non-lethal 17XNL strain of Plasmodium yoelii. This is a very strange phenomenon because the functions of conventional T cells, especially CD8(+) T cells, are known to be somewhat lower in DBA/2 mice than in other strains of mice. We examined herein how immune responses differed between DBA/2 mice and B6 mice during malarial infection. DBA/2 mice and (DBA/2 x B6)F(1) (BDF(1), H-2(b/d)) mice were found to have milder parasitaemia and to recover more quickly from malarial infection than B6 mice. These DBA/2 and BDF(1) mice were also found to experience a marked expansion of interleukin (IL)-2Rbeta(+) CD3(int) cells and gammadelta T cells in the liver, especially in the recovery phase. The expansion of unconventional T cells (i.e. B220(+) T cells) was also marked in DBA/2 and BDF(1) mice. The majority of B220(+) T cells were gammadelta T cells and these T cells were double-negative CD4(-) CD8(-). More importantly, the production of immunoglobulin M (IgM)-type anti-DNA autoantibody was also higher in DBA/2 and BDF(1) mice than in B6 mice. In conjunction with data on cytokine production, these results indicate that primitive T and B cells, namely autoreactive extrathymic T cells and autoantibody-producing B cells, may be much more activated in DBA/2 mice and therefore resistant to the non-lethal 17XNL strain of P. yoelii.
Collapse
MESH Headings
- Animals
- Antibodies, Antinuclear/biosynthesis
- Antibodies, Monoclonal/immunology
- CD3 Complex/analysis
- Cytokines/biosynthesis
- Immunity, Innate
- Immunoglobulin M/biosynthesis
- Immunophenotyping
- Leukocyte Common Antigens/analysis
- Liver/immunology
- Malaria/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Parasitemia/immunology
- Plasmodium yoelii
- Receptors, Antigen, T-Cell, gamma-delta/analysis
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
Collapse
Affiliation(s)
- Hanaa Y Bakir
- Department of Immunology, Niigata University School of Medicine, Niigata, Japan
| | | | | | | | | | | | | |
Collapse
|
8
|
Engwerda C, Belnoue E, Grüner AC, Rénia L. ExperimentalModels of Cerebral Malaria. Curr Top Microbiol Immunol 2005. [DOI: 10.1007/3-540-29967-x_4] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
9
|
Collette A, Bagot S, Ferrandiz ME, Cazenave PA, Six A, Pied S. A profound alteration of blood TCRB repertoire allows prediction of cerebral malaria. THE JOURNAL OF IMMUNOLOGY 2004; 173:4568-75. [PMID: 15383590 DOI: 10.4049/jimmunol.173.7.4568] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cerebral malaria (CM) is one of the severe complications of Plasmodium infection. In murine models of CM, Talphabeta cells have been implicated in the neuropathogenesis. To obtain insights into the TCRB repertoire during CM, we used high throughput CDR3 spectratyping and set up new methods and software tools to analyze data. We compared PBL and spleen repertoires of mice infected with Plasmodium berghei ANKA that developed CM (CM(+)) or not (CM(-)) to evidence modifications of the TCRB repertoire associated with neuropathology. Using distinct statistical multivariate methods, the PBL repertoires of CM(+) mice were found to be specifically altered. This alteration is partly due to recurrently expanded T cell clones. Strikingly, alteration of the PBL repertoire can be used to distinguish between CM(+) and CM(-). This study provides the first ex vivo demonstration of modifications of Talphabeta cell compartment during CM. Finally, our original approach for deciphering lymphocyte repertoires can be transposed to various pathological conditions.
