Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Sci Transl Med ; 15(678): eabo0205, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36630481

ABSTRACT

The common γ chain (γc; IL-2RG) is a subunit of the interleukin (IL) receptors for the γc cytokines IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The lack of appropriate neutralizing antibodies recognizing IL-2RG has made it difficult to thoroughly interrogate the role of γc cytokines in inflammatory and autoimmune disease settings. Here, we generated a γc cytokine receptor antibody, REGN7257, to determine whether γc cytokines might be targeted for T cell-mediated disease prevention and treatment. Biochemical, structural, and in vitro analysis showed that REGN7257 binds with high affinity to IL-2RG and potently blocks signaling of all γc cytokines. In nonhuman primates, REGN7257 efficiently suppressed T cells without affecting granulocytes, platelets, or red blood cells. Using REGN7257, we showed that γc cytokines drive T cell-mediated disease in mouse models of graft-versus-host disease (GVHD) and multiple sclerosis by affecting multiple aspects of the pathogenic response. We found that our xenogeneic GVHD mouse model recapitulates hallmarks of acute and chronic GVHD, with T cell expansion/infiltration into tissues and liver fibrosis, as well as hallmarks of immune aplastic anemia, with bone marrow aplasia and peripheral cytopenia. Our findings indicate that γc cytokines contribute to GVHD and aplastic anemia pathology by promoting these characteristic features. By demonstrating that broad inhibition of γc cytokine signaling with REGN7257 protects from immune-mediated disorders, our data provide evidence of γc cytokines as key drivers of pathogenic T cell responses, offering a potential strategy for the management of T cell-mediated diseases.


Subject(s)
Anemia, Aplastic , Graft vs Host Disease , Interleukin Receptor Common gamma Subunit , T-Lymphocytes , Animals , Mice , Anemia, Aplastic/metabolism , Antibodies, Monoclonal/metabolism , Cytokines/metabolism , Graft vs Host Disease/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Interleukin Receptor Common gamma Subunit/antagonists & inhibitors , Interleukin Receptor Common gamma Subunit/metabolism , Primates
2.
J Autoimmun ; 128: 102808, 2022 04.
Article in English | MEDLINE | ID: mdl-35276587

ABSTRACT

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.


Subject(s)
Gastrointestinal Microbiome , Hepatitis, Autoimmune , Animals , Central Tolerance , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , Thymus Gland
3.
Sci Immunol ; 6(66): eabj4026, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34919442

ABSTRACT

Despite the enormous promise of T cell therapies, the isolation and study of human T cell receptors (TCRs) of dedicated specificity remains a major challenge. To overcome this limitation, we generated mice with a genetically humanized system of T cell immunity. We used VelociGene technology to replace the murine TCRαß variable regions, along with regions encoding the extracellular domains of co-receptors CD4 and CD8, and major histocompatibility complex (MHC) class I and II, with corresponding human sequences. The resulting "VelociT" mice have normal myeloid and lymphoid immune cell populations, including thymic and peripheral αß T cell subsets comparable with wild-type mice. VelociT mice expressed a diverse TCR repertoire, mounted functional T cell responses to lymphocytic choriomeningitis virus infection, and could develop experimental autoimmune encephalomyelitis. Immunization of VelociT mice with human tumor-associated peptide antigens generated robust, antigen-specific responses and led to identification of a TCR against tumor antigen New York esophageal squamous cell carcinoma-1 with potent antitumor activity. These studies demonstrate that VelociT mice mount clinically relevant T cell responses to both MHC-I­ and MHC-II­restricted antigens, providing a powerful new model for analyzing T cell function in human disease. Moreover, VelociT mice are a new platform for de novo discovery of therapeutic human TCRs.


Subject(s)
Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes/immunology , Animals , Humans , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/genetics
4.
Sci Rep ; 7(1): 4111, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28646220

