Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Front Immunol ; 12: 692127, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34305924

RESUMO

Mucus secretion is an important feature of asthma that highly correlates with morbidity. Current therapies, including administration of mucolytics and anti-inflammatory drugs, show limited effectiveness and durability, underscoring the need for novel effective and longer lasting therapeutic approaches. Here we show that mucus production in the lungs is regulated by the TNF superfamily member 15 (TL1A) acting through the mucus-inducing cytokine IL-13. TL1A induces IL13 expression by innate lymphoid cells leading to mucus production, in addition to promoting airway inflammation and fibrosis. Reciprocally, neutralization of IL13 signaling through its receptor (IL4Rα), completely reverses TL1A-induced mucus secretion, while maintaining airway inflammation and fibrosis. Importance of TL1A is further demonstrated using a preclinical asthma model induced by chronic house dust mite exposure where TL1A neutralization by genetic deletion or antagonistic blockade of its receptor DR3 protected against mucus production and fibrosis. Thus, TL1A presents a promising therapeutic target that out benefits IL13 in reversing mucus production, airway inflammation and fibrosis, cardinal features of severe asthma in humans.


Assuntos
Asma/imunologia , Interleucina-13/imunologia , Subunidade alfa de Receptor de Interleucina-4/imunologia , Pulmão/imunologia , Muco/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Asma/patologia , Proteínas de Ligação a DNA/genética , Feminino , Fibrose , Pulmão/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Transdução de Sinais
2.
J Immunol ; 205(12): 3348-3357, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33177161

RESUMO

Relative control of HIV-1 infection has been linked to genetic and immune host factors. In this study, we analyzed 96 plasma proteome arrays from chronic untreated HIV-1-infected individuals using the classificatory random forest approach to discriminate between uncontrolled disease (plasma viral load [pVL] >50,000 RNA copies/ml; CD4 counts 283 cells/mm3, n = 47) and relatively controlled disease (pVL <10,000 RNA copies/ml; CD4 counts 657 cells/mm3, n = 49). Our analysis highlighted the TNF molecule's relevance, in particular, TL1A (TNFSF15) and its cognate DR3 (TNFSRF25), both of which increased in the relative virus control phenotype. DR3 levels (in plasma and PBMCs) were validated in unrelated cohorts (including long-term nonprogressors), thus confirming their independence from CD4 counts and pVL. Further analysis in combined antiretroviral treatment (cART)-treated individuals with a wide range of CD4 counts (137-1835 cells/mm3) indicated that neither TL1A nor DR3 levels reflected recovery of CD4 counts with cART. Interestingly, in cART-treated individuals, plasma TL1A levels correlated with regulatory T cell frequencies, whereas soluble DR3 was strongly associated with the abundance of effector HLA-DR+CD8+ T cells. A positive correlation was also observed between plasma DR3 levels and the HIV-1-specific T cell responses. In vitro, costimulation of PBMC with DR3-specific mAb increased the magnitude of HIV-1-specific responses. Finally, in splenocytes of DNA.HTI-vaccinated mice, costimulation of HTI peptides and a DR3 agonist (4C12) intensified the magnitude of T cell responses by 27%. These data describe the role of the TL1A-DR3 axis in the natural control of HIV-1 infection and point to the use of DR3 agonists in HIV-1 vaccine regimens.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Anticorpos Monoclonais Murinos/farmacologia , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/metabolismo , Feminino , Infecções por HIV/sangue , HIV-1/metabolismo , Humanos , Masculino , Camundongos , Membro 25 de Receptores de Fatores de Necrose Tumoral/sangue , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/sangue
3.
J Immunol ; 205(9): 2414-2422, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32958689

