Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
JCI Insight ; 8(15)2023 08 08.
Article in English | MEDLINE | ID: mdl-37338983

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder that causes debilitating swelling and destruction of the joints. People with RA are treated with drugs that actively suppress one or more parts of their immune system, and these may alter the response to vaccination against SARS-CoV-2. In this study, we analyzed blood samples from a cohort of patients with RA after receiving a 2-dose mRNA COVID-19 vaccine regimen. Our data show that individuals on the cytotoxic T lymphocyte antigen 4-Ig therapy abatacept had reduced levels of SARS-CoV-2-neutralizing antibodies after vaccination. At the cellular level, these patients showed reduced activation and class switching of SARS-CoV-2-specific B cells, as well as reduced numbers and impaired helper cytokine production by SARS-CoV-2-specific CD4+ T cells. Individuals on methotrexate showed similar but less severe defects in vaccine response, whereas individuals on the B cell-depleting therapy rituximab had a near-total loss of antibody production after vaccination. These data define a specific cellular phenotype associated with impaired response to SARS-CoV-2 vaccination in patients with RA on different immune-modifying therapies and help inform efforts to improve vaccination strategies in this vulnerable population.


Subject(s)
Arthritis, Rheumatoid , COVID-19 , Humans , COVID-19 Vaccines , COVID-19/prevention & control , SARS-CoV-2 , Arthritis, Rheumatoid/drug therapy , Antibodies, Viral , RNA, Messenger
2.
JCI Insight ; 7(12)2022 06 22.
Article in English | MEDLINE | ID: mdl-35587373

ABSTRACT

The transcription factor STAT1 plays a critical role in modulating the differentiation of CD4+ T cells producing IL-17 and GM-CSF, which promote the development of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). The protective role of STAT1 in MS and EAE has been largely attributed to its ability to limit pathogenic Th cells and promote Tregs. Using mice with selective deletion of STAT1 in T cells (STAT1CD4-Cre), we identified a potentially novel mechanism by which STAT1 regulates neuroinflammation independently of Foxp3+ Tregs. STAT1-deficient effector T cells became the target of NK cell-mediated killing, limiting their capacity to induce EAE. STAT1-deficient T cells promoted their own killing by producing more IL-2 that, in return, activated NK cells. Elimination of NK cells restored EAE susceptibility in STAT1CD4-Cre mice. Therefore, our study suggests that the STAT1 pathway can be manipulated to limit autoreactive T cells during autoimmunity directed against the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Autoimmunity , CD4-Positive T-Lymphocytes , Mice , Neuroinflammatory Diseases , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
3.
J Immunol ; 208(7): 1519-1524, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35288472

ABSTRACT

Multiple sclerosis (MS) is a demyelinating inflammatory disease of the CNS treated by diverse disease-modifying therapies that suppress the immune system. Severe acute respiratory syndrome coronavirus 2 mRNA vaccines have been very effective in immunocompetent individuals, but whether MS patients treated with modifying therapies are afforded the same protection is not known. This study determined that dimethyl fumarate caused a momentary reduction in anti-Spike (S)-specific Abs and CD8 T cell response. MS patients treated with B cell-depleting (anti-CD20) or sphingosine 1-phosphate receptor agonist (fingolimod) therapies lack significant S-specific Ab response. Whereas S-specific CD4 and CD8 T cell responses were largely compromised by fingolimod treatment, T cell responses were robustly generated in anti-CD20-treated MS patients, but with a reduced proportion of CD4+CXCR5+ circulating follicular Th cells. These data provide novel information regarding vaccine immune response in patients with autoimmunity useful to help improve vaccine effectiveness in these populations.