Collapse
MESH Headings
- Animals
- Cell Separation
- Clone Cells
- Complementarity Determining Regions/biosynthesis
- Complementarity Determining Regions/blood
- Complementarity Determining Regions/genetics
- Female
- Immunoglobulin Constant Regions/biosynthesis
- Immunoglobulin Constant Regions/blood
- Immunoglobulin Constant Regions/genetics
- Immunoglobulin Variable Region/biosynthesis
- Immunoglobulin Variable Region/blood
- Immunoglobulin Variable Region/genetics
- Malaria, Cerebral/genetics
- Malaria, Cerebral/immunology
- Malaria, Cerebral/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Plasmodium berghei/immunology
- Plasmodium berghei/pathogenicity
- Polymerase Chain Reaction/methods
- Predictive Value of Tests
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/blood
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Recurrence
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- Alexis Collette
- Immunophysiopathologie Infectieuse, Centre National de la Recherche Scientifique Unité de Recherche Associée 1961, Institut Pasteur, and Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | |
Collapse
|
10
|
Bagot S, Nogueira F, Collette A, do Rosario V, Lemonier F, Cazenave PA, Pied S. Comparative study of brain CD8+ T cells induced by sporozoites and those induced by blood-stage Plasmodium berghei ANKA involved in the development of cerebral malaria. Infect Immun 2004; 72:2817-26. [PMID: 15102792 PMCID: PMC387860 DOI: 10.1128/iai.72.5.2817-2826.2004] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To obtain insight into the mechanisms that contribute to the pathogenesis of Plasmodium infections, we developed an improved rodent model that mimics human malaria closely by inducing cerebral malaria (CM) through sporozoite infection. We used this model to carry out a detailed study on isolated T cells recruited from the brains of mice during the development of CM. We compared several aspects of the immune response related to the experimental model of Plasmodium berghei ANKA infection induced by sporozoites in C57BL/6 mice and those related to a blood-stage infection. Our data show that in both models, oligoclonal TCRVbeta4(+), TCRVbeta6(+), TCRVbeta8.1(+), and TCRVbeta11(+) major histocompatibility complex class I-restricted CD8 T cells were present in the brains of CM(+) mice. These CD8(+) T cells display an activated phenotype, do not undergo apoptosis, secrete gamma interferon or tumor necrosis factor alpha, and are associated with the development of the neurological syndrome.
Collapse
Affiliation(s)
- Sébastien Bagot
- Unité d'Immunophysiopathologie Infectieuse, CNRS URA 1961, Université Pierre et Marie Curie, Paris, France
| | | | | | | | | | | | | |
Collapse
|
11
|
Kang H, Liesenfeld O, Remington JS, Claflin J, Wang X, Suzuki Y. TCR V beta 8+ T cells prevent development of toxoplasmic encephalitis in BALB/c mice genetically resistant to the disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4254-9. [PMID: 12682259 DOI: 10.4049/jimmunol.170.8.4254] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BALB/c are genetically resistant to development of toxoplasmic encephalitis (TE) when infected with Toxoplasma gondii, whereas CBA/Ca mice are susceptible. We compared TCR Vbeta chain usage in lymphocytes infiltrated into brains between these animals following infection. TCR Vbeta8(+) cells were the most frequent T cell population in brains of infected, resistant BALB/c mice, whereas TCR Vbeta6(+) T cells were more prevalent than Vbeta8(+) T cells in brains of infected, susceptible CBA/Ca mice. Adoptive transfer of Vbeta8(+) immune T cells, obtained from infected BALB/c mice, prevented development of TE and mortality in infected athymic nude mice that lack T cells. In contrast, adoptive transfer of Vbeta6(+) immune T cells did not prevent development of TE or mortality in the nude mice. The protective activity of Vbeta8(+) immune T cells was greater than that of the total Vbeta8(-) population. In addition, Vbeta8(+) immune T cells produced markedly greater amounts of IFN-gamma than did the Vbeta8(-) population after stimulation with tachyzoite lysate Ags in vitro. Thus, Vbeta8(+) T cells appear to play a crucial role in the genetic resistance of BALB/c mice against development of TE.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Biomarkers/analysis
- Brain/immunology
- Brain/pathology
- Cell Movement/genetics
- Cell Movement/immunology
- Encephalitis/genetics
- Encephalitis/immunology
- Encephalitis/pathology
- Encephalitis/prevention & control
- Female
- Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Genetic Predisposition to Disease
- Immunity, Innate/genetics
- Interferon-gamma/biosynthesis
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Count
- Mice
- Mice, Inbred BALB C
- Mice, Inbred CBA
- Mice, Nude
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Toxoplasma/immunology
- Toxoplasmosis, Animal/genetics
- Toxoplasmosis, Animal/immunology
- Toxoplasmosis, Animal/pathology
- Toxoplasmosis, Animal/prevention & control
- Toxoplasmosis, Cerebral/genetics
- Toxoplasmosis, Cerebral/immunology
- Toxoplasmosis, Cerebral/pathology
- Toxoplasmosis, Cerebral/prevention & control
Collapse
Affiliation(s)
- Hoil Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|