ABSTRACT

Angiotensin II (AngII) promotes hypertension, atherogenesis, vascular aneurysm and impairs post-ischemic cardiac remodeling through concerted roles on vascular cells, monocytes and T lymphocytes. However, the role of AngII in B lymphocyte responses is largely unexplored. Here, we show that chronic B cell depletion (Baffr deficiency) significantly reduces atherosclerosis in Apoe -/- mice infused with AngII. While adoptive transfer of B cells in Apoe -/- /Baffr -/- mice reversed atheroprotection in the absence of AngII, infusion of AngII in B cell replenished Apoe -/- /Baffr -/- mice unexpectedly prevented the progression of atherosclerosis. Atheroprotection observed in these mice was associated with a significant increase in regulatory CD1dhiCD5+ B cells, which produced high levels of interleukin (IL)-10 (B10 cells). Replenishment of Apoe -/- /Baffr -/- mice with Il10 -/- B cells reversed AngII-induced B cell-dependent atheroprotection, thus highlighting a protective role of IL-10+ regulatory B cells in this setting. Transfer of AngII type 1A receptor deficient (Agtr1a -/-) B cells into Apoe -/- /Baffr -/- mice substantially reduced the production of IL-10 by B cells and prevented the AngII-dependent atheroprotective B cell phenotype. Consistent with the in vivo data, AngII synergized with BAFF to induce IL-10 production by B cells in vitro via AngII type 1A receptor. Our data demonstrate a previously unknown synergy between AngII and BAFF in inducing IL-10 production by B cells, resulting in atheroprotection.


Subject(s)
Angiotensin II/metabolism , Atherosclerosis/etiology , Atherosclerosis/metabolism , B-Cell Activating Factor/genetics , B-Lymphocytes, Regulatory/metabolism , Animals , Atherosclerosis/pathology , B-Lymphocytes, Regulatory/immunology , Biomarkers , Cells, Cultured , Disease Models, Animal , Immunohistochemistry , Interleukin-10/biosynthesis , Mice , Mice, Knockout , Phenotype , Receptor, Angiotensin, Type 1/deficiency
5.
J Autoimmun ; 75: 141-149, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27543048

ABSTRACT

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.


Subject(s)
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
6.
Immunol Cell Biol ; 94(10): 981-993, 2016 11.
Article in English | MEDLINE | ID: mdl-27359298

ABSTRACT

The immunological synapse formed between a T-cell and an antigen-presenting cell is important for cell-cell communication during T-cell-mediated immune responses. Immunological synapse formation begins with stimulation of the T-cell receptor (TCR). TCR microclusters are assembled and transported to the center of the immunological synapse in an actin polymerization-dependent process. However, the physical link between TCR and actin remains elusive. Here we show that lymphocyte-specific Crk-associated substrate (Cas-L), a member of a force sensing protein family, is required for transport of TCR microclusters and for establishing synapse stability. We found that Cas-L is phosphorylated at TCR microclusters in an actin polymerization-dependent fashion. Furthermore, Cas-L participates in a positive feedback loop leading to amplification of Ca2+ signaling, inside-out integrin activation, and actomyosin contraction. We propose a new role for Cas-L in T-cell activation as a mechanical transducer linking TCR microclusters to the underlying actin network and coordinating multiple actin-dependent structures in the immunological synapse. Our studies highlight the importance of mechanotransduction processes in T-cell-mediated immune responses.


Subject(s)
Actins/metabolism , Crk-Associated Substrate Protein/metabolism , Immunological Synapses/metabolism , Polymerization , Animals , Calcium/metabolism , Cell Adhesion , Crk-Associated Substrate Protein/deficiency , Integrins/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Models, Immunological , Phosphorylation , Protein Transport , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
7.
Arterioscler Thromb Vasc Biol ; 36(9): 1791-801, 2016 09.
Article in English | MEDLINE | ID: mdl-27417580

ABSTRACT

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.


Subject(s)
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
9.
Nat Neurosci ; 18(4): 511-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25706475

ABSTRACT

Axonal damage has been associated with aberrant protein trafficking. We examined a newly characterized class of compounds that target nucleo-cytoplasmic shuttling by binding to the catalytic groove of the nuclear export protein XPO1 (also known as CRM1, chromosome region maintenance protein 1). Oral administration of reversible CRM1 inhibitors in preclinical murine models of demyelination significantly attenuated disease progression, even when started after the onset of paralysis. Clinical efficacy was associated with decreased proliferation of immune cells, characterized by nuclear accumulation of cell cycle inhibitors, and preservation of cytoskeletal integrity even in demyelinated axons. Neuroprotection was not limited to models of demyelination, but was also observed in another mouse model of axonal damage (that is, kainic acid injection) and detected in cultured neurons after knockdown of Xpo1, the gene encoding CRM1. A proteomic screen for target molecules revealed that CRM1 inhibitors in neurons prevented nuclear export of molecules associated with axonal damage while retaining transcription factors modulating neuroprotection.


