Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 8(8)2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-37092548

RESUMO

Fibroblastic reticular cells (FRCs) play important roles in tolerance by producing laminin α4 (Lama4) and altering lymph node (LN) structure and function. The present study revealed the specific roles of extracellular matrix Lama4 in regulating LN conduits using FRC-specific KO mouse strains. FRC-derived Lama4 maintained conduit fiber integrity, as its depletion altered conduit morphology and structure and reduced homeostatic conduit flow. Lama4 regulated the lymphotoxin ß receptor (LTßR) pathway, which is critical for conduit and LN integrity. Depleting LTßR in FRCs further reduced conduits and impaired reticular fibers. Lama4 was indispensable for FRC generation and survival, as FRCs lacking Lama4 displayed reduced proliferation but upregulated senescence and apoptosis. During acute immunization, FRC Lama4 deficiency increased antigen flow through conduits. Importantly, adoptive transfer of WT FRCs to FRC Lama4-deficient mice rescued conduit structure, ameliorated Treg and chemokine distribution, and restored transplant allograft acceptance, which were all impaired by FRC Lama4 depletion. Single-cell RNA sequencing analysis of LN stromal cells indicated that the laminin and collagen signaling pathways linked crosstalk among FRC subsets and endothelial cells. This study demonstrated that FRC Lama4 is responsible for maintaining conduits by FRCs and can be harnessed to potentiate FRC-based immunomodulation.


Assuntos
Células Endoteliais , Laminina , Camundongos , Animais , Laminina/genética , Laminina/metabolismo , Linfonodos , Transdução de Sinais , Quimiocinas/metabolismo
2.
Nat Commun ; 14(1): 681, 2023 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-36755035

RESUMO

Antigen-specific tolerance is a key goal of experimental immunotherapies for autoimmune disease and allograft rejection. This outcome could selectively inhibit detrimental inflammatory immune responses without compromising functional protective immunity. A major challenge facing antigen-specific immunotherapies is ineffective control over immune signal targeting and integration, limiting efficacy and causing systemic non-specific suppression. Here we use intra-lymph node injection of diffusion-limited degradable microparticles that encapsulate self-antigens with the immunomodulatory small molecule, rapamycin. We show this strategy potently inhibits disease during pre-clinical type 1 diabetes and allogenic islet transplantation. Antigen and rapamycin are required for maximal efficacy, and tolerance is accompanied by expansion of antigen-specific regulatory T cells in treated and untreated lymph nodes. The antigen-specific tolerance in type 1 diabetes is systemic but avoids non-specific immune suppression. Further, microparticle treatment results in the development of tolerogenic structural microdomains in lymph nodes. Finally, these local structural and functional changes in lymph nodes promote memory markers among antigen-specific regulatory T cells, and tolerance that is durable. This work supports intra-lymph node injection of tolerogenic microparticles as a powerful platform to promote antigen-dependent efficacy in type 1 diabetes and allogenic islet transplantation.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Humanos , Tolerância Imunológica , Autoantígenos , Linfonodos/patologia , Sirolimo
3.
J Clin Invest ; 132(13)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35775481

RESUMO

Lymph node (LN) fibroblastic reticular cells (FRCs) define LN niches and regulate lymphocyte homeostasis through producing diverse extracellular matrix (ECM) components. We examined the role of ECM laminin α4 (Lama4) using FRC-Lama4 conditional KO Pdgfrb-Cre-/- × Lama4fl/fl mice. Single-cell RNA-sequencing (scRNA-Seq) data showed the promoter gene Pdgfrb was exclusively expressed in FRCs. Depleting FRC-Lama4 reduced Tregs and dendritic cells, decreased high endothelial venules, impaired the conduit system, and downregulated T cell survival factors in LNs. FRC-Lama4 depletion impaired the homing of lymphocytes to LNs in homeostasis and after allografting. Alloantigen-specific T cells proliferated, were activated to greater degrees in LNs lacking FRC-Lama4, and were more prone to differentiate into effector phenotypes relative to the Treg phenotype. In murine cardiac transplantation, tolerogenic immunosuppression was not effective in FRC-Lama4 recipients, which produced more alloantibodies than WT. After lung transplantation, FRC-Lama4-KO mice had more severe graft rejection with fewer Tregs in their LNs. Overall, FRC-Lama4 critically contributes to a tolerogenic LN niche by supporting T cell migration, constraining T cell activation and proliferation, and promoting Treg differentiation. Hence, it serves as a therapeutic target for immunoengineering.


