Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
J Autoimmun ; 128: 102808, 2022 04.
Article En | MEDLINE | ID: mdl-35276587

Medullary thymic epithelial cells (mTECs) induce T cell tolerance in the thymus through the elimination of self-reactive thymocytes. Commensal bacteria are also critical for shaping T cell responses in the gut and distal organs. We previously showed that mice depleted of mTECs (Traf6ΔTEC) generated autoreactive T cells and developed autoimmune hepatitis (AIH). In this report, we found that Toll-like receptor (TLR)-mediated microbial sensing on liver hematopoietic cells and the gut microbiota contributed to AIH development in Traf6ΔTEC mice. While adoptive transfer of thymic Traf6ΔTEC T cells in immune-deficient mice was sufficient for AIH development, colonization of germ-free mice with Traf6ΔTEC microbiota failed to induce AIH, suggesting that the gut microbiota contributes to but is not sufficient for AIH development. Microbiota-mediated exacerbation of AIH associated with increased numbers of hepatic Foxp3+ T cells and their increase was proportional to the degree of inflammation. The contribution of the gut microbiota to AIH development associated with an altered microbial signature whose composition was influenced by the qualitative nature of the thymic T cell compartment. These results suggest that aberrant selection of T cells in the thymus can induce changes in the gut microbiota that lead to exacerbation of organ-specific autoimmunity and AIH. Our results add to our understanding of the mechanisms of AIH development and create a platform towards developing novel therapeutic approaches for treating this disease.


Gastrointestinal Microbiome , Hepatitis, Autoimmune , Animals , Central Tolerance , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , Thymus Gland
2.
Cell Host Microbe ; 29(8): 1305-1315.e6, 2021 Aug 11.
Article En | MEDLINE | ID: mdl-34320399

Anelloviruses are a ubiquitous component of healthy human viromes and remain highly prevalent after being acquired early in life. The full extent of "anellome" diversity and its evolutionary dynamics remain unexplored. We employed in-depth sequencing of blood-transfusion donor(s)-recipient pairs coupled with public genomic resources for a large-scale assembly of anellovirus genomes and used the data to characterize global and personal anellovirus diversity through time. The breadth of the anellome is much greater than previously appreciated, and individuals harbor unique anellomes and transmit lineages that can persist for several months within a diverse milieu of endemic host lineages. Anellovirus sequence diversity is shaped by extensive recombination at all levels of divergence, hindering traditional phylogenetic analyses. Our findings illuminate the transmission dynamics and vast diversity of anelloviruses and set the foundation for future studies to characterize their biology.


Anelloviridae/classification , Anelloviridae/genetics , DNA Virus Infections/virology , Phylogeny , Virome , Blood Transfusion , Coinfection , Genome, Viral , Genomics , Humans
3.
J Autoimmun ; 75: 141-149, 2016 Dec.
Article En | MEDLINE | ID: mdl-27543048

In the thymus, antigen presenting cells (APCs) namely, medullary thymic epithelial cells (mTECs) and thymic dendritic cells (tDCs) regulate T cell tolerance through elimination of autoreactive T cells and production of thymic T regulatory (tTreg) cells. How the different APCs in the thymus share the burden of tolerazing the emerging T cell repertoire remains unclear. For example, while mutations that inhibit mTEC development or function associate with peripheral autoimmunity, the role of tDCs in organ-specific autoimmunity and tTreg cell production remains controversial. In this report we used mice depleted of mTECs and/or CD8α+ DCs, to examine the contributions of these cell populations in thymic tolerance. We found that while mice depleted of CD8α+ DCs or mTECs were normal or developed liver inflammation respectively, combined depletion of mTECs and CD8α+ DCs resulted in overt peripheral autoimmunity. The autoimmune manifestations in mice depleted of both mTECs and CD8α+ cDCs associated with increased percentages of CD4+ and CD8+ T cells in the thymus. In contrast, while mTEC depletion resulted in reduced percentages of tTreg cells, no additional effect was observed when CD8α+ DCs were also depleted. These results reveal that: 1) mTECs and CD8α+ DCs cooperatively safeguard against peripheral autoimmunity through thymic T cell deletion; 2) CD8α+ DCs are dispensable for tTreg cell production, whereas mTECs play a non-redundant role in this process; 3) mTECs and CD8α+ DCs make unique contributions to tolerance induction that cannot be compensated for by other thymic APCs such as migratory SIRPα+ or plasmacytoid DCs.