Subject(s)
Axons , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Karyopherins/metabolism , Neuroprotective Agents/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Acrylamides/administration & dosage , Acrylamides/pharmacokinetics , Acrylamides/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cell Nucleus/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Drug Evaluation, Preclinical , Female , Karyopherins/antagonists & inhibitors , Karyopherins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Proteomics , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Treatment Outcome , Exportin 1 Protein
10.
Int J Mol Sci ; 16(1): 1980-2000, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25603179

ABSTRACT

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.


Subject(s)
Central Tolerance/immunology , Disease Models, Animal , Epithelial Cells/pathology , Hepatitis, Autoimmune/immunology , Thymus Gland/pathology , Animals , Mice , T-Lymphocytes, Regulatory/immunology
11.
Arterioscler Thromb Vasc Biol ; 33(10): 2374-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23908246

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm is an inflammatory disease leading to destructive vascular remodeling and ultimately to lethal aortic rupture. Despite its frequent association with atherosclerosis, compelling studies have shown striking differences and potentially opposite roles of T-cell helper responses in aneurysm as compared with atherosclerosis, casting doubt on the relevance and suitability of T-cell-targeted therapies in this context. APPROACH AND RESULTS: Here, we show that selective depletion of T regulatory (Treg) cells using a CD25-specific monoclonal antibody significantly enhances the susceptibility of C57Bl/6 mice to angiotensin II-induced abdominal aortic aneurysm and promotes aortic rupture (n=25-44 mice/group). Similar results are observed in angiotensin II-treated Cd80(-/-)/Cd86(-/-) or Cd28(-/-) mice with impaired Treg cell homeostasis (n=18-23 mice/group). Treg cell depletion is associated with increased immune cell activation and a blunted interleukin (IL)-10 anti-inflammatory response, suggesting an immunoinflammatory imbalance. Interestingly, Il-10(-/-) mice (n=20 mice/group) show increased susceptibility to angiotensin II-induced abdominal aortic aneurysm and aortic rupture and are insensitive to Treg cell depletion. Finally, reconstitution of Cd28(-/-) Treg-deficient mice with Treg cells (n=22 mice/group) restores a balance in the immunoinflammatory response, rescues the animals from increased susceptibility to aneurysm, and prevents aortic dissection. CONCLUSIONS: These results identify a critical role for Treg cells and IL-10 in the control of aneurysm formation and its progression to rupture and suggest that therapies targeting Treg responses may be most suited to treat aneurysmal disease.


Subject(s)
Angiotensin II , Aorta, Abdominal/immunology , Aortic Aneurysm, Abdominal/prevention & control , Aortic Rupture/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/pathology , Aortic Rupture/chemically induced , Aortic Rupture/immunology , Aortic Rupture/pathology , B7-1 Antigen/deficiency , B7-1 Antigen/genetics , B7-2 Antigen/deficiency , B7-2 Antigen/genetics , CD28 Antigens/deficiency , CD28 Antigens/genetics , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Leukocyte Reduction Procedures , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/transplantation , Time Factors
12.
Arterioscler Thromb Vasc Biol ; 33(3): 466-73, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23349189

ABSTRACT

OBJECTIVE: Several secreted phospholipases A2 (sPLA2s), including group IIA, III, V, and X, have been linked to the development of atherosclerosis, which led to the clinical testing of A-002 (varespladib), a broad sPLA2 inhibitor for the treatment of coronary artery disease. Group X sPLA2 (PLA2G10) has the most potent hydrolyzing activity toward phosphatidylcholine and is believed to play a proatherogenic role. METHODS AND RESULTS: Here, we show that Ldlr(-/-) mice reconstituted with bone marrow from mouse group X-deficient mice (Pla2g10(-/-)) unexpectedly display a doubling of plaque size compared with Pla2g10(+/+) chimeric mice. Macrophages of Pla2g10(-/-) mice are more susceptible to apoptosis in vitro, which is associated with a 4-fold increase of plaque necrotic core in vivo. In addition, chimeric Pla2g10(-/-) mice show exaggerated T lymphocyte (Th)1 immune response, associated with enhanced T-cell infiltration in atherosclerotic plaques. Interestingly, overexpression of human PLA2G10 in murine bone marrow cells leads to significant reduction of Th1 response and to 50% reduction of lesion size. CONCLUSIONS: PLA2G10 expression in bone marrow cells controls a proatherogenic Th1 response and limits the development of atherosclerosis. The results may provide an explanation for the recently reported inefficacy of A-002 (varespladib) to treat patients with coronary artery disease. Indeed, A-002 is a nonselective sPLA2 inhibitor that inhibits both proatherogenic (groups IIA and V) and antiatherogenic (group X) sPLA2s. Our results suggest that selective targeting of individual sPLA2 enzymes may be a better strategy to treat cardiovascular diseases.