Assuntos
Laminina , Linfonodos , Reticulina , Linfócitos T , Animais , Laminina/imunologia , Linfonodos/imunologia , Camundongos , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Reticulina/imunologia , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Imunologia de Transplantes
4.
Cell Rep ; 39(3): 110727, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-35443187

RESUMO

Regulatory T cell (Treg) lymphatic migration is required for resolving inflammation and prolonging allograft survival. Focusing on Treg interactions with lymphatic endothelial cells (LECs), we dissect mechanisms and functional consequences of Treg transendothelial migration (TEM). Using three genetic mouse models of pancreatic islet transplantation, we show that Treg lymphotoxin (LT) αß and LEC LTß receptor (LTßR) signaling are required for efficient Treg migration and suppressive function to prolong allograft survival. Inhibition of LT signaling increases Treg conversion to Foxp3loCD25lo exTregs. In a transwell-based model of TEM across polarized LECs, non-migrated Tregs become exTregs. Such conversion is regulated by LTßR nuclear factor κB (NF-κB) signaling in LECs, which increases interleukin-6 (IL-6) production and drives exTreg conversion. Migrating Tregs are ectonucleotidase CD39hi and resist exTreg conversion in an adenosine-receptor-2A-dependent fashion. Human Tregs migrating across human LECs behave similarly. These molecular interactions can be targeted for therapeutic manipulation of immunity and suppression.


Assuntos
Células Endoteliais , Linfócitos T Reguladores , Adenosina , Animais , Fatores de Transcrição Forkhead/genética , Linfotoxina-beta , Camundongos , NF-kappa B
6.
J Clin Invest ; 130(5): 2602-2619, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32017712

RESUMO

Lymph node stromal cells (LNSCs) regulate immunity through constructing lymphocyte niches. LNSC-produced laminin α5 (Lama5) regulates CD4+ T cells but the underlying mechanisms of its functions are poorly understood. Here we show that depleting Lama5 in LNSCs resulted in decreased Lama5 protein in the LN cortical ridge (CR) and around high endothelial venules (HEVs). Lama5 depletion affected LN structure with increased HEVs, upregulated chemokines, and cell adhesion molecules, and led to greater numbers of Tregs in the T cell zone. Mouse and human T cell transendothelial migration and T cell entry into LNs were suppressed by Lama5 through the receptors α6 integrin and α-dystroglycan. During immune responses and allograft transplantation, depleting Lama5 promoted antigen-specific CD4+ T cell entry into the CR through HEVs, suppressed T cell activation, and altered T cell differentiation to suppressive regulatory phenotypes. Enhanced allograft acceptance resulted from depleting Lama5 or blockade of T cell Lama5 receptors. Lama5 and Lama4/Lama5 ratios in allografts were associated with the rejection severity. Overall, our results demonstrated that stromal Lama5 regulated immune responses through altering LN structures and T cell behaviors. This study delineated a stromal Lama5-T cell receptor axis that can be targeted for immune tolerance modulation.


Assuntos
Laminina/imunologia , Linfonodos/imunologia , Tolerância ao Transplante/imunologia , Animais , Distroglicanas/metabolismo , Humanos , Integrina alfa6/metabolismo , Laminina/genética , Laminina/metabolismo , Linfonodos/citologia , Linfonodos/metabolismo , Vasos Linfáticos/citologia , Vasos Linfáticos/imunologia , Vasos Linfáticos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Estromais/citologia , Células Estromais/imunologia , Células Estromais/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Migração Transendotelial e Transepitelial/imunologia
7.
Cell Rep ; 30(4): 1052-1062.e5, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31995749

RESUMO

Regulatory T cells (Tregs) express high levels of cell surface lymphotoxin alpha beta (LTα1ß2) to activate the LT beta receptor (LTßR) on the lymphatic endothelial cells (LECs), modulating LEC adhesion molecules, intercellular junctions, and chemokines. We demonstrate a role for Tregs through this pathway to condition the permissiveness of lymphatic endothelia for transendothelial migration (TEM), thus gating leukocyte traffic. Human Tregs share the same property with murine Tregs. Activation of TLR2 on Tregs during inflammation specifically augments LTα1ß2-LTßR signaling, which further enhances the permissiveness of LECs to facilitate TEM. The conditioning of endothelia may promote the resolution of inflammation by directing leukocytes out of tissues to lymphatic vessels and draining lymph nodes (dLNs). Thus, Tregs interact with lymphatic endothelia under homeostasis and inflammation and dictate endothelial permissiveness and gating mechanisms for subsequent leukocyte migration through endothelial barriers.


