Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cell ; 184(2): 422-440.e17, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33450207

ABSTRACT

Itch is an evolutionarily conserved sensation that facilitates expulsion of pathogens and noxious stimuli from the skin. However, in organ failure, cancer, and chronic inflammatory disorders such as atopic dermatitis (AD), itch becomes chronic, intractable, and debilitating. In addition to chronic itch, patients often experience intense acute itch exacerbations. Recent discoveries have unearthed the neuroimmune circuitry of itch, leading to the development of anti-itch treatments. However, mechanisms underlying acute itch exacerbations remain overlooked. Herein, we identify that a large proportion of patients with AD harbor allergen-specific immunoglobulin E (IgE) and exhibit a propensity for acute itch flares. In mice, while allergen-provoked acute itch is mediated by the mast cell-histamine axis in steady state, AD-associated inflammation renders this pathway dispensable. Instead, a previously unrecognized basophil-leukotriene (LT) axis emerges as critical for acute itch flares. By probing fundamental itch mechanisms, our study highlights a basophil-neuronal circuit that may underlie a variety of neuroimmune processes.


Subject(s)
Basophils/pathology , Neurons/pathology , Pruritus/pathology , Acute Disease , Allergens/immunology , Animals , Chronic Disease , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Disease Models, Animal , Histamine/metabolism , Humans , Immunoglobulin E/immunology , Inflammation/pathology , Leukotrienes/metabolism , Mast Cells/immunology , Mice, Inbred C57BL , Phenotype , Pruritus/immunology , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
2.
Cell ; 171(1): 217-228.e13, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28890086

ABSTRACT

Mammals have evolved neurophysiologic reflexes, such as coughing and scratching, to expel invading pathogens and noxious environmental stimuli. It is well established that these responses are also associated with chronic inflammatory diseases, including asthma and atopic dermatitis. However, the mechanisms by which inflammatory pathways promote sensations such as itch remain poorly understood. Here, we show that type 2 cytokines directly activate sensory neurons in both mice and humans. Further, we demonstrate that chronic itch is dependent on neuronal IL-4Rα and JAK1 signaling. We also observe that patients with recalcitrant chronic itch that failed other immunosuppressive therapies markedly improve when treated with JAK inhibitors. Thus, signaling mechanisms previously ascribed to the immune system may represent novel therapeutic targets within the nervous system. Collectively, this study reveals an evolutionarily conserved paradigm in which the sensory nervous system employs classical immune signaling pathways to influence mammalian behavior.


Subject(s)
Pruritus/immunology , Sensory Receptor Cells/immunology , Sensory Receptor Cells/metabolism , Signal Transduction , Skin Diseases/immunology , Animals , Ganglia, Spinal , Humans , Interleukin-13/immunology , Interleukin-4/immunology , Janus Kinase 1/metabolism , Mice , Mice, Inbred C57BL , Pruritus/metabolism , Skin Diseases/pathology
3.
Immunity ; 54(12): 2795-2811.e9, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34788601

ABSTRACT

Lymphangitis and the formation of tertiary lymphoid organs (TLOs) in the mesentery are features of Crohn's disease. Here, we examined the genesis of these TLOs and their impact on disease progression. Whole-mount and intravital imaging of the ileum and ileum-draining collecting lymphatic vessels (CLVs) draining to mesenteric lymph nodes from TNFΔARE mice, a model of ileitis, revealed TLO formation at valves of CLVs. TLOs obstructed cellular and molecular outflow from the gut and were sites of lymph leakage and backflow. Tumor necrosis factor (TNF) neutralization begun at early stages of TLO formation restored lymph transport. However, robustly developed, chronic TLOs resisted regression and restoration of flow after TNF neutralization. TNF stimulation of cultured lymphatic endothelial cells reprogrammed responses to oscillatory shear stress, preventing the induction of valve-associated genes. Disrupted transport of immune cells, driven by loss of valve integrity and TLO formation, may contribute to the pathology of Crohn's disease.