CD8 Antigens/immunology , Central Tolerance/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Autoimmunity/immunology , CD8 Antigens/metabolism , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Flow Cytometry , Immune Tolerance/immunology , Lymphocyte Depletion , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
4.
Arterioscler Thromb Vasc Biol ; 36(9): 1791-801, 2016 09.
Article En | MEDLINE | ID: mdl-27417580

OBJECTIVE: The chronic inflammation associated with atherosclerosis is caused by lipid deposition followed by leukocyte recruitment to the arterial wall. We previously showed that the hematopoietic cell-specific adaptor protein Cas- and Hef1-associated signal transducer hematopoietic isoform (Chat-H)/SHEP1 regulated lymphocyte adhesion and migration. In this study, we analyzed the role of Chat-H in atherosclerosis development. APPROACH AND RESULTS: Using Chat-H-deficient bone marrow transplantation in low-density lipoprotein receptor-deficient mice, we found that Chat-H regulated atherosclerotic plaque formation. Chat-H deficiency in hematopoietic cells associated with lower plaque complexity and fewer leukocytes in the lesions, whereas myeloid-specific deletion of Chat-H was sufficient for conferring atheroprotection. Chat-H deficiency resulted in reduced recruitment of classical Ly6c(high) and nonclassical Ly6c(low) monocytes to the plaques, which was accompanied by increased numbers of both monocyte subsets in the blood. This associated with defective adhesion of Chat-H-deficient Ly6c(high) and Ly6c(low) monocytes to vascular cell adhesion molecule-1 in vitro and impaired infiltration of fluorescent bead-loaded monocytes to atherosclerotic plaques. In contrast, Chat-H was dispensable for CX3CL1 and CCR1/CCR5-dependent migration of monocytes. CONCLUSIONS: Our findings highlight Chat-H as a key protein that regulates atherosclerosis development by controlling monocyte adhesion and recruitment to the plaques and identify a novel target that may be exploited for treating atherosclerosis.


Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Cell Adhesion , Chemotaxis, Leukocyte , Monocytes/metabolism , Plaque, Atherosclerotic , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, Ly/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Bone Marrow Transplantation , Cells, Cultured , Disease Models, Animal , Genotype , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Neutrophils/metabolism , Neutrophils/pathology , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Vascular Cell Adhesion Molecule-1/metabolism
5.
J Clin Invest ; 126(4): 1581-91, 2016 Apr 01.
Article En | MEDLINE | ID: mdl-26999610

Lymphatic collecting vessels direct lymph into and from lymph nodes (LNs) and can become hyperpermeable as the result of a previous infection. Enhanced permeability has been implicated in compromised immunity due to reduced flow of lymph and immune cells to LNs, which are the primary site of antigen presentation to T cells. Presently, very little is known about the molecular signals that affect lymphatic collecting vessel permeability. Here, we have shown that lymphatic collecting vessel permeability is controlled by CCR7 and that the chronic hyperpermeability of collecting vessels observed in Ccr7-/- mice is followed by vessel fibrosis. Reexpression of CCR7 in DCs, however, was sufficient to reverse the development of such fibrosis. IFN regulatory factor 4-positive (IRF4+) DCs constitutively interacted with collecting lymphatics, and selective ablation of this DC subset in Cd11c-Cre Irf4fl/fl mice also rendered lymphatic collecting vessels hyperpermeable and fibrotic. Together, our data reveal that CCR7 plays multifaceted roles in regulating collecting vessel permeability and fibrosis, with one of the key players being IRF4-dependent DCs.