RESUMO

Lung fibrosis and tissue remodeling are features of chronic diseases such as severe asthma, idiopathic pulmonary fibrosis, and systemic sclerosis. However, fibrosis-targeted therapies are currently limited. We demonstrate in mouse models of allergen- and bleomycin-driven airway inflammation that neutralization of the TNF family cytokine TL1A through Ab blocking or genetic deletion of its receptor DR3 restricted increases in peribronchial smooth muscle mass and accumulation of lung collagen, primary features of remodeling. TL1A was found as a soluble molecule in the airways and expressed on the surface of alveolar macrophages, dendritic cells, innate lymphoid type 2 cells, and subpopulations of lung structural cells. DR3 was found on CD4 T cells, innate lymphoid type 2 cells, macrophages, fibroblasts, and some epithelial cells. Suggesting in part a direct activity on lung structural cells, administration of recombinant TL1A into the naive mouse airways drove remodeling in the absence of other inflammatory stimuli, innate lymphoid cells, and adaptive immunity. Correspondingly, human lung fibroblasts and bronchial epithelial cells were found to express DR3 and responded to TL1A by proliferating and/or producing fibrotic molecules such as collagen and periostin. Reagents that disrupt the interaction of TL1A with DR3 then have the potential to prevent deregulated tissue cell activity in lung diseases that involve fibrosis and remodeling.


Assuntos
Remodelação das Vias Aéreas/imunologia , Fibrose Pulmonar Idiopática/imunologia , Pulmão/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Imunidade Adaptativa/imunologia , Animais , Asma/imunologia , Bleomicina/imunologia , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Feminino , Humanos , Imunidade Inata/imunologia , Inflamação/imunologia , Linfócitos/imunologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos C57BL
4.
Dev Comp Immunol ; 108: 103685, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32213302

RESUMO

Tumor necrosis factor superfamily (TNFSF)15 is a member of TNFSF which shares a high homology with other TNFSFs, especially lymphotoxin (LT)-α in teleost. In this study, we have cloned a putative TNFSF15 gene in rock bream which was highly homologous with other fish TNFSF15 and performed bioinformatic analysis to confirm the membership. The RB-TNFSF15 cDNA consists of 3192 bp (193 bp of 5'-untranslated region (UTR), 732 bp of ORF, and 2267 bp of 3'-UTR) and encodes a polypeptide of 243 amino acids containing a predicted TNF superfamily signature with 43-61% identities with fish TNFSF15. The predicted 3D structure was similar to human TNFSF15 with ß barrel structure containing 10 ß strands and 1 α helix while human LT-α and ß contain 10 ß strands and 2 α helices. Consequently, the synteny and phylogenetic analysis of fish TNFSF15 genes and structural similarity of the predicted protein to mammalian TNFSF15 implicate that they can be identified as TNFSF15. In healthy rock bream, RB-TNFSF15 gene expression level was the highest in fin and the lowest in blood. In vitro, TNFSF15 gene expression was up-regulated by lipopolysaccharide, polyinosinic:polycytidylic acid (poly I:C) and rock bream iridovirus (RBIV) in head kidney, while up-regulated by poly I:C and RBIV at later time in spleen. In vivo, RB-TNFSF15 gene expression was up-regulated in head kidney, liver and blood after vaccination with a formalin inactivated RBIV. After challenging with RBIV, RB-TNFSF15 gene expression was up-regulated in unvaccinated group at day 3 post-infection in head kidney. In gill, it was significantly up-regulated in vaccinated group at day 1 post-challenge and all groups at day 7, indicating that RB-TNFSF may play a key role in mucosal immunity during viral infection. Since the regulation mechanism of TNFSF15 gene expression in fish has not yet been elucidated, the present study will help to understand the roles of TNFSF15 in fish immune system.


Assuntos
Doenças dos Peixes/imunologia , Proteínas de Peixes/metabolismo , Peixes/imunologia , Iridovirus/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Clonagem Molecular , Doenças dos Peixes/virologia , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Peixes/genética , Peixes/virologia , Regulação da Expressão Gênica/imunologia , Iridovirus/patogenicidade , Filogenia , Poli I-C/imunologia , Alinhamento de Sequência , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Vacinas Virais/administração & dosagem
5.
J Immunol ; 203(11): 2970-2978, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31628153