Assuntos
Movimento Celular/imunologia , Endotélio Linfático/metabolismo , Inflamação/metabolismo , Linfócitos T Reguladores/metabolismo , Receptor 2 Toll-Like/metabolismo , Migração Transendotelial e Transepitelial/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Caderinas/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Quimiocina CCL21/metabolismo , Endotélio Linfático/efeitos dos fármacos , Humanos , Inflamação/imunologia , Inflamação/patologia , Interleucina-2/farmacologia , Ilhotas Pancreáticas/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Receptor beta de Linfotoxina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Protocaderinas , Receptores de Interleucina-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Receptor 2 Toll-Like/imunologia , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo
8.
Nat Commun ; 10(1): 2927, 2019 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-31249309

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

9.
J Exp Med ; 216(7): 1582-1598, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31147448

RESUMO

Sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) activate G protein-coupled receptors (GPCRs) to regulate biological processes. Using a genome-wide CRISPR/dCas9-based GPCR signaling screen, LPAR1 was identified as an inducer of S1PR1/ß-arrestin coupling while suppressing Gαi signaling. S1pr1 and Lpar1-positive lymphatic endothelial cells (LECs) of lymph nodes exhibit constitutive S1PR1/ß-arrestin signaling, which was suppressed by LPAR1 antagonism. Pharmacological inhibition or genetic loss of function of Lpar1 reduced the frequency of punctate junctions at sinus-lining LECs. Ligand activation of transfected LPAR1 in endothelial cells remodeled junctions from continuous to punctate structures and increased transendothelial permeability. In addition, LPAR1 antagonism in mice increased lymph node retention of adoptively transferred lymphocytes. These data suggest that cross-talk between LPAR1 and S1PR1 promotes the porous junctional architecture of sinus-lining LECs, which enables efficient lymphocyte trafficking. Heterotypic inter-GPCR coupling may regulate complex cellular phenotypes in physiological milieu containing many GPCR ligands.


Assuntos
Células Endoteliais/metabolismo , Linfonodos/metabolismo , Receptor Cross-Talk , Animais , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Imunofluorescência , Edição de Genes , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Lisofosfolipídeos/metabolismo , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo
10.
Sci Immunol ; 4(33)2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30877143

RESUMO

Sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs) regulate migration of lymphocytes out of thymus to blood and lymph nodes (LNs) to efferent lymph, whereas their role in other tissue sites is not known. Here, we investigated the question of how these molecules regulate leukocyte migration from tissues through afferent lymphatics to draining LNs (dLNs). S1P, but not other chemokines, selectively enhanced human and murine CD4 T cell migration across lymphatic endothelial cells (LECs). T cell S1PR1 and S1PR4, and LEC S1PR2, were required for migration across LECs and into lymphatic vessels and dLNs. S1PR1 and S1PR4 differentially regulated T cell motility and vascular cell adhesion molecule-1 (VCAM-1) binding. S1PR2 regulated LEC layer structure, permeability, and expression of the junction molecules VE-cadherin, occludin, and zonulin-1 through the ERK pathway. S1PR2 facilitated T cell transcellular migration through VCAM-1 expression and recruitment of T cells to LEC migration sites. These results demonstrated distinct roles for S1PRs in comodulating T cell and LEC functions in migration and suggest previously unknown levels of regulation of leukocytes and endothelial cells during homeostasis and immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Movimento Celular/imunologia , Células Endoteliais/imunologia , Vasos Linfáticos/imunologia , Receptores de Esfingosina-1-Fosfato/imunologia , Animais , Linfócitos T CD4-Positivos/fisiologia , Linhagem Celular , Células Endoteliais/fisiologia , Humanos , Lisofosfolipídeos/imunologia , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esfingosina/análogos & derivados , Esfingosina/imunologia , Receptores de Esfingosina-1-Fosfato/genética , Proteínas de Junções Íntimas/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia
11.
JCI Insight ; 3(19)2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30282817