Subject(s)
Crohn Disease/immunology , Endothelial Cells/immunology , Ileum/immunology , Lymph/metabolism , Lymphatic Vessels/immunology , Mesentery/immunology , Tertiary Lymphoid Structures/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Humans , Ileitis , Lymphangitis , Mice , Mice, Knockout , Stress, Mechanical
4.
Immunity ; 54(11): 2547-2564.e7, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34715017

ABSTRACT

Cryptosporidium can cause severe diarrhea and morbidity, but many infections are asymptomatic. Here, we studied the immune response to a commensal strain of Cryptosporidium tyzzeri (Ct-STL) serendipitously discovered when conventional type 1 dendritic cell (cDC1)-deficient mice developed cryptosporidiosis. Ct-STL was vertically transmitted without negative health effects in wild-type mice. Yet, Ct-STL provoked profound changes in the intestinal immune system, including induction of an IFN-γ-producing Th1 response. TCR sequencing coupled with in vitro and in vivo analysis of common Th1 TCRs revealed that Ct-STL elicited a dominant antigen-specific Th1 response. In contrast, deficiency in cDC1s skewed the Ct-STL CD4 T cell response toward Th17 and regulatory T cells. Although Ct-STL predominantly colonized the small intestine, colon Th1 responses were enhanced and associated with protection against Citrobacter rodentium infection and exacerbation of dextran sodium sulfate and anti-IL10R-triggered colitis. Thus, Ct-STL represents a commensal pathobiont that elicits Th1-mediated intestinal homeostasis that may reflect asymptomatic human Cryptosporidium infection.


Subject(s)
Cryptosporidiosis/immunology , Cryptosporidiosis/parasitology , Cryptosporidium/immunology , Dendritic Cells/immunology , Host-Parasite Interactions/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Th1 Cells/immunology , Animals , Dendritic Cells/metabolism , Disease Models, Animal , Homeostasis , Intestinal Mucosa/metabolism , Mice , Microbiota , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/metabolism
5.
Immunity ; 48(5): 923-936.e4, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29752065

ABSTRACT

The development of T cell tolerance in the thymus requires the presentation of host proteins by multiple antigen-presenting cell (APC) types. However, the importance of transferring host antigens from transcription factor AIRE-dependent medullary thymic epithelial cells (mTECs) to bone marrow (BM) APCs is unknown. We report that antigen was primarily transferred from mTECs to CD8α+ dendritic cells (DCs) and showed that CD36, a scavenger receptor selectively expressed on CD8α+ DCs, mediated the transfer of cell-surface, but not cytoplasmic, antigens. The absence of CD8α+ DCs or CD36 altered thymic T cell selection, as evidenced by TCR repertoire analysis and the loss of allo-tolerance in murine allogeneic BM transplantation (allo-BMT) studies. Decreases in these DCs and CD36 expression in peripheral blood of human allo-BMT patients correlated with graft-versus-host disease. Our findings suggest that CD36 facilitates transfer of mTEC-derived cell-surface antigen on CD8α+ DCs to promote tolerance to host antigens during homeostasis and allo-BMT.


Subject(s)
Antigens, Surface/immunology , CD36 Antigens/immunology , Immune Tolerance/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Antigens, Surface/metabolism , Bone Marrow Transplantation , CD36 Antigens/genetics , CD36 Antigens/metabolism , CD8 Antigens/immunology , CD8 Antigens/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/metabolism , Transplantation, Homologous
6.
Immunity ; 41(3): 414-426, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25220213

ABSTRACT

The contribution of thymic antigen-presenting-cell (APC) subsets in selecting a self-tolerant T cell population remains unclear. We show that bone marrow (BM) APCs and medullary thymic epithelial cells (mTECs) played nonoverlapping roles in shaping the T cell receptor (TCR) repertoire by deletion and regulatory T (Treg) cell selection of distinct TCRs. Aire, which induces tissue-specific antigen expression in mTECs, affected the TCR repertoire in a manner distinct from mTEC presentation. Approximately half of Aire-dependent deletion or Treg cell selection utilized a pathway dependent on antigen presentation by BM APCs. Batf3-dependent CD8α⁺ dendritic cells (DCs) were the crucial BM APCs for Treg cell selection via this pathway, showing enhanced ability to present antigens from stromal cells. These results demonstrate the division of function between thymic APCs in shaping the self-tolerant TCR repertoire and reveal an unappreciated cooperation between mTECs and CD8α⁺ DCs for presentation of Aire-induced self-antigens to developing thymocytes.