RESUMO

Mucosal-associated invariant T (MAIT) cells are semi-invariant T cells specifically recognizing riboflavin derivatives that are synthesized by many bacteria and fungi presented by MHC class I-related MR1 molecules. Accumulating evidence, however, indicates that MAIT cell functions are inducible by cytokine stimuli in the absence of TCR ligation, identifying MAIT cells as innate sentinels in inflammatory environments. In this study, we demonstrate that death receptor 3 (DR3), a member of the TNFR superfamily, is ex vivo expressed and predominantly upregulated on the surface of human MAIT cells by innate cytokine stimulation. In turn, the DR3 ligand TNF-like protein 1A (TL1A) licenses innate TNF-α production in the absence of cognate triggers, being sufficient to promote activation of primary endothelial cells in vitro. TL1A further amplifies synthesis of IFN-γ and granzyme B in the presence of otherwise weak innate stimuli and strongly augments polyfunctionality. Mechanistically, TL1A potentiates T-bet expression, early NF-κB, and late p38 MAP kinase phosphorylation, with the latter being indispensable for TNF-α production by MAIT cells. Of note, endogenous TL1A is also rapidly released from PBMC cultures in response to bacterial triggering, thereby equally augmenting Ag-specific MAIT cell effector functions. In summary, to our knowledge, we identify a new inflammatory mechanism in MAIT cells linking the DR3/TL1A axis with amplification of TCR-dependent and -independent effector functions, particularly inducing excessive innate TNF-α production. Given that both TL1A and TNF-α are abundantly present at sites of chronic inflammation, the contribution of MAIT cells in such scenarios needs to be determined.


Assuntos
Células T Invariantes Associadas à Mucosa/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Humanos , Inflamação/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
6.
Front Immunol ; 10: 1624, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31379829

RESUMO

Death receptor 3 (DR3) is a tumor necrosis factor receptor superfamily member (TNFRSF25), which is minimally expressed on resting conventional T cells (though readily inducible upon cell activation), yet highly expressed on resting FoxP3+ regulatory T cells (Treg). We recently demonstrated that activation of DR3 with an agonistic antibody (4C12) leads to selective expansion and activation of Treg in healthy mice and suppression of graft-versus-host disease (GVHD) in recipient mice when donor mice are treated. However, given the long antibody half-life and concomitant safety concerns, along with the lack of a humanized agonistic antibody to DR3, both human and murine fusion proteins incorporating the natural DR3 ligand TL1A (TL1A-Ig) have been developed. Herein, we show that DR3 activation with 4C12 or with TL1A-Ig, with or without the addition of low dose IL-2 to the treatment regimen, led to a significant expansion of murine Treg in spleen, lymph nodes, and peripheral blood. Bioluminescent imaging revealed peak Treg expansion around day 7-8, with return to near baseline after 2-3 weeks. In addition to expansion, all DR3 agonist treatment regimens led to increased activation of Tregs, with significant upregulation of the activation markers ICOS, KLRG-1, PD-1, and CD103, and the proliferation marker Ki-67. The near absence of activated Treg populations in control treated spleens was also detected on tSNE analysis of flow cytometry data. Subtly different patterns of splenic Treg activation by the different DR3 agonists were noted in both tSNE analysis of flow cytometry data and RNA-sequencing analysis. However, upregulation of gene transcripts which play important roles in cell proliferation, trafficking, activation, and effector function were observed regardless of the DR3 agonist treatment regimen used. In the major MHC-mismatch model of hematopoietic cell transplantation, DR3 agonist-mediated expansion and activation of Tregs in donor mice led to a significant improvement in GVHD in recipient mice. These data provide important preclinical information regarding the outcome of DR3 activation with an agonistic antibody or natural ligand and provide insight into the therapeutic use of this approach to reduce GVHD in recipients and improve outcomes of hematopoietic cell transplantation.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Ativação Linfocitária/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Linfócitos T Reguladores/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Feminino , Transplante de Células-Tronco Hematopoéticas/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Doadores de Tecidos
7.
Egypt J Immunol ; 26(1): 43-54, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31332995