RESUMO

We hypothesized that the gut microbiota influences survival of murine cardiac allografts through modulation of immunity. Antibiotic pretreated mice received vascularized cardiac allografts and fecal microbiota transfer (FMT), along with tacrolimus immunosuppression. FMT source samples were from normal, pregnant (immune suppressed), or spontaneously colitic (inflammation) mice. Bifidobacterium pseudolongum (B. pseudolongum) in pregnant FMT recipients was associated with prolonged allograft survival and lower inflammation and fibrosis, while normal or colitic FMT resulted in inferior survival and worse histology. Transfer of B. pseudolongum alone resulted in reduced inflammation and fibrosis. Stimulation of DC and macrophage lines with B. pseudolongum induced the antiinflammatory cytokine IL-10 and homeostatic chemokine CCL19 but induced lesser amounts of the proinflammatory cytokines TNFα and IL-6. In contrast, LPS and Desulfovibrio desulfuricans (D. desulfuricans), more abundant in colitic FMT, induced a more inflammatory cytokine response. Analysis of mesenteric and peripheral lymph node structure showed that B. pseudolongum gavage resulted in a higher laminin α4/α5 ratio in the lymph node cortical ridge, indicative of a suppressive environment, while D. desulfuricans resulted in a lower laminin α4/α5 ratio, supportive of inflammation. Discrete gut bacterial species alter immunity and may predict graft outcomes through stimulation of myeloid cells and shifts in lymph node structure and permissiveness.


Assuntos
Microbioma Gastrointestinal/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração/efeitos adversos , Imunidade Inata , Linfonodos/imunologia , Aloenxertos/imunologia , Aloenxertos/patologia , Animais , Antibacterianos/administração & dosagem , Linhagem Celular Tumoral , Colite/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Rejeição de Enxerto/patologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Humanos , Imunossupressores/administração & dosagem , Camundongos , Miocárdio/patologia , Gravidez , Células RAW 264.7 , Tacrolimo/administração & dosagem , Resultado do Tratamento
12.
Nat Commun ; 9(1): 3020, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30069025

RESUMO

Lymphotoxin-beta receptor (LTßR) signaling in lymphatic endothelial cells (LEC) regulates leukocyte afferent lymphatic transendothelial migration (TEM). The function of individual signaling pathways for different leukocyte subsets is currently unknown. Here, we show that LTßR signals predominantly via the constitutive and ligand-driven non-classical NIK pathway. Targeting LTßR-NIK by an LTßR-derived decoy peptide (nciLT) suppresses the production of chemokines CCL21 and CXCL12, and enhances the expression of classical NFκB-driven VCAM-1 and integrin ß4 to retain T cells on LEC and precludes T cell and dendritic cell TEM. nciLT inhibits contact hypersensitivity (CHS) at both the sensitization and elicitation stages, likely by inhibiting leukocyte migration. By contrast, targeting LTßR-classical NFκB signaling during the elicitation and resolution stages attenuates CHS, possibly by promoting leukocyte egress. These findings demonstrate the importance of LTßR signaling in leukocyte migration and LEC and lymphatic vessel function, and show that antagonist peptides may serve as lead compounds for therapeutic applications.


Assuntos
Movimento Celular , Vasos Linfáticos/metabolismo , Receptor beta de Linfotoxina/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Animais , Anticorpos Monoclonais/farmacologia , Movimento Celular/efeitos dos fármacos , Dermatite de Contato/imunologia , Dermatite de Contato/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Inflamação/patologia , Camundongos Endogâmicos C57BL , Peptídeos/metabolismo , Linfócitos T/efeitos dos fármacos
13.
Genome Announc ; 5(40)2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28983003

RESUMO

Here, we report the complete genome sequence of Bifidobacterium pseudolongum strain UMB-MBP-01, isolated from the feces of C57BL/6J mice. This strain was identified in microbiome profiling studies and associated with improved transplant outcome in a murine model of cardiac heterotypic transplantation.