Subject(s)
COVID-19 , Multiple Sclerosis , COVID-19 Vaccines , Humans , Immunologic Memory , Multiple Sclerosis/drug therapy , SARS-CoV-2
4.
J Immunol ; 207(10): 2417-2422, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34663621

ABSTRACT

Dedicator of cytokinesis 8 (DOCK8) is a guanine nucleotide exchange factor with an essential role in cytoskeletal rearrangement, cell migration, and survival of various immune cells. Interestingly, DOCK8-deficient mice are resistant to the development of experimental autoimmune encephalomyelitis (EAE). To understand if EAE resistance in these mice results from an alteration in dendritic cell (DC) functions, we generated mice with conditional deletion of DOCK8 in DCs and observed attenuated EAE in these mice compared with control mice. Additionally, we demonstrated that DOCK8 is important for the existence of splenic conventional DC2 and lymph node migratory DCs and further established that migratory DC, rather than resident DC, are essential for the generation and proliferation of pathogenic T cell populations upon immunization with myelin Ag in adjuvant. Therefore, our data suggest that limiting migratory DCs through DOCK8 deletion and possibly other mechanisms could limit the development of CNS autoimmunity.


Subject(s)
Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Guanine Nucleotide Exchange Factors/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL
6.
J Immunol ; 202(5): 1373-1382, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30683697

ABSTRACT

Abatacept is a CTLA-4-Ig fusion protein that binds to the costimulatory ligands CD80 and CD86 and blocks their interaction with the CD28 and CTLA-4 receptors expressed by T cells, therefore inhibiting T cell activation and function. Abatacept has shown clinical efficacy in treating some autoimmune diseases but has failed to show clinical benefit in other autoimmune conditions. The reasons for these disparate results are not clear and warrant further investigation of abatacept's mode of action. Longitudinal specimens from the Immune Tolerance Network's A Cooperative Clinical Study of Abatacept in Multiple Sclerosis trial were used to examine the effects of abatacept treatment on the frequency and transcriptional profile of specific T cell populations in peripheral blood. We found that the relative abundance of CD4+ T follicular helper (Tfh) cells and regulatory T cells was selectively decreased in participants following abatacept treatment. Within both cell types, abatacept reduced the proportion of activated cells expressing CD38 and ICOS and was associated with decreased expression of genes that regulate cell-cycle and chromatin dynamics during cell proliferation, thereby linking changes in costimulatory signaling to impaired activation, proliferation, and decreased abundance. All cellular and molecular changes were reversed following termination of abatacept treatment. These data expand upon the mechanism of action of abatacept reported in other autoimmune diseases and identify new transcriptional targets of CD28-mediated costimulatory signaling in human regulatory T and Tfh cells, further informing on its potential use in diseases associated with dysregulated Tfh activity.


Subject(s)
Abatacept/pharmacology , Immunosuppressive Agents/pharmacology , Multiple Sclerosis/drug therapy , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Cell Proliferation/drug effects , Double-Blind Method , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
7.
Curr Opin Immunol ; 55: 38-43, 2018 12.
Article in English | MEDLINE | ID: mdl-30268837

ABSTRACT

CD4+ T helper (Th) cells play a central role in orchestrating protective immunity but also in autoimmunity. Multiple Sclerosis (MS) is a human autoimmune disease of the central nervous system (CNS) characterized by the infiltration of inflammatory lymphocytes and myeloid cells into the brain and spinal cord, leading to demyelination, axonal damage, and progressive loss of motor functions. The release of T cells in the circulation and their migration in the central nervous system are key and tightly regulated processes which have been targeted to decrease CD4+ T cell presence in the CNS and limit disease progression. Here, we review two of these pathways and discuss how their blockade modulate different subsets of CD4+ T cells.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Animals , Humans
8.
Article in English | MEDLINE | ID: mdl-29311122