RESUMO

Different cytokines play roles in the pathogenesis and tissue damage of Rheumatoid Arthritis (RA) including, Tumor necrosis factor superfamily (TNFSF) and their receptors particularly TNF-like ligand 1A (TL1A), and its decoy receptor DcR3. This study included 150 subjects, of them 50 patients having Rheumatoid Arthritis (RA), 50 patients with Osteoarthritis (OA), and 50 normal controls. Clinical examination was done and data was collected from patient's sheets, routine laboratory investigations included, rheumatoid factor (RF) antibody, anti-cyclic citrullinated peptide (anti-CCP) antibody, erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP). Disease activity score 28 was calculated and used to measure the activity of RA. Serum and synovial fluid (SF) TL1A and DcR3 levels were measured by (ELISA), while IL-17 was measured in supernatant fluid of PBMC culture after stimulation with recombinant human (rh) TL1A. Results showed significantly higher levels of TL1A and its decoy receptor DcR3 in RA patients than the other two groups. It was also found that TL1A is significantly related to the disease activity and enhances IL-17 production after stimulation of PBMC. These results can guide scientists to the future substitutions in the way of treatment of various inflammatory and autoimmune diseases.


Assuntos
Artrite Reumatoide/fisiopatologia , Autoanticorpos/imunologia , Membro 6b de Receptores do Fator de Necrose Tumoral/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Artrite Reumatoide/imunologia , Estudos de Casos e Controles , Células Cultivadas , Humanos , Interleucina-17/imunologia , Leucócitos Mononucleares
8.
Life Sci ; 231: 116536, 2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31176785

RESUMO

AIMS: TL1A was reported to contribute to the susceptibility to ulcerative colitis (UC). However, the molecular mechanisms of TL1A in UC development are poorly understood. We aimed to investigate the role of TL1A in colitis, and reveal the regulatory mechanism of TL1A in chronic colitis development. MAIN METHODS: Wild-type mice and transgenic mice with overexpressing TL1A in lymphocytes were used to construct chronic DSS colitis models. To investigate the molecular mechanism in vitro, CD4+ T cells were sorted from spleens and mesenteric lymph node cells to induce Th9 cells. Biopsy specimens from ulcerative colitis patients were collected for in vivo validation. KEY FINDINGS: The elevated TL1A expression in chronic DSS colitis models exacerbated intestinal inflammation. The differentiation of Th9 cells, IL-9 secretion and production of TGF-ß, IL-4 and PU.1 was significantly enhanced in transgenic mice with TL1A overexpression. In vitro results showed that TL1A enhanced the Th9 cells, IL-9 and PU.1 production, while TL1A antibodies inhibited their production. In human translational studies, patients with ulcerative colitis with elevated TL1A expression also exhibited more serious inflammation with higher levels of Th9 cells, IL-9 and PU.1 expression. SIGNIFICANCE: We presented a possible mechanism of TL1A in UC development that TL1A may promote the differentiation of Th9 cells and enhanced IL-9 secretion by up-regulating the expression of TGF-ß, IL-4 and PU.1, which provided a novel perspective to study the UC pathogenesis, and indicated that targeting of TL1A signal pathway may by a likely strategy for the treatment of chronic colitis.


Assuntos
Colite/imunologia , Interleucina-9/imunologia , Linfócitos T/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Diferenciação Celular/imunologia , Colite/induzido quimicamente , Colite/patologia , Citocinas/imunologia , Citocinas/metabolismo , Glutationa/metabolismo , Interleucina-17/imunologia , Interleucina-1beta/metabolismo , Interleucina-9/metabolismo , Mucosa Intestinal/imunologia , Intestinos/imunologia , Intestinos/patologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Fator de Crescimento Transformador beta/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/biossíntese , Fator de Necrose Tumoral alfa/metabolismo
9.
J Allergy Clin Immunol Pract ; 7(5): 1394-1403, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31076057