14.
Sci Rep ; 7(1): 1633, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28487567

RESUMO

Trans-endothelial migration (TEM) is essential for leukocyte circulation. While much is known about trans-blood endothelial migration, far less is known about trans-lymphatic endothelial migration. We established an in vitro system to evaluate lymphatic TEM for various cell types across primary mouse and human lymphatic endothelial cells (LEC), and validated the model for the murine LEC cell line SVEC4-10. T cells exhibited enhanced unidirectional migration from the basal (abluminal) to the apical (luminal) surface across LEC, whereas for blood endothelial cells (BEC) they migrated similarly in both directions. This preferential, vectorial migration was chemotactic toward many different chemoattractants and dose-dependent. Stromal protein fibers, interstitial type fluid flow, distribution of chemokines in the stromal layer, and inflammatory cytokines influenced LEC phenotype and leukocyte TEM. Activated and memory CD4 T cells, macrophages, and dendritic cell (DC) showed chemoattractantΔdriven vectorial migration, while CD8 T cell migration across LEC was not. The system was further validated for studying cancer cell transmigration across lymphatic endothelium. This model for lymphatic TEM for various migrating and endothelial cell types possesses the capacity to be high-throughput, highly reproducible and integrate the complexities of lymphatic biology, stromal variability, chemoattractant distribution, and fluid flow.


Assuntos
Movimento Celular , Células Endoteliais/citologia , Endotélio Linfático/citologia , Modelos Biológicos , Animais , Linfócitos T CD4-Positivos/citologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Microambiente Celular , Quimiocinas/metabolismo , Quimiotaxia , Humanos , Inflamação/patologia , Camundongos Endogâmicos C57BL , Fenótipo
15.
Nat Commun ; 7: 12021, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27323847

RESUMO

Regulatory T cells (Tregs) are essential to suppress unwanted immunity or inflammation. After islet allo-transplant Tregs must migrate from blood to allograft, then via afferent lymphatics to draining LN to protect allografts. Here we show that Tregs but not non-Treg T cells use lymphotoxin (LT) during migration from allograft to draining LN, and that LT deficiency or blockade prevents normal migration and allograft protection. Treg LTαß rapidly modulates cytoskeletal and membrane structure of lymphatic endothelial cells; dependent on VCAM-1 and non-canonical NFκB signalling via LTßR. These results demonstrate a form of T-cell migration used only by Treg in tissues that serves an important role in their suppressive function and is a unique therapeutic focus for modulating suppression.


Assuntos
Diabetes Mellitus Experimental/terapia , Rejeição de Enxerto/imunologia , Transplante das Ilhotas Pancreáticas , Heterotrímero de Linfotoxina alfa1 e beta2/imunologia , Receptor beta de Linfotoxina/imunologia , Linfócitos T Reguladores/imunologia , Migração Transendotelial e Transepitelial/imunologia , Animais , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/mortalidade , Células Endoteliais/citologia , Células Endoteliais/imunologia , Regulação da Expressão Gênica , Rejeição de Enxerto/genética , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/genética , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/cirurgia , Linfonodos/citologia , Linfonodos/imunologia , Vasos Linfáticos/citologia , Vasos Linfáticos/imunologia , Heterotrímero de Linfotoxina alfa1 e beta2/genética , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/imunologia , Transdução de Sinais , Análise de Sobrevida , Linfócitos T Reguladores/citologia , Transplante Homólogo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
16.
J Immunol ; 196(6): 2526-40, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26880765

RESUMO

T-bet is essential for natural regulatory T cells (nTreg) to regulate Th1 inflammation, but whether T-bet controls other Treg functions after entering the inflammatory site is unknown. In an islet allograft model, T-bet(-/-) nTreg, but not induced Treg, failed to prolong graft survival as effectively as wild-type Treg. T-bet(-/-) nTreg had no functional deficiency in vitro but failed to home from the graft to draining lymph nodes (dLN) as efficiently as wild type. T-bet regulated expression of adhesion- and migration-related molecules, influencing nTreg distribution in tissues, so that T-bet(-/-) nTreg remained in the grafts rather than migrating to lymphatics and dLN. In contrast, both wild-type and T-bet(-/-) CD4(+) conventional T cells and induced Treg migrated normally toward afferent lymphatics. T-bet(-/-) nTreg displayed instability in the graft, failing to suppress Ag-specific CD4(+) T cells and prevent their infiltration into the graft and dLN. Thus, T-bet regulates nTreg migration into afferent lymphatics and dLN and consequently their suppressive stability in vivo.