ABSTRACT

Multiple sclerosis (MS) is a multifocal demyelinating disease of the central nervous system (CNS) leading to the progressive destruction of the myelin sheath surrounding axons. It can present with variable clinical and pathological manifestations, which might reflect the involvement of distinct pathogenic processes. Although the mechanisms leading to the development of the disease are not fully understood, numerous evidences indicate that MS is an autoimmune disease, the initiation and progression of which are dependent on an autoimmune response against myelin antigens. In addition, genetic susceptibility and environmental triggers likely contribute to the initiation of the disease. At this time, there is no cure for MS, but several disease-modifying therapies (DMTs) are available to control and slow down disease progression. A good number of these DMTs were identified and tested using animal models of MS referred to as experimental autoimmune encephalomyelitis (EAE). In this review, we will recapitulate the characteristics of EAE models and discuss how they help shed light on MS pathogenesis and help test new treatments for MS patients.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/physiopathology , Animals , Antigens/immunology , B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Multiple Sclerosis/immunology , Myelin Basic Protein/physiology , Myelin Proteolipid Protein/physiology , Myelin-Oligodendrocyte Glycoprotein/physiology
9.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978795

ABSTRACT

Foxp3+ Tregs possess potent immunosuppressive activity, which is critical for maintaining immune homeostasis and self-tolerance. Defects in Treg development or function result in inadvertent immune activation and autoimmunity. Despite recent advances in Treg biology, we still do not completely understand the molecular and cellular mechanisms governing the development and suppressive function of these cells. Here, we have demonstrated an essential role of the dedicator of cytokinesis 8 (DOCK8), guanine nucleotide exchange factors required for cytoskeleton rearrangement, cell migration, and immune cell survival in controlling Treg fitness and their function. Treg-specific DOCK8 deletion led to spontaneous multiorgan inflammation in mice due to uncontrolled T cell activation and production of proinflammatory cytokines. In addition, we show that DOCK8-deficient Tregs are defective in competitive fitness and in vivo suppressive function. Furthermore, DOCK8 controls IL-2 signaling, crucial for maintenance and competitive fitness of Tregs, via a STAT5-dependent manner. Our study provides potentially novel insights into the essential function of DOCK8 in Tregs and immune regulation, and it explains the autoimmune manifestations associated with DOCK8 deficiency.


Subject(s)
Guanine Nucleotide Exchange Factors/immunology , Interleukin-2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Guanine Nucleotide Exchange Factors/deficiency , Immune Tolerance/immunology , Inflammation/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation/immunology , Mice, Knockout , STAT5 Transcription Factor/immunology , Signal Transduction/immunology
10.
Sci Rep ; 7(1): 12905, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29018225

ABSTRACT

Sphingosine-1 phosphate receptor 1 (S1P1) is critical for the egress of T and B cells out of lymphoid organs. Although S1P1 agonist fingolimod is currently used for the treatment of multiple sclerosis (MS) little is known how S1P1 signaling regulates Th17 and Treg cell homeostasis. To study the impact of S1P1 signaling on Th17 and Treg cell biology, we specifically deleted S1P1 in Th17 and Treg cells using IL-17A Cre and Foxp3 Cre mice, respectively. Deletion of S1P1 in Th17 cells conferred resistance to experimental autoimmune encephalomyelitis (EAE). On the other hand, permanent deletion of S1P1 in Treg cells resulted in autoimmunity and acute deletion rendered mice more susceptible to EAE. Importantly, our study revealed that S1P1 not only regulated the egress of Treg cells out of lymphoid organs and subsequent non-lymphoid tissue distribution but also their phenotypic diversity. Most of the Treg cells found in S1P1-deficient mice as well as MS patients on fingolimod therapy had an activated phenotype and were more prone to apoptosis, thus converted to effector Treg. Our results provide novel insight into the functions of S1P1 and potential impact of long term fingolimod use on Th17 and Treg cell biology and general health in MS patients.