Subject(s)
Aorta, Thoracic/enzymology , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Group X Phospholipases A2/metabolism , Receptors, LDL/deficiency , Adaptive Immunity , Animals , Aorta, Thoracic/immunology , Aorta, Thoracic/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Apoptosis , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Transplantation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Group X Phospholipases A2/deficiency , Group X Phospholipases A2/genetics , Humans , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Necrosis , Plaque, Atherosclerotic , Receptors, LDL/genetics , Th1 Cells/immunology , Time Factors
13.
Arterioscler Thromb Vasc Biol ; 32(3): 605-12, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22223728

ABSTRACT

OBJECTIVE: The immunoinflammatory response plays a critical role in the development and progression of atherosclerosis. Recent studies suggested an important role for regulatory T (Treg) cells in the inhibition of disease-related vascular inflammation. We hypothesized that induction of a specific Treg cell response to atherosclerosis-relevant antigens would be an attractive strategy to limit the development and progression of atherosclerosis through the promotion of immune tolerance. METHODS AND RESULTS: Young or old Apoe-/- mice were subcutaneously infused for 2 weeks with either a control ovalbumin (OVA) peptide or with apolipoprotein B100 (ApoB100)-derived peptides without adjuvant. Atherosclerosis development, progression and immunologic status were assessed at 8 weeks after the end of the infusion. Treatment with ApoB100 peptides led to significant reduction of lesion development in young Apoe-/- mice (P=0.001 versus OVA group) and abrogated atherosclerosis progression in old Apoe-/- mice with already established lesions (0% progression in ApoB100 versus 17% in OVA group, P<0.005). Limitation of plaque progression was associated with reduced vascular inflammation and increased collagen content, indicative of plaque stabilization. Infusion of ApoB100 peptides did not alterantibody production but promoted a specific Treg cell response, which was associated with a reduction of both T helper type 1-related and T helper type 2-related cytokines. Interestingly, depletion of CD4+CD25+ Treg cells abrogated ApoB100 peptides-dependent immune modulation and atheroprotection. CONCLUSION: Subcutaneous infusion of adjuvant-free ApoB100-derived peptides to Apoe-/- mice reduces atherosclerosis through the induction of a specific Treg cell response.


Subject(s)
Apolipoprotein B-100/administration & dosage , Atherosclerosis/prevention & control , Immune Tolerance , Immunotherapy/methods , Lymphocyte Activation , Peptides/administration & dosage , T-Lymphocytes, Regulatory/immunology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Infusions, Subcutaneous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , T-Lymphocytes, Regulatory/metabolism , Time Factors
14.
Nat Rev Cardiol ; 8(6): 348-58, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21502963

ABSTRACT

Chronic inflammation drives the development of atherosclerosis, and adaptive immunity is deeply involved in this process. Initial studies attributed a pathogenic role to T cells in atherosclerosis, mainly owing to the proatherogenic role of the T-helper (T(H))-1 cell subset, whereas the influence of T(H)2 and T(H)17 subsets is still debated. Today we know that T regulatory cells play a critical role in the protection against atherosclerotic lesion development and inflammation. In contrast to T cells, B cells were initially considered to be protective in atherosclerosis, assumingly through the production of protective antibodies against oxidized LDL. This concept has now been refined and proatherogenic roles of certain mature B cell subsets have been identified. We review the current knowledge about the role of various lymphocyte subsets in the development and progression of atherosclerosis and highlight future targets for immunomodulatory therapy.