Subject(s)
Bone Marrow Cells/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Self Tolerance/immunology , Thymus Gland/immunology , Transcription Factors/genetics , Animals , Antigen Presentation/immunology , Autoantigens/immunology , Basic-Leucine Zipper Transcription Factors/genetics , CD8 Antigens/biosynthesis , Cell Differentiation/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Transcription Factors/biosynthesis , AIRE Protein
7.
J Immunol ; 207(2): 398-407, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34193598

ABSTRACT

Alterations in gut microbiota in early life have been associated with the development of asthma; however, the role of gut bacteria or the IgA response to gut bacteria in school-aged children with asthma is unclear. To address this question, we profiled the microbial populations in fecal and nasal swab samples by 16S rRNA sequencing from 40 asthma and 40 control children aged 9-17 y from Peru. Clinical history and laboratory evaluation of asthma and allergy were obtained. Fecal samples were analyzed by flow cytometry and sorted into IgA+ and IgA- subsets for 16S rRNA sequencing. We found that the fecal or nasal microbial 16S rRNA diversity and frequency of IgA+ fecal bacteria did not differ between children with or without asthma. However, the α diversity of fecal IgA+ bacteria was decreased in asthma compared with control. Machine learning analysis of fecal bacterial IgA-enrichment data revealed loss of IgA binding to the Blautia, Ruminococcus, and Lachnospiraceae taxa in children with asthma compared with controls. In addition, this loss of IgA binding was associated with worse asthma control (Asthma Control Test) and increased odds of severe as opposed to mild to moderate asthma. Thus, despite little to no change in the microbiota, children with asthma exhibit an altered host IgA response to gut bacteria compared with control participants. Notably, the signature of altered IgA responses is loss of IgA binding, in particular to members of Clostridia spp., which is associated with greater severity of asthma.


Subject(s)
Asthma/immunology , Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Adolescent , Bacteria/genetics , Bacteria/immunology , Case-Control Studies , Child , Feces/microbiology , Female , Humans , Hypersensitivity/immunology , Male , Microbiota/genetics , Microbiota/immunology , Peru , RNA, Ribosomal, 16S/genetics , Young Adult
9.
Nat Immunol ; 10(6): 610-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19430476

ABSTRACT

Because the deletion of self-reactive T cells is incomplete, thymic development of natural Foxp3+CD4+ regulatory T cells (Treg cells) is required for preventing autoimmunity. However, the function of T cell antigen receptor (TCR) specificity in thymic Treg cell development remains controversial. To address this issue, we generated a transgenic line expressing a naturally occurring Treg cell-derived TCR. Unexpectedly, we found that efficient thymic Treg cell development occurred only when the antigen-specific Treg cell precursors were present at low clonal frequency (o1%) in a normal thymus. Using retroviral vectors and bone marrow chimeras, we observed similar activity with two other Treg cell-derived TCRs. Our data demonstrate that thymic Treg cell development is a 'TCR-instructive' process involving a niche that can be saturable at much lower clonal frequencies than is the niche for positive selection.