RESUMO

Mechanistic studies have improved our understanding of molecular and cellular components involved in asthma and our ability to treat severe patients. An mAb directed against IgE (omalizumab) has become an established add-on therapy for patients with uncontrolled allergic asthma and mAbs specific for IL-5 (reslizumab, mepolizumab), IL-5R (benralizumab), and IL-4R (dupilumab) have been approved as add-on treatments for uncontrolled eosinophilic (type 2) asthma. While these medications have proven highly effective, some patients with severe allergic and/or eosinophilic asthma, as well as most patients with severe non-type-2 disease, have poorly controlled disease. Agents that have recently been evaluated in clinical trials include an antibody directed against thymic stromal lymphopoietin, small molecule antagonists to the chemoattractant receptor-homologous molecule expressed on TH2 cells (CRTH2) and the receptor for stem cell factor on mast cells (KIT), and a DNA enzyme directed at GATA3. Antibodies to IL-33 and its receptor, ST2, are being evaluated in ongoing clinical studies. In addition, a number of antagonists directed against other potential targets are under consideration for future trials, including IL-25, IL-6, TNF-like ligand 1A, CD6, and activated cell adhesion molecule (ALCAM). Clinical data from ongoing and future trials will be important in determining whether these new medications will offer benefits in place of or in addition to existing therapies for asthma.


Assuntos
Antiasmáticos/uso terapêutico , Asma/tratamento farmacológico , Molécula de Adesão de Leucócito Ativado/imunologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Asma/imunologia , Asma/fisiopatologia , Citocinas/antagonistas & inibidores , Citocinas/imunologia , DNA Catalítico/uso terapêutico , Eosinófilos/imunologia , Fator de Transcrição GATA3 , Humanos , Mesilato de Imatinib/uso terapêutico , Ácidos Indolacéticos/uso terapêutico , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Interleucina-6/imunologia , Linfócitos/imunologia , Mastócitos/imunologia , Terapia de Alvo Molecular , Omalizumab/uso terapêutico , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/imunologia , Piridinas/uso terapêutico , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/imunologia , Receptores de Interleucina-17/antagonistas & inibidores , Receptores de Interleucina-17/imunologia , Receptores de Prostaglandina/antagonistas & inibidores , Receptores de Prostaglandina/imunologia , Ribonucleases/uso terapêutico , Células Th2/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
10.
Immunity ; 49(6): 1077-1089.e5, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30552020

RESUMO

Inflammatory bowel disease (IBD) results from a dysregulated interaction between the microbiota and a genetically susceptible host. Genetic studies have linked TNFSF15 polymorphisms and its protein TNF-like ligand 1A (TL1A) with IBD, but the functional role of TL1A is not known. Here, we found that adherent IBD-associated microbiota induced TL1A release from CX3CR1+ mononuclear phagocytes (MNPs). Using cell-specific genetic deletion models, we identified an essential role for CX3CR1+MNP-derived TL1A in driving group 3 innate lymphoid cell (ILC3) production of interleukin-22 and mucosal healing during acute colitis. In contrast to this protective role in acute colitis, TL1A-dependent expression of co-stimulatory molecule OX40L in MHCII+ ILC3s during colitis led to co-stimulation of antigen-specific T cells that was required for chronic T cell colitis. These results identify a role for ILC3s in activating intestinal T cells and reveal a central role for TL1A in promoting ILC3 barrier immunity during colitis.


Assuntos
Colite/imunologia , Imunidade Inata/imunologia , Linfócitos/imunologia , Microbiota/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Adulto , Idoso , Animais , Colite/genética , Colite/metabolismo , Feminino , Humanos , Imunidade Inata/genética , Interleucinas/genética , Interleucinas/imunologia , Interleucinas/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Linfócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microbiota/fisiologia , Pessoa de Meia-Idade , Fagócitos/citologia , Fagócitos/imunologia , Fagócitos/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Adulto Jovem , Interleucina 22
11.
Sci Immunol ; 3(21)2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29500225