Assuntos
Quimiotaxia de Leucócito/imunologia , Tolerância Imunológica/imunologia , Proteínas com Domínio T/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Transplante das Ilhotas Pancreáticas/imunologia , Linfonodos/imunologia , Vasos Linfáticos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real
17.
J Innate Immun ; 8(2): 171-84, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26457672

RESUMO

Endotoxin tolerance protects the host by limiting excessive 'cytokine storm' during sepsis, but compromises the ability to counteract infections in septic shock survivors. It reprograms Toll-like receptor (TLR) 4 responses by attenuating the expression of proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators, but the mechanisms of reprogramming remain unclear. In this study, we demonstrate that the induction of endotoxin tolerance in human monocytes, THP-1 and MonoMac-6 cells inhibited lipopolysaccharide (LPS)-mediated phosphorylation of Lyn, c-Src and their recruitment to TLR4, but increased total protein phosphatase (PP) activity and the expression of protein tyrosine phosphatase (PTP) 1B, PP2A, PTP nonreceptor type (PTPN) 22 and mitogen-activated protein kinase phosphatase (MKP)-1. Chemical PP inhibitors, okadaic acid, dephostatin and cantharidic acid markedly decreased or completely abolished LPS tolerance, indicating the importance of phosphatases in endotoxin tolerization. Overexpression of PTPN22 decreased LPS-mediated nuclear factor (NF)-x03BA;B activation, p38 phosphorylation and CXCL8 gene expression, while PTPN22 ablation upregulated LPS-induced p65 NF-x03BA;B and p38 phosphorylation and the expression of TNF-α and pro-IL-1ß mRNA, indicating PTPN22 as an inhibitor of TLR4 signaling. Thus, LPS tolerance interferes with TLR4 signaling by inhibiting Lyn and c-Src phosphorylation and their recruitment to TLR4, while increasing the phosphatase activity and expression of PP2A, PTPN22, PTP1B and MKP1.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Tolerância Imunológica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Monócitos/imunologia , Fosfoproteínas Fosfatases/imunologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/imunologia , Quinases da Família src/imunologia , Proteína Tirosina Quinase CSK , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica/imunologia , Humanos , Tolerância Imunológica/genética , Lipopolissacarídeos/imunologia , Fosfoproteínas Fosfatases/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/genética , Quinases da Família src/genética
18.
J Leukoc Biol ; 98(6): 963-74, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26310831

RESUMO

Development of endotoxin tolerance in macrophages during sepsis reprograms Toll-like receptor 4 signaling to inhibit proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators and protects the host from excessive inflammation and tissue damage. However, endotoxin tolerance renders septic patients immunocompromised and unable to control secondary infections. Although previous studies have revealed the importance of several negative regulators of Toll-like receptor signaling in endotoxin tolerance, the role of Pellino proteins has not been addressed. The present report shows that the induction of endotoxin tolerance in vivo in mice and in vitro in human monocytes and THP-1 and MonoMac-6 macrophages increases the expression of Pellino-3. Overexpression of Pellino-3 in human embryonic kidney 293/Toll-like receptor 2 or 293/Toll-like receptor 4/myeloid differentiation factor-2 cells inhibited Toll-like receptor 2/4-mediated activation of nuclear factor-κB and induction of CXCL-8 mRNA, and Pellino-3 ablation increased these responses. Pellino-3-deficient THP-1 cells had elevated Toll-like receptor 2/4-driven tumor necrosis factor-α, interleukin-6 mRNA, and Toll-like receptor 4-driven CCL5 gene expression in response to Toll-like receptor agonists and heat-killed Escherichia coli and Staphylococcus aureus, cytokines controlled by the MyD88 and Toll-interleukin-1R domain-containing protein inducing interferon-ß-mediated pathways, respectively. In addition, deficiency in Pellino-3 slightly increased phagocytosis of heat-killed bacteria. Transfected Pellino-3 inhibited nuclear factor-κB activation driven by overexpression of MyD88, TIR domain-containing adapter inducing interferon-ß, interleukin-1R-associated kinase-1, and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase-1, TGF-ß-activated kinase 1, and tumor necrosis factor receptor-associated factor-6, and inhibited interleukin-1R-associated kinase 1 modifications and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase 1 phosphorylation. Finally, Pellino-3 ablation in THP-1 decreased the extent of endotoxin tolerization. Thus, Pellino-3 is involved in endotoxin tolerance and functions as a negative regulator of Toll-like receptor 2/4 signaling.