Subject(s)
Gene Deletion , Receptors, Lysosphingolipid/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Autoimmunity , Case-Control Studies , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , Inflammation/pathology , Lymphadenopathy/pathology , Lymphoid Tissue/pathology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Organ Specificity , Phenotype , Receptors, Lysosphingolipid/metabolism , Transcription, Genetic
12.
J Immunol ; 196(9): 3542-6, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27016608

ABSTRACT

The neutralization of integrin α4 (Itga4) is currently used as treatment in multiple sclerosis. Although most studies have focused on its function on lymphocyte migration to the CNS, we have uncovered the importance of Itga4 for the generation of regulatory B cells in peripheral immune organs and their control of pathogenic T cell response and CNS pathology. Our study underscores the importance of looking at the dual role of B cells in CNS autoimmunity and provides important perspectives regarding the efficacy and side effects associated with Itga4 neutralization and other B cell-targeting therapies.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/immunology , Animals , B-Lymphocytes, Regulatory/physiology , Cell Movement , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Humans , Integrin alpha4/genetics , Mice , Multiple Sclerosis/immunology , T-Lymphocytes/immunology
13.
J Immunol ; 195(5): 1974-83, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26223651

ABSTRACT

IL-17-producing CD4(+) T (Th17) cells, along with IFN-γ-expressing Th1 cells, represent two major pathogenic T cell subsets in experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis (MS). The cytokines and transcription factors involved in the development and effector functions of Th1 and Th17 cells have been largely characterized. Among them, IL-23 is essential for the generation of stable and encephalitogenic Th17 cells and for the development of EAE. The IL-7/IL-7R signaling axis participates in cell survival, and perturbation of this pathway has been associated with enhanced susceptibility to MS. A link between IL-23-driven pathogenic T cells and IL-7/IL-7R signaling has previously been proposed, but has not been formally addressed. In the current study, we showed that Th17 cells from mice with EAE express high levels of IL-7Rα compared with Th1 cells. Using mice that constitutively express IL-7Rα on T cells, we determined that sustained IL-7R expression in IL-23R-deficient mice could not drive pathogenic T cells and the development of EAE. IL-7 inhibited the differentiation of Th17 cells, but promoted IFN-γ and GM-CSF secretion in vitro. In vivo IL-7/anti-IL-7 mAb complexes selectively expanded and enhanced the proliferation of CXCR3-expressing Th1 cells, but did not impact Th17 cells and EAE development in wild-type and IL-23R-deficient mice. Importantly, high IL-7 expression was detected in the CNS during EAE and could drive the plasticity of Th17 cells to IFN-γ-producing T cells. Together, these data address the contribution of IL-23/IL-23R and IL-7/IL-7R signaling in Th17 and Th1 cell dynamics during CNS autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-7/immunology , Receptors, Interleukin-7/immunology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Flow Cytometry , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-7/genetics , Interleukin-7/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Receptors, Interleukin/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Signal Transduction/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
14.
Sci Rep ; 5: 7834, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25592296

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis (MS), results from an autoimmune attack of the central nervous system (CNS) by effector T helper (Th) 1 and Th17 cells. Regulatory T cells (Treg) can control effector T cells and limit the progression of CNS autoimmunity. Integrin alpha 4 (Itga4) is critical for the entry of Th1 but not Th17 cells into the CNS during EAE. Whether Itga4 controls the homing of Tregs in the CNS and whether Tregs can limit Th17-mediated EAE has, however, not been addressed. Through selective elimination of Itga4 in Foxp3-expressing cells, we show here that Tregs can suppress Th17-mediated EAE and enter into the CNS independently of Itga4. Furthermore, similarly to Th17 cells and in contrast to Th1 cells, Tregs depend on LFA-1 for their entry into the CNS in the absence of Itga4. Therefore, these data suggest that the efficacy of Itga4 neutralization on MS progression may be associated with the prevention of Th1 cells and the maintenance of Tregs migration into the CNS.