Subject(s)
Genes, T-Cell Receptor beta , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , Bone Marrow Cells/immunology , Cell Differentiation , Chimera/immunology , Forkhead Transcription Factors/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics
10.
Immunity ; 37(3): 475-86, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22921379

ABSTRACT

The degree of T cell self-reactivity considered dangerous by the immune system, thereby requiring thymic selection processes to prevent autoimmunity, is unknown. Here, we analyzed a panel of T cell receptors (TCRs) with a broad range of reactivity to ovalbumin (OVA(323-339)) in the rat insulin promoter (RIP)-mOVA self-antigen model for their ability to trigger thymic self-tolerance mechanisms. Thymic regulatory T (Treg) cell generation in vivo was directly correlated with in vitro TCR reactivity to OVA-peptide in a broad ~1,000-fold range. Interestingly, higher TCR affinity was associated with a larger Treg cell developmental "niche" size, even though the amount of antigen should remain constant. The TCR-reactivity threshold to elicit thymic negative selection and peripheral T cell responses was ~100-fold higher than that of Treg cell differentiation. Thus, these data suggest that the broad range of self-reactivity that elicits thymic Treg cell generation is tuned to secure peripheral tolerance to self.


Subject(s)
Autoantigens/immunology , Receptors, Antigen, T-Cell/immunology , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Cell Differentiation/immunology , Female , Flow Cytometry , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Ovalbumin/chemistry , Ovalbumin/immunology , Peptides/immunology , Peripheral Tolerance/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism
11.
Immunol Rev ; 271(1): 141-55, 2016 May.
Article in English | MEDLINE | ID: mdl-27088912

ABSTRACT

The development of T-cell self-tolerance in the thymus is important for establishing immune homeostasis and preventing autoimmunity. Here, we review the components of T-cell tolerance, which includes T-cell receptor (TCR) self-reactivity, costimulation, cytokines, and antigen presentation by a variety of antigen-presenting cells (APCs) subsets. We discuss the current evidence on the process of regulatory T (Treg) cell and negative selection and the importance of TCR signaling. We then examine recent evidence showing unique roles for bone marrow (BM)-derived APCs and medullary thymic epithelial cells (mTECs) on the conventional and Treg TCR repertoire, as well as emerging data on the role of B cells in tolerance. Finally, we review the accumulating data that suggest that cooperative antigen presentation is a prominent component of T -ell tolerance. With the development of tools to interrogate the function of individual APC subsets in the medulla, we have gained greater understanding of the complex cellular and molecular events that determine T-cell tolerance.


Subject(s)
Bone Marrow Cells/physiology , Epithelial Cells/physiology , Self Tolerance , T-Lymphocytes/physiology , Thymus Gland/immunology , Antigen Presentation , Autoantigens/immunology , Autoimmunity , B-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
12.
Immunity ; 31(6): 852-3, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-20064443

ABSTRACT

In this issue of Immunity, reports by Long et al. (2009) and Ruan et al. (2009) suggest that the transcription factor NF-kappaB-c-Rel is an important molecular mechanism by which T cell receptor-specificity for self-antigens instructs the selection of Foxp3(+) regulatory T cells.


Subject(s)
Forkhead Transcription Factors/immunology , NF-kappa B/immunology , Proto-Oncogene Proteins c-rel/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Mice , NF-kappa B/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism
13.
J Immunol ; 197(9): 3512-3519, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27671109

ABSTRACT

Foxp3+retinoic acid-related orphan receptor (ROR)γt+ T cells have recently been characterized as an immunoregulatory population highly enriched in the colon lamina propria. However, their developmental origin and relationship to RORγt- regulatory T and Th17 cells remain unclear. In this study, we use a fixed TCRß system to show that the TCR repertoire of the Foxp3+RORγt+ population is mostly distinct compared with other colonic T cell subsets. However, of these TCRs, a fraction is also found in the Th17 subset, suggesting that TCR repertoire overlap may contribute to the reported ability of Foxp3+RORγt+ cells to regulate Th17 immunity. Naive transgenic T cells expressing a Foxp3+RORγt+-restricted TCR first acquire a Foxp3+RORγt- phenotype before coexpressing RORγt, suggesting that Foxp3+RORγt+ cell development can occur via an RORγt- regulatory T cell intermediate.