Assuntos
Tolerância Imunológica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Macrófagos Peritoneais/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/imunologia , Escherichia coli/imunologia , Humanos , Tolerância Imunológica/genética , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Staphylococcus aureus/imunologia , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Ubiquitina-Proteína Ligases/genética
19.
J Biol Chem ; 290(31): 19218-32, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26082489

RESUMO

Endotoxin tolerance reprograms Toll-like receptor (TLR) 4-mediated macrophage responses by attenuating induction of proinflammatory cytokines while retaining expression of anti-inflammatory and antimicrobial mediators. We previously demonstrated deficient TLR4-induced activation of IL-1 receptor-associated kinase (IRAK) 4, IRAK1, and TANK-binding kinase (TBK) 1 as critical hallmarks of endotoxin tolerance, but mechanisms remain unclear. In this study, we examined the role of the E3 ubiquitin ligase Pellino-1 in endotoxin tolerance and TLR signaling. LPS stimulation increased Pellino-1 mRNA and protein expression in macrophages from mice injected with saline and in medium-pretreated human monocytes, THP-1, and MonoMac-6 cells, whereas endotoxin tolerization abrogated LPS inducibility of Pellino-1. Overexpression of Pellino-1 in 293/TLR2 and 293/TLR4/MD2 cells enhanced TLR2- and TLR4-induced nuclear factor κB (NF-κB) and expression of IL-8 mRNA, whereas Pellino-1 knockdown reduced these responses. Pellino-1 ablation in THP-1 cells impaired induction of myeloid differentiation primary response protein (MyD88), and Toll-IL-1R domain-containing adapter inducing IFN-ß (TRIF)-dependent cytokine genes in response to TLR4 and TLR2 agonists and heat-killed Escherichia coli and Staphylococcus aureus, whereas only weakly affecting phagocytosis of heat-killed bacteria. Co-expressed Pellino-1 potentiated NF-κB activation driven by transfected MyD88, TRIF, IRAK1, TBK1, TGF-ß-activated kinase (TAK) 1, and TNFR-associated factor 6, whereas not affecting p65-induced responses. Mechanistically, Pellino-1 increased LPS-driven K63-linked polyubiquitination of IRAK1, TBK1, TAK1, and phosphorylation of TBK1 and IFN regulatory factor 3. These results reveal a novel mechanism by which endotoxin tolerance re-programs TLR4 signaling via suppression of Pellino-1, a positive regulator of MyD88- and TRIF-dependent signaling that promotes K63-linked polyubiquitination of IRAK1, TBK1, and TAK1.


Assuntos
Proteínas Nucleares/fisiologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Animais , Células HEK293 , Humanos , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase Quinases/metabolismo , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Monócitos/metabolismo , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo , Ativação Transcricional/imunologia , Ubiquitinação
20.
Curr Opin Organ Transplant ; 18(4): 393-401, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23838643

RESUMO

PURPOSE OF REVIEW: The mechanisms of tolerance induction and maintenance remain incompletely understood and have yet to be translated to clinical practice. Advances in imaging techniques have allowed precise examination of cell interactions in the lymph node, often in real time. Herein we review evidence that lymph node structure is dynamic and controls the character of the immune response in a multistep, multiplayer dance. T-cell responses in particular can be initiated or influenced in regions beyond the canonical T-cell zone. We propose that the cortical ridge is one such region required for induction and maintenance of tolerance. RECENT FINDINGS: Lymph node domains are more complex than T-cell and B-cell zones. Different domains are important for different types of immune responses. These domains are in part defined by dynamic, malleable physical structures that guide cell interactions and influence immune outcomes. SUMMARY: Further probing as to how lymph node stromal cells and fibers interact with and determine the character of immune responses should yield fundamental insights into tolerance and immunity. Manipulation of lymph node structure and associated unique cell types and molecules may allow therapeutic interventions in the tolerogenic process.


Assuntos
Linfócitos B/imunologia , Tolerância Imunológica/fisiologia , Linfonodos/imunologia , Linfócitos T/imunologia , Animais , Comunicação Celular , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...