Subject(s)
Adaptive Immunity , Atherosclerosis/immunology , B-Lymphocyte Subsets/immunology , Inflammation/immunology , T-Lymphocyte Subsets/immunology , Adaptive Immunity/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , B-Lymphocyte Subsets/drug effects , Humans , Immunologic Factors/therapeutic use , Inflammation/drug therapy , Inflammation Mediators/metabolism , T-Lymphocyte Subsets/drug effects , Vaccines
16.
J Exp Med ; 207(8): 1579-87, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-20603314

ABSTRACT

B cell depletion significantly reduces the burden of several immune-mediated diseases. However, B cell activation has been until now associated with a protection against atherosclerosis, suggesting that B cell-depleting therapies would enhance cardiovascular risk. We unexpectedly show that mature B cell depletion using a CD20-specific monoclonal antibody induces a significant reduction of atherosclerosis in various mouse models of the disease. This treatment preserves the production of natural and potentially protective anti-oxidized low-density lipoprotein (oxLDL) IgM autoantibodies over IgG type anti-oxLDL antibodies, and markedly reduces pathogenic T cell activation. B cell depletion diminished T cell-derived IFN-gamma secretion and enhanced production of IL-17; neutralization of the latter abrogated CD20 antibody-mediated atheroprotection. These results challenge the current paradigm that B cell activation plays an overall protective role in atherogenesis and identify new antiatherogenic strategies based on B cell modulation.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/prevention & control , B-Lymphocytes/immunology , Lymphocyte Depletion , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , Apolipoproteins E/genetics , Atherosclerosis/pathology , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Proliferation , Cholesterol/blood , Dendritic Cells/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Lipoproteins, LDL/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/genetics , Sinus of Valsalva/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
17.
J Clin Invest ; 120(2): 422-32, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20101093

ABSTRACT

Complicated abdominal aortic aneurysm (AAA) is a major cause of mortality in elderly men. Ang II-dependent TGF-beta activity promotes aortic aneurysm progression in experimental Marfan syndrome. However, the role of TGF-beta in experimental models of AAA has not been comprehensively assessed. Here, we show that systemic neutralization of TGF-beta activity breaks the resistance of normocholesterolemic C57BL/6 mice to Ang II-induced AAA formation and markedly increases their susceptibility to the disease. These aneurysms displayed a large spectrum of complications on echography, including fissuration, double channel formation, and rupture, leading to death from aneurysm complications. The disease was refractory to inhibition of IFN-gamma, IL-4, IL-6, or TNF-alpha signaling. Genetic deletion of T and B cells or inhibition of the CX3CR1 pathway resulted in partial protection. Interestingly, neutralization of TGF-beta activity enhanced monocyte invasiveness, and monocyte depletion markedly inhibited aneurysm progression and complications. Finally, TGF-beta neutralization increased MMP-12 activity, and MMP-12 deficiency prevented aneurysm rupture. These results clearly identify a critical role for TGF-beta in the taming of the innate immune response and the preservation of vessel integrity in C57BL/6 mice, which contrasts with its reported pathogenic role in Marfan syndrome.


Subject(s)
Angiotensin II/toxicity , Aortic Aneurysm, Abdominal/drug therapy , Inflammation/physiopathology , Transforming Growth Factor beta/pharmacology , Angiotensin II/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Disease Progression , Inflammation/immunology , Inflammation/prevention & control , Male , Matrix Metalloproteinase 12/deficiency , Matrix Metalloproteinase 2/deficiency , Mice , Mice, Inbred C57BL , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Ultrasonography
18.
Circ Cardiovasc Genet ; 2(4): 379-88, 2009 Aug.
Article in English | MEDLINE | ID: mdl-20031610

ABSTRACT

BACKGROUND: Microparticles (MPs) with procoagulant activity are present in human atherosclerosis, but no detailed information is available on their composition. METHODS AND RESULTS: To obtain insights into the role of MPs in atherogenesis, MP proteins were identified by tandem mass spectrometry, metabolite profiles were determined by high-resolution nuclear magnetic resonance spectroscopy, and antibody reactivity was assessed against combinatorial antigen libraries. Plaque MPs expressed surface antigens consistent with their leukocyte origin, including major histocompatibility complex classes I and II, and induced a dose-dependent stimulatory effect on T-cell proliferation. Notably, taurine, the most abundant free organic acid in human neutrophils, which scavenges myeloperoxidase-catalyzed free radicals, was highly enriched in plaque MPs. Moreover, fluorescent labeling of proteins on the MP surface suggested immunoglobulins to be trapped inside, which was confirmed by flow cytometry analysis on permeabilized and nonpermeabilized plaque MPs. Colabeling for CD14 and IgG established that more than 90% of the IgG containing MPs were CD14(+), indicating a macrophage origin. Screening against an antigen library revealed that the immunologic profiles of antibodies in MPs were similar to those found in plaques but differed profoundly from antibodies in plasma and unexpectedly, showed strong reactions with oligosaccharide antigens, in particular blood group antigen A. CONCLUSIONS: This study provides the first evidence that immunoglobulins are present within MPs derived from plaque macrophages, that the portfolio of plaque antibodies is different from circulating antibodies in plasma, and that anticarbohydrate antibodies are retained in human atherosclerotic lesions.