RESUMO

Atopic dermatitis is an allergic inflammatory skin disease characterized by the production of the type 2 cytokines in the skin by type 2 innate lymphoid cells (ILC2s) and T helper 2 (TH2) cells, and tissue eosinophilia. Using two distinct mouse models of atopic dermatitis, we show that expression of retinoid-related orphan receptor α (RORα) in skin-resident T regulatory cells (Tregs) is important for restraining allergic skin inflammation. In both models, targeted deletion of RORα in mouse Tregs led to exaggerated eosinophilia driven by interleukin-5 (IL-5) production by ILC2s and TH2 cells. Expression of RORα in skin-resident Tregs suppressed IL-4 expression and enhanced expression of death receptor 3 (DR3), which is the receptor for tumor necrosis factor (TNF) family cytokine, TNF ligand-related molecule 1 (TL1A), which promotes Treg functions. DR3 is expressed on both ILC2s and skin-resident Tregs Upon deletion of RORα in skin-resident Tregs, we found that Tregs were no longer able to sequester TL1A, resulting in enhanced ILC2 activation. We also documented higher expression of RORα in skin-resident Tregs than in peripheral blood circulating Tregs in humans, suggesting that RORα and the TL1A-DR3 circuit could be therapeutically targeted in atopic dermatitis.


Assuntos
Dermatite Atópica/imunologia , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Linfócitos T Reguladores/imunologia , Animais , Feminino , Humanos , Imunidade Inata , Camundongos Transgênicos , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Pele/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
12.
J Immunol ; 200(4): 1360-1369, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29335258

RESUMO

TNF superfamily cytokines play major roles in the regulation of adaptive and innate immunity. The TNF superfamily cytokine TL1A (TNFSF15), through its cognate receptor DR3 (TNFRSF25), promotes T cell immunity to pathogens and directly costimulates group 2 and 3 innate lymphoid cells. Polymorphisms in the TNFSF15 gene are associated with the risk for various human diseases, including inflammatory bowel disease. Like other cytokines in the TNF superfamily, TL1A is synthesized as a type II transmembrane protein and cleaved from the plasma membrane by metalloproteinases. Membrane cleavage has been shown to alter or abrogate certain activities of other TNF family cytokines; however, the functional capabilities of membrane-bound and soluble forms TL1A are not known. Constitutive expression of TL1A in transgenic mice results in expansion of activated T cells and promotes intestinal hyperplasia and inflammation through stimulation of group 2 innate lymphoid cells. Through the generation of membrane-restricted TL1A-transgenic mice, we demonstrate that membrane TL1A promotes expression of inflammatory cytokines in the lung, dependent upon DR3 expression on T cells. Soluble TL1A alone was unable to produce this phenotype but was still able to induce intestinal type 2 inflammation independently of T cells. These data suggest differential roles for membrane and soluble TL1A on adaptive and innate immune cells and have implications for the consequences of blocking these two forms of TL1A.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Ativação Linfocitária/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Linfócitos T/imunologia
13.
Pathol Res Pract ; 214(2): 217-227, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29254800

RESUMO

Tumor necrosis factor like cytokine 1A (TL1A) is a member of the TNF superfamily. Accumulating evidence demonstrated the importance of TL1A in the pathogenesis of inflammatory bowel disease (IBD) and suggested a potential role of TL1A blocking in IBD therapy. Here we aimed to explore whether the anti-TL1A antibody could ameliorate intestinal inflammation and fibrosis in IBD. A T cell transfer model of chronic colitis was induced by intraperitoneal injection of CD4+CD45RBhigh naive T cells isolated from either C57BL/6 wild type (WT) mice or LCK-CD2-Tl1a-GFP transgenic (L-Tg) mice into recombinase activating gene-1-deficient (RAG-/-) mice. The colitis model mice were treated prophylactically or therapeutically with anti-Tl1a antibody or IgG isotype control. Haematoxylin and eosin staining (H&E staining), Masson's trichrome staining (MT staining) and sirius red staining were used to detect histopathological changes in colonic tissue; immunohistochemical staining was used to detect the expressions of collagen I, collagen III, TIMP1, vimentin, α-SMA and TGF-ß1/Smad3. Results showed that anti-Tl1a antibody could reduce intestinal inflammation and fibrosis by inhibiting the activation of intestinal fibroblasts and reducing the collagen synthesis in the T cell transfer model of chronic colitis. The mechanism may be related to the inhibition of TGF-1/Smad3 signaling pathway.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Colite/metabolismo , Inflamação/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Animais , Doença Crônica , Modelos Animais de Doenças , Fibrose/metabolismo , Homeostase/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo
14.
MAbs ; 10(1): 95-103, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29135326