Subject(s)
Autoimmunity , CD11a Antigen/metabolism , Central Nervous System/metabolism , Integrin alpha4/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD11a Antigen/genetics , Cell Differentiation , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Forkhead Transcription Factors/metabolism , Integrin alpha4/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
15.
J Clin Invest ; 125(1): 129-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25415436

ABSTRACT

Podoplanin (PDPN, also known as Gp38) is highly expressed on the surface of lymphatic endothelial cells, where it regulates development of lymphatic vessels. We have recently observed that PDPN is also expressed on effector T cells that infiltrate target tissues during autoimmune inflammation; however, the function of PDPN in T cells is largely unclear. Here, we demonstrated that global deletion of Pdpn results in exaggerated T cell responses and spontaneous experimental autoimmune encephalomyelitis (EAE) in mice with a susceptible genetic background. In contrast, T cell-specific overexpression of PDPN resulted in profound defects in IL-7-mediated T cell expansion and survival. Consequently, these animals exhibited a more rapid resolution of CNS inflammation, characterized by a reduced effector CD4+ T cell population in the CNS. Mice harboring a T cell-specific deletion of Pdpn developed exacerbated EAE, with increased accumulation of effector CD4+ T cells in the CNS. Transcriptional profiling of naturally occurring PDPN+ effector T cells in the CNS revealed increased expression of other inhibitory receptors, such as Pd1 and Tim3, and decreased expression of prosurvival factors, including Il7ra. Together, our data suggest that PDPN functions as an inhibitory molecule on T cells, thereby promoting tissue tolerance by limiting long-term survival and maintenance of CD4+ effector T cells in target organs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Membrane Glycoproteins/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Survival , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Lymphopenia/immunology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout
16.
Nat Commun ; 5: 4603, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25091235

ABSTRACT

Retinoic acid receptor-related orphan receptor-γt-positive (RORγt(+)) innate lymphoid cells (ILCs) produce interleukin (IL)-22 and IL-17, which are critical for protective immunity against enteric pathogens. The molecular mechanism underlying the development and survival of RORγt(+) ILCs is not thoroughly understood. Here, we show that Dedicator of cytokinesis 8 (DOCK8), a scaffolding protein involved in cytoskeletal rearrangement and cell migration, is essential for the protective immunity against Citrobacter rodentium. A comparative RNA sequencing-based analysis reveals an impaired induction of antimicrobial peptides in the colon of DOCK8-deficient mice, which correlates with high susceptibility to infection and a very low number of IL-22-producing RORγt(+) ILCs in their GI tract. Furthermore, DOCK8-deficient RORγt(+) ILCs are less responsive to IL-7 mediated signalling, more prone to apoptosis and produce less IL-22 due to a defect in IL-23-mediated STAT3 phosphorylation. Our studies reveal an unsuspected role of DOCK8 for the function, generation and survival of RORγt(+) ILCs.


Subject(s)
Enterobacteriaceae Infections/immunology , Guanine Nucleotide Exchange Factors/immunology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Animals , Apoptosis , Bone Marrow Cells/cytology , Cell Survival , Citrobacter rodentium , Colon/immunology , Colon/metabolism , Female , Interleukin-17/metabolism , Interleukins/metabolism , Interleukins/pharmacology , Lymphocytes/microbiology , Male , Mice , Mice, Knockout , Phosphorylation , Sequence Analysis, RNA , Interleukin-22
17.
J Immunol ; 190(9): 4478-82, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23543757

ABSTRACT

During the development of experimental autoimmune encephalomyelitis (EAE), the proportion of pathogenic and myelin-specific cells within CNS-infiltrating cytokine-producing Th cells is unknown. Using an IL-17A/IFN-γ double reporter mouse and I-A(b)/myelin oligodendrocyte glycoprotein 38-49 tetramer, we show in this study that IL-17(+)IFN-γ(+) Th cells, which are expanded in the CNS during EAE, are highly enriched in myelin oligodendrocyte glycoprotein-specific T cells. We further demonstrate that IL-23 is essential for the generation and expansion of IFN-γ-producing Th17 cells independently of the Th1-associated transcription factors T-bet, STAT1, and STAT4. Furthermore, Th17 and IL-17(+)IFN-γ(+) Th cells can induce CNS autoimmunity independently of T-bet. Whereas T-bet is crucial for Th1-mediated EAE, it is dispensable for Th17 cell-mediated autoimmunity. Our results suggest the existence of different epigenetic programs that regulate IFN-γ expression in Th1 and Th17 cells.