Subject(s)
Atherosclerosis/immunology , Cell-Derived Microparticles/immunology , Amino Acid Sequence , Antibodies/immunology , Antibodies/metabolism , Atherosclerosis/metabolism , Cell-Derived Microparticles/metabolism , Databases, Genetic , Electrophoresis, Gel, Two-Dimensional , Flow Cytometry , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Lipopolysaccharide Receptors/metabolism , Metabolomics , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Particle Size , Proteomics , Tandem Mass Spectrometry , Taurine/metabolism
19.
J Exp Med ; 206(10): 2067-77, 2009 Sep 28.
Article in English | MEDLINE | ID: mdl-19737863

ABSTRACT

Atherosclerosis is an inflammatory vascular disease responsible for the first cause of mortality worldwide. Recent studies have clearly highlighted the critical role of the immunoinflammatory balance in the modulation of disease development and progression. However, the immunoregulatory pathways that control atherosclerosis remain largely unknown. We show that loss of suppressor of cytokine signaling (SOCS) 3 in T cells increases both interleukin (IL)-17 and IL-10 production, induces an antiinflammatory macrophage phenotype, and leads to unexpected IL-17-dependent reduction in lesion development and vascular inflammation. In vivo administration of IL-17 reduces endothelial vascular cell adhesion molecule-1 expression and vascular T cell infiltration, and significantly limits atherosclerotic lesion development. In contrast, overexpression of SOCS3 in T cells reduces IL-17 and accelerates atherosclerosis. We also show that in human lesions, increased levels of signal transducer and activator of transcription (STAT) 3 phosphorylation and IL-17 are associated with a stable plaque phenotype. These results identify novel SOCS3-controlled IL-17 regulatory pathways in atherosclerosis and may have important implications for the understanding of the increased susceptibility to vascular inflammation in patients with dominant-negative STAT3 mutations and defective Th17 cell differentiation.


Subject(s)
Atherosclerosis/etiology , Interleukin-17/physiology , Suppressor of Cytokine Signaling Proteins/physiology , T-Lymphocytes/physiology , Animals , Female , Humans , Interleukin-10/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Receptors, LDL/physiology , STAT3 Transcription Factor/physiology , Suppressor of Cytokine Signaling 3 Protein , Vascular Cell Adhesion Molecule-1/analysis , Vasculitis/etiology
20.
Circulation ; 116(15): 1707-13, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17875970

ABSTRACT

BACKGROUND: Recent studies clearly suggest that regulatory T cells play a critical role in the control of the immunoinflammatory response in atherosclerosis and substantially limit lesion development. Measles virus infection or vaccination is associated with immune depression, in part through the induction of an antiinflammatory response by measles virus nucleoprotein. We hypothesized that the antiinflammatory properties of measles virus nucleoprotein may limit the development atherosclerosis. METHODS AND RESULTS: Here, we show for the first time that repetitive administration of measles virus nucleoprotein to apolipoprotein E-deficient mice promotes an antiinflammatory T-regulatory-cell type 1-like response and inhibits macrophage and T-cell accumulation within the lesions. Treatment with measles virus nucleoprotein significantly reduces the development of new atherosclerotic plaques and markedly inhibits the progression of established lesions. The antiatherosclerotic potential of nucleoprotein is retained in its short N-terminal segment. The protective effects on lesion size are lost in mice with lymphocyte deficiency. CONCLUSIONS: Our findings identify a novel mechanism of immune modulation by measles virus nucleoprotein through the promotion of a regulatory T-cell response and suggest that this property may be harnessed for treating atherosclerosis, the first cause of heart disease and stroke.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/immunology , Atherosclerosis/prevention & control , Measles virus/immunology , Nucleoproteins/immunology , Viral Proteins/immunology , Animals , Baculoviridae , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Genetic Vectors , Inflammation/immunology , Inflammation/prevention & control , Male , Mice , Mice, Knockout , Nucleoproteins/genetics , Nucleoproteins/therapeutic use , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , T-Lymphocytes, Regulatory/immunology , Viral Proteins/genetics , Viral Proteins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...