RESUMO

TL1A, a tumor necrosis factor-like cytokine, is a ligand for the death domain receptor DR3. TL1A, upon binding to DR3, can stimulate lymphocytes and trigger secretion of proinflammatory cytokines. Therefore, blockade of TL1A/DR3 interaction may be a potential therapeutic strategy for autoimmune and inflammatory diseases. Recently, the anti-TL1A monoclonal antibody 1 (mAb1) with a strong potency in blocking the TL1A/DR3 interaction was identified. Here, we report on the use of hydrogen/deuterium exchange mass spectrometry (HDX-MS) to obtain molecular-level details of mAb1's binding epitope on TL1A. HDX coupled with electron-transfer dissociation MS provided residue-level epitope information. The HDX dataset, in combination with solvent accessible surface area (SASA) analysis and computational modeling, revealed a discontinuous epitope within the predicted interaction interface of TL1A and DR3. The epitope regions span a distance within the approximate size of the variable domains of mAb1's heavy and light chains, indicating it uses a unique mechanism of action to block the TL1A/DR3 interaction.


Assuntos
Anticorpos Monoclonais/metabolismo , Medição da Troca de Deutério/métodos , Mapeamento de Epitopos/métodos , Epitopos/imunologia , Espectrometria de Massas/métodos , Simulação de Acoplamento Molecular , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Sítios de Ligação de Anticorpos , Células CHO , Cricetulus , Humanos , Cinética , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/química , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
15.
Biol Blood Marrow Transplant ; 23(5): 757-766, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28219835

RESUMO

Regulatory T cells (Tregs) are critical for self-tolerance. Although adoptive transfer of expanded Tregs limits graft-versus-host disease (GVHD) after hematopoietic stem cell transplantation (HSCT), ex vivo generation of large numbers of functional Tregs remains difficult. Here, we demonstrate that in vivo targeting of the TNF superfamily receptor TNFRSF25 using the TL1A-Ig fusion protein, along with IL-2, resulted in transient but massive Treg expansion in donor mice, which peaked within days and was nontoxic. Tregs increased in multiple compartments, including blood, lymph nodes, spleen, and colon (GVHD target tissue). Tregs did not expand in bone marrow, a critical site for graft-versus-malignancy responses. Adoptive transfer of in vivo-expanded Tregs in the setting of MHC-mismatched or MHC-matched allogeneic HSCT significantly ameliorated GVHD. Critically, transplantation of Treg-expanded donor cells facilitated transplant tolerance without GVHD, with complete sparing of graft-versus-malignancy. This approach may prove valuable as a therapeutic strategy promoting transplantation tolerance.


Assuntos
Transferência Adotiva/métodos , Doença Enxerto-Hospedeiro/prevenção & controle , Efeito Enxerto vs Leucemia , Transplante de Células-Tronco Hematopoéticas/métodos , Linfócitos T Reguladores/transplante , Animais , Proliferação de Células/efeitos dos fármacos , Feminino , Doença Enxerto-Hospedeiro/tratamento farmacológico , Imunoglobulinas/farmacologia , Interleucina-2/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Tolerância a Antígenos Próprios , Linfócitos T Reguladores/citologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
16.
Oncotarget ; 7(43): 69436-69449, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27589684