Capillary Permeability/immunology , Dendritic Cells/immunology , Interferon Regulatory Factors/immunology , Lymphatic Vessels/immunology , Receptors, CCR7/immunology , Animals , Capillary Permeability/genetics , Dendritic Cells/pathology , Fibrosis/genetics , Fibrosis/immunology , Fibrosis/pathology , Interferon Regulatory Factors/genetics , Lymphatic Vessels/pathology , Mice , Mice, Knockout , Receptors, CCR7/genetics
6.
J Exp Med ; 213(1): 53-73, 2016 Jan 11.
Article En | MEDLINE | ID: mdl-26712806

Protective immunoglobulin A (IgA) responses to oral antigens are usually orchestrated by gut dendritic cells (DCs). Here, we show that lung CD103(+) and CD24(+)CD11b(+) DCs induced IgA class-switch recombination (CSR) by activating B cells through T cell-dependent or -independent pathways. Compared with lung DCs (LDC), lung CD64(+) macrophages had decreased expression of B cell activation genes and induced significantly less IgA production. Microbial stimuli, acting through Toll-like receptors, induced transforming growth factor-ß (TGF-ß) production by LDCs and exerted a profound influence on LDC-mediated IgA CSR. After intranasal immunization with inactive cholera toxin (CT), LDCs stimulated retinoic acid-dependent up-regulation of α4ß7 and CCR9 gut-homing receptors on local IgA-expressing B cells. Migration of these B cells to the gut resulted in IgA-mediated protection against an oral challenge with active CT. However, in germ-free mice, the levels of LDC-induced, CT-specific IgA in the gut are significantly reduced. Herein, we demonstrate an unexpected role of the microbiota in modulating the protective efficacy of intranasal vaccination through their effect on the IgA class-switching function of LDCs.


Dendritic Cells/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Immunoglobulin Class Switching/genetics , Lung/immunology , Microbiota , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antigens, CD/metabolism , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD24 Antigen/metabolism , Cell Movement/genetics , Dendritic Cells/metabolism , Gastrointestinal Tract/metabolism , Gene Expression , Immunoglobulin Class Switching/drug effects , Integrin alpha Chains/metabolism , Integrins/genetics , Integrins/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Receptors, CCR/genetics , Receptors, CCR/metabolism , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
7.
Int J Mol Sci ; 16(1): 1980-2000, 2015 Jan 16.
Article En | MEDLINE | ID: mdl-25603179

Autoimmune hepatitis (AIH) is an immune-mediated disorder that affects the liver parenchyma. Diagnosis usually occurs at the later stages of the disease, complicating efforts towards understanding the causes of disease development. While animal models are useful for studying the etiology of autoimmune disorders, most of the existing animal models of AIH do not recapitulate the chronic course of the human condition. In addition, approaches to mimic AIH-associated liver inflammation have instead led to liver tolerance, consistent with the high tolerogenic capacity of the liver. Recently, we described a new mouse model that exhibited spontaneous and chronic liver inflammation that recapitulated the known histopathological and immunological parameters of AIH. The approach involved liver-extrinsic genetic engineering that interfered with the induction of T-cell tolerance in the thymus, the very process thought to inhibit AIH induction by liver-specific expression of exogenous antigens. The mutation led to depletion of specialized thymic epithelial cells that present self-antigens and eliminate autoreactive T-cells before they exit the thymus. Based on our findings, which are summarized below, we believe that this mouse model represents a relevant experimental tool towards elucidating the cellular and molecular aspects of AIH development and developing novel therapeutic strategies for treating this disease.