Subject(s)
Intestinal Mucosa/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Precursor Cells, T-Lymphoid/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cell Differentiation , Cells, Cultured , Forkhead Transcription Factors/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/genetics
14.
Immunol Rev ; 259(1): 60-74, 2014 May.
Article in English | MEDLINE | ID: mdl-24712459

ABSTRACT

Although intestinal bacteria live deep within the body, they are topographically on the exterior surface and thus outside the host. According to the classic notion that the immune system targets non-self rather than self, these intestinal bacteria should be considered foreign and therefore attacked and eliminated. While this appears to be true for some commensal bacterial species, recent data suggest that the immune system actively becomes tolerant to many bacterial organisms. The induction or activation of regulatory T (Treg) cells that inhibit, rather than promote, inflammatory responses to commensal bacteria appears to be a central component of mucosal tolerance. Loss of this mechanism can lead to inappropriate immune reactivity toward commensal organisms, perhaps contributing to mucosal inflammation characteristic of disorders such as inflammatory bowel disease.


Subject(s)
Clonal Selection, Antigen-Mediated/immunology , Homeostasis/immunology , Intestines/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bacteria/immunology , Cell Differentiation/immunology , Cellular Microenvironment/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Host-Pathogen Interactions/immunology , Humans , Immune System/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestines/microbiology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/microbiology , Transcription Factors/metabolism
15.
Nature ; 478(7368): 250-4, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21937990

ABSTRACT

The instruction of the immune system to be tolerant of self, thereby preventing autoimmunity, is facilitated by the education of T cells in a specialized organ, the thymus, in which self-reactive cells are either eliminated or differentiated into tolerogenic Foxp3(+) regulatory T (T(reg)) cells. However, it is unknown whether T cells are also educated to be tolerant of foreign antigens, such as those from commensal bacteria, to prevent immunopathology such as inflammatory bowel disease. Here we show that encounter with commensal microbiota results in the peripheral generation of T(reg) cells rather than pathogenic effectors. We observed that colonic T(reg) cells used T-cell antigen receptors (TCRs) different from those used by T(reg) cells in other locations, implying an important role for local antigens in shaping the colonic T(reg)-cell population. Many of the local antigens seemed to be derived from commensal bacteria, on the basis of the in vitro reactivity of common colon T(reg) TCRs. These TCRs did not facilitate thymic T(reg)-cell development, implying that many colonic T(reg) cells arise instead by means of antigen-driven peripheral T(reg)-cell development. Further analysis of two of these TCRs by the creation of retroviral bone marrow chimaeras and a TCR transgenic line revealed that microbiota indigenous to our mouse colony was required for the generation of colonic T(reg) cells from otherwise naive T cells. If T cells expressing these TCRs fail to undergo T(reg)-cell development and instead become effector cells, they have the potential to induce colitis, as evidenced by adoptive transfer studies. These results suggest that the efficient peripheral generation of antigen-specific populations of T(reg) cells in response to an individual's microbiota provides important post-thymic education of the immune system to foreign antigens, thereby providing tolerance to commensal microbiota.


Subject(s)
Colon/immunology , Colon/microbiology , Immune System/immunology , Metagenome/immunology , Animals , Colitis/immunology , Colitis/prevention & control , Colon/cytology , Immune System/cytology , Immune Tolerance/immunology , Immunity, Mucosal/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymus Gland/immunology
16.
J Immunol ; 193(4): 1778-86, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015825