Subject(s)
Interferon-gamma/immunology , Interleukin-17/immunology , T-Box Domain Proteins/immunology , Th17 Cells/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/metabolism , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , STAT4 Transcription Factor/immunology , STAT4 Transcription Factor/metabolism , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/metabolism
18.
Immunity ; 35(6): 986-96, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22177922

ABSTRACT

Ectopic lymphoid follicles are hallmarks of chronic autoimmune inflammatory diseases such as multiple sclerosis (MS), rheumatoid arthritis, Sjögren's syndrome, and myasthenia gravis. However, the effector cells and mechanisms that induce their development are unknown. Here we showed that in experimental autoimmune encephalomyelitis (EAE), the animal model of MS, Th17 cells specifically induced ectopic lymphoid follicles in the central nervous system (CNS). Development of ectopic lymphoid follicles was partly dependent on the cytokine interleukin 17 (IL-17) and on the cell surface molecule Podoplanin (Pdp), which was expressed on Th17 cells, but not on other effector T cell subsets. Pdp was also crucial for the development of secondary lymphoid structures: Pdp-deficient mice lacked peripheral lymph nodes and had a defect in forming normal lymphoid follicles and germinal centers in spleen and lymph node remnants. Thus, Th17 cells are uniquely endowed to induce tissue inflammation, characterized by ectopic lymphoid follicles within the target organ.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Th17 Cells/immunology , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Th17 Cells/metabolism
19.
J Immunol ; 187(12): 6176-9, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22084440

ABSTRACT

The neutralization of α4 integrin is currently used as treatment in several autoimmune diseases and is thought to prevent the entry of most immune cells in target tissues. In this study, we showed that selective deletion of α4 integrin in T cells did not prevent but delayed the development of experimental autoimmune encephalomyelitis. Whereas both Th1 and Th17 cells infiltrate the CNS of wild-type mice, T cells present in the CNS of mice lacking α4 integrin were mainly enriched in Th17 cells, suggesting that this T cell subset uses other integrins to access the CNS. In contrast, α4 integrin expression is important for Th1 cells to enter the CNS and for the stability of their Th1-associated genetic program. Therefore, our data suggest that anti-α4 integrin Ab treatment may be more efficient in the treatment of Th1- rather than Th17-mediated disease.


Subject(s)
Cell Migration Inhibition/immunology , Gene Deletion , Gene Expression Regulation/immunology , Integrin alpha4/genetics , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Migration Inhibition/genetics , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Integrin alpha4/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/metabolism , Th17 Cells/pathology
20.
Proc Natl Acad Sci U S A ; 107(32): 14292-7, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20660725

ABSTRACT

T helper type 17 (TH17) cells are highly proinflammatory effector T cells that are characterized by the production of high amounts of IL-17A, IL-17F, IL-21, and IL-22. Furthermore, TH17 cells have been associated with a number of autoimmune diseases. However, it is not clear whether TH17 cells can also serve as effective helper cells. Here we show that TH17 cells can function as B-cell helpers in that they not only induce a strong proliferative response of B cells in vitro but also trigger antibody production with class switch recombination in vivo. Transfer of TH17 cells into WT or T-cell receptor alpha-deficient mice, which lack endogenous T cells, induces a pronounced antibody response with preferential isotype class switching to IgG1, IgG2a, IgG2b, and IgG3, as well as the formation of germinal centers. Conversely, blockade of IL-17 signaling results in a significant reduction in both number and size of germinal centers. Whereas IL-21 is known to help B cells, IL-17 on its own drives B cells to undergo preferential isotype class switching to IgG2a and IgG3 subtypes. These observations provide insights into the unappreciated role of TH17 cells and their signature cytokines in mediating B-cell differentiation and class switch recombination.


Subject(s)
B-Lymphocytes/immunology , Interleukin-17/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation , Cell Differentiation , Cell Proliferation , Immunoglobulin Class Switching , Immunoglobulin Isotypes/analysis , Inflammation/immunology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...