Central Tolerance/immunology , Disease Models, Animal , Epithelial Cells/pathology , Hepatitis, Autoimmune/immunology , Thymus Gland/pathology , Animals , Mice , T-Lymphocytes, Regulatory/immunology
8.
J Clin Invest ; 123(8): 3510-24, 2013 Aug.
Article En | MEDLINE | ID: mdl-23867620

TRAF6, an E3 ubiquitin protein ligase, plays a critical role in T cell tolerance by regulating medullary thymic epithelial cell (mTEC) development. mTECs regulate T cell tolerance by ectopically expressing self-antigens and eliminating autoreactive T cells in the thymus. Here we show that mice with mTEC depletion due to conditional deletion of Traf6 expression in murine thymic epithelial cells (Traf6ΔTEC mice) showed a surprisingly narrow spectrum of autoimmunity affecting the liver. The liver inflammation in Traf6ΔTEC mice exhibited all the histological and immunological characteristics of human autoimmune hepatitis (AIH). The role of T cells in AIH establishment was supported by intrahepatic T cell population changes and AIH development after transfer of liver T cells into immunodeficient mice. Despite a 50% reduction in natural Treg thymic output, peripheral tolerance in Traf6ΔTEC mice was normal, whereas compensatory T regulatory mechanisms were evident in the liver of these animals. These data indicate that mTECs exert a cell-autonomous role in central T cell tolerance and organ-specific autoimmunity, but play a redundant role in peripheral tolerance. These findings also demonstrate that Traf6ΔTEC mice are a relevant model with which to study the pathophysiology of AIH, as well as autoantigen-specific T cell responses and regulatory mechanisms underlying this disease.


Epithelial Cells/pathology , Hepatitis, Autoimmune/pathology , Thymus Gland/pathology , Adoptive Transfer , Animals , Antibodies, Antinuclear/metabolism , Disease Models, Animal , Epithelial Cells/immunology , Hepatitis, Autoimmune/immunology , Humans , Immune Tolerance , Liver/immunology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells/pathology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , T-Lymphocytes, Regulatory/immunology , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/genetics , Thymus Gland/immunology
9.
Immunol Invest ; 36(5-6): 581-605, 2007.
Article En | MEDLINE | ID: mdl-18161520

Flow cytometric analyses of immune cell proliferation, differentiation, and function are limited by the number of different fluorochromes that can be resolved simultaneously. Additional colors to expand functional analytic capability will facilitate higher dimensional analyses of heterogeneous cell populations by basic and clinical scientists. Our aim in these studies was to evaluate CellVue Claret, a fluorescent, far-red emitting, membrane intercalating dye (excitation maximum: 655 nm, emission maximum 677 nm), as an alternative and/or complementary probe to PKH26 and CFSE(1) for polychromatic studies of immune cell proliferation and function. Using a BD FACSCalibur and human peripheral blood mononuclear cells (PBMCs) from 8 different donors (2 donors studied twice), we compared CellVue Claret with the two most commonly used visible-emitting proliferation dyes, PKH26 and CFSE, in terms of: (1) compatibility with 7-Amino-actinomycin D (7-AAD) as a viability marker; (2) effect of dye labeling on lymphocyte viability; and (3) the proliferative response of CD3+ T lymphocytes from 0-96 hours as assessed by dilution of each of the 3 cell tracking dyes in cultures stimulated with anti-CD3 plus IL-2. Post-labeling recoveries and viabilities were similar for all 3 dyes, with modestly higher initial staining intensities and coefficients of variation for CellVue Claret than for CFSE or PKH26. Lymphocyte viabilities in stimulated or unstimulated cultures were also unaffected by choice of dye. Proliferative responses of viable CD3+ lymphocytes were comparable for all three dyes, whether results were reported as Proliferative Fraction (percent of cells that had divided one or more times) or as Precursor Frequency (percent of parent population that had gone on to proliferate in response to anti-CD3 plus IL-2). In summary, T cell proliferation analysis using CellVue Claret gives results equivalent to those obtained with PKH26 or CFSE, expanding the choice of proliferation dyes suitable for use in high dimensional polychromatic studies on flow cytometers with far red (633 nm-658 nm) excitation capabilities.


Cell Proliferation , Fluorescent Dyes , Leukocytes, Mononuclear/cytology , T-Lymphocytes/cytology , Cells, Cultured , Evaluation Studies as Topic , Humans , Rhodamines , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
...