ABSTRACT

Thymic selection is designed to ensure TCR reactivity to foreign Ags presented by self-MHC while minimizing reactivity to self-Ags. We hypothesized that the repertoire of T cells with unwanted specificities such as alloreactivity or autoreactivity are a consequence of simultaneous rearrangement of both TCRα loci. We hypothesized that this process helps maximize production of thymocytes capable of successfully completing thymic selection, but results in secondary TCRs that escape stringent selection. In T cells expressing two TCRs, one TCR can mediate positive selection and mask secondary TCR from negative selection. Examination of mice heterozygous for TRAC (TCRα(+/-)), capable of only one functional TCRα rearrangement, demonstrated a defect in generating mature T cells attributable to decreased positive selection. Elimination of secondary TCRs did not broadly alter the peripheral T cell compartment, though deep sequencing of TCRα repertoires of dual TCR T cells and TCRα(+/-) T cells demonstrated unique TCRs in the presence of secondary rearrangements. The functional impact of secondary TCRs on the naive peripheral repertoire was evidenced by reduced frequencies of T cells responding to autoantigen and alloantigen peptide-MHC tetramers in TCRα(+/-) mice. T cell populations with secondary TCRs had significantly increased ability to respond to altered peptide ligands related to their allogeneic ligand as compared with TCRα(+/-) cells, suggesting increased breadth in peptide recognition may be a mechanism for their reactivity. Our results imply that the role of secondary TCRs in forming the T cell repertoire is perhaps more significant than what has been assumed.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology , Thymocytes/immunology , Amino Acid Sequence , Animals , Autoantigens/immunology , CHO Cells , Cell Differentiation/immunology , Cells, Cultured , Cricetulus , Genetic Variation , Graft vs Host Disease/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocyte Subsets/immunology
17.
J Immunol ; 191(6): 3082-9, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23956428

ABSTRACT

The CD28 costimulatory receptor is a critical regulator of T cell function, making it an attractive therapeutic target for the treatment of immune-mediated diseases. CTLA4Ig, now approved for use in humans, prevents naive T cell activation by binding to B7 proteins and blocking engagement of CD28. However, CTLA4Ig suppresses inflammation even if administered when disease is established, suggesting alternative mechanisms. We identified a novel, CD28-independent mechanism by which CTLA4Ig inhibits activated T cells. We show that in vitro, CTLA4Ig synergizes with NO from bone marrow-derived macrophages to inhibit T cell proliferation. Depletion of regulatory T cells (Tregs) or interference with TGF-ß signaling abrogated the inhibitory effect of CTLA4Ig. Parallel in vivo experiments using an allergic airway inflammation model demonstrated that this novel mechanism required both macrophages and regulatory T cells. Furthermore, CTLA4Ig was ineffective in SMAD3-deficient mice, supporting a requirement for TGF-ß signaling. Thus, in addition to preventing naive T cells from being fully activated, CTLA4Ig can turn off already activated effector T cells by an NO/regulatory T cell/TGF-ß-dependent pathway. This mechanism is similar to cell-extrinsic effects of endogenous CTLA4 and may be particularly important in the ability of CTLA4Ig to treat chronic inflammatory disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , CTLA-4 Antigen/immunology , Immunoglobulin G/pharmacology , Lymphocyte Activation/drug effects , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Adoptive Transfer , Animals , Coculture Techniques , Disease Models, Animal , Flow Cytometry , Immunoconjugates/pharmacology , Lymphocyte Activation/immunology , Macrophages , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins/pharmacology , Respiratory Hypersensitivity/immunology , T-Lymphocytes, Regulatory/drug effects
18.
Eur J Immunol ; 43(8): 2089-100, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23640763

ABSTRACT

Multiple sclerosis (MS) is a presumed autoimmune disease directed against central nervous system (CNS) myelin, in which diet and obesity are implicated as risk factors. Immune responses can be influenced by molecules produced by fat cells, called adipokines. Adiponectin is an adipokine with anti-inflammatory effects. We tested the hypothesis that adiponectin has a protective role in the EAE model for MS, that can be induced by immunization with myelin antigens or transfer of myelin-specific T lymphocytes. Adiponectin deficient (ADPKO) mice developed worse EAE with greater CNS inflammation, demyelination, and axon injury. Lymphocytes from myelin-immunized ADPKO mice proliferated more, produced higher amounts of IFN-γ, IL-17, TNF-α, IL-6, and transferred more severe EAE than wild type (WT) lymphocytes. At EAE peak, the spleen and CNS of ADPKO had fewer regulatory T (Treg) cells than WT mice and during EAE recovery, Foxp3, IL-10 and TGF-ß expression levels in the CNS were reduced in ADPKO compared with WT mice. Treatment with globular adiponectin in vivo ameliorated EAE, and was associated with an increase in Treg cells. These data indicate that adiponectin is an important regulator of T-cell functions during EAE, suggesting a new avenue of investigation for MS treatment.


Subject(s)
Adiponectin/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Adiponectin/administration & dosage , Adiponectin/deficiency , Adiponectin/genetics , Adoptive Transfer , Animals , Autoimmunity , Cell Proliferation , Cells, Cultured , Central Nervous System/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Forkhead Transcription Factors/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-17/biosynthesis , Interleukin-6/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Myelin Sheath/immunology , Risk Factors , Spleen/immunology , Th1 Cells/immunology , Th1 Cells/transplantation , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
19.
bioRxiv ; 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38712036

ABSTRACT

Antigen specificity is the central trait distinguishing adaptive from innate immune function. Assembly of antigen-specific T cell and B cell receptors occurs through V(D)J recombination mediated by the Recombinase Activating Gene endonucleases RAG1 and RAG2 (collectively called RAG). In the absence of RAG, mature T and B cells do not develop and thus RAG is critically associated with adaptive immune function. In addition to adaptive T helper 2 (Th2) cells, group 2 innate lymphoid cells (ILC2s) contribute to type 2 immune responses by producing cytokines like Interleukin-5 (IL-5) and IL-13. Although it has been reported that RAG expression modulates the function of innate natural killer (NK) cells, whether other innate immune cells such as ILC2s are affected by RAG remains unclear. We find that in RAG-deficient mice, ILC2 populations expand and produce increased IL-5 and IL-13 at steady state and contribute to increased inflammation in atopic dermatitis (AD)-like disease. Further, we show that RAG modulates ILC2 function in a cell-intrinsic manner independent of the absence or presence of adaptive T and B lymphocytes. Lastly, employing multiomic single cell analyses of RAG1 lineage-traced cells, we identify key transcriptional and epigenomic ILC2 functional programs that are suppressed by a history of RAG expression. Collectively, our data reveal a novel role for RAG in modulating innate type 2 immunity through suppression of ILC2s.

20.
bioRxiv ; 2024 Jan 07.
Article in English | MEDLINE | ID: mdl-38260555

ABSTRACT

Humoral immune responses within the gut play diverse roles including pathogen clearance during enteric infections, maintaining tolerance, and facilitating the assemblage and stability of the gut microbiota. How these humoral immune responses are initiated and contribute to these processes are well studied. However, the signals promoting the expansion of these responses and their rapid mobilization to the gut mucosa are less well understood. Intestinal goblet cells form goblet cell-associated antigen passages (GAPs) to deliver luminal antigens to the underlying immune system and facilitate tolerance. GAPs are rapidly inhibited during enteric infection to prevent inflammatory responses to innocuous luminal antigens. Here we interrogate GAP inhibition as a key physiological response required for effective humoral immunity. Independent of infection, GAP inhibition resulted in enrichment of transcripts representing B cell recruitment, expansion, and differentiation into plasma cells in the small intestine (SI), which were confirmed by flow cytometry and ELISpot assays. Further we observed an expansion of isolated lymphoid follicles within the SI, as well as expansion of plasma cells in the bone marrow upon GAP inhibition. S1PR1-induced blockade of leukocyte trafficking during GAP inhibition resulted in a blunting of SI plasma cell expansion, suggesting that mobilization of plasma cells from the bone marrow contributes to their expansion in the gut. However, luminal IgA secretion was only observed in the presence of S. typhimurium infection, suggesting that although GAP inhibition mobilizes a mucosal humoral immune response, a second signal is required for full effector function. Overriding GAP inhibition during enteric infection abrogated the expansion of laminar propria IgA+ plasma cells. We conclude that GAP inhibition is a required physiological response for efficiently mobilizing mucosal humoral immunity in response to enteric infection.

SELECTION OF CITATIONS
SEARCH DETAIL