RESUMO

Vascular endothelial cell growth factor (VEGF) plays a pivotal role in promoting neovascularization. VEGF gene expression in vascular endothelial cells in normal tissues is maintained at low levels but becomes highly up-regulated in a variety of disease settings including cancers. Tumor necrosis factor superfamily 15 (TNFSF15; VEGI; TL1A) is an anti-angiogenic cytokine prominently produced by endothelial cells in a normal vasculature. We report here that VEGF production in mouse endothelial cell line bEnd.3 can be inhibited by TNFSF15 via microRNA-29b (miR-29b) that targets the 3'-UTR of VEGF transcript. Blocking TNFSF15 activity by using either siRNA against the TNFSF15 receptor known as death domain-containing receptor-3 (DR3; TNFRSF25), or a neutralizing antibody 4-3H against TNFSF15, led to inhibition of miR-29b expression and reinvigoration of VEGF production. In addition, we found that TNFSF15 activated the JNK signaling pathway as well as the transcription factor GATA3, resulting in enhanced miR-29b production. Treatment of the cells either with SP600125, an inhibitor of JNK, or with JNK siRNA, led to eradication of TNFSF15-induced GATA3 expression. Moreover, GATA3 siRNA suppressed TNFSF15-induced miR-29b expression. These findings suggest that VEGF gene expression can be suppressed by TNFSF15-stimulated activation of the JNK-GATA3 signaling pathway which gives rise to up-regulation of miR-29b.


Assuntos
Células Endoteliais/efeitos dos fármacos , Fator de Transcrição GATA3/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , MicroRNAs/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Antracenos/farmacologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Linhagem Celular , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Fator de Transcrição GATA3/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos Endogâmicos C57BL , Proteína Quinase 8 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Interferência de RNA , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Mol Med Rep ; 13(4): 3647-52, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26956410

RESUMO

Decoy receptor 3 (DcR3) competitively binds to three ligands, Fas ligand, lymphotoxin­related inducible ligand that competes for glycoprotein D binding to herpesvirus entry mediator on T cells and tumor necrosis factor­like ligand 1A (TL1A), to prevent their effects. Recent studies have suggested that DcR3 directly affects cells as a ligand. Using a microarray assay, our group newly identified interleukin (IL)­12B, which encodes the p40 subunit common to IL­12 and IL­23, as one of the genes for which expression in fibroblast­like synoviocytes from patients with rheumatoid arthritis (RA­FLS) is induced by DcR3. The present study demonstrated that IL­12B mRNA expression was upregulated by DcR3­Fc in RA­FLS in a dose­dependent manner, but not in OA­FLS. IL­12B p40 protein in RA­FLS was increased when stimulated with DcR3­Fc. Pre­treatment with anti­TL1A antibody suppressed the upregulation of IL­12B mRNA in RA­FLS stimulated with DcR3­Fc. DcR3 mRNA expression in RA­FLS was induced by IL­23, but not by IL­12. These results indicated that DcR3 may increase IL­12 or IL­23 by inducing IL­12B p40 expression via membrane­bound TL1A on RA­FLS and that IL­23 reciprocally induces DcR3 expression in RA­FLS. DcR3 and IL­23 may interact in a feedback loop that aggravates local inflammation in patients with RA.


Assuntos
Subunidade p40 da Interleucina-12/metabolismo , Membro 6b de Receptores do Fator de Necrose Tumoral/metabolismo , Idoso , Anticorpos/farmacologia , Artrite Reumatoide/patologia , Western Blotting , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Subunidade p40 da Interleucina-12/genética , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Membro 6b de Receptores do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/farmacologia , Membrana Sinovial/citologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Regulação para Cima/efeitos dos fármacos
18.
Inflamm Bowel Dis ; 21(8): 1942-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26389819

RESUMO

On March 18 and 19, 2015, the Institute for Biomedical Sciences at Georgia State University hosted the Second Shanthi V. Sitaraman Intestinal Pathobiology Symposium in memory of Dr. Shanthi V. Sitaraman, an outstanding clinician and scientist in gastroenterology. The recent advances in basic and translational science related to gastroenterology, which makes the timely exchange of ideas critical; the need to recruit mentor and young MD, MD/PhD, and PhD scientists in the field; and the overwhelming success of the First Shanthi V. Sitaraman Intestinal Pathobiology Symposium (2012) in achieving similar goals motivated the project of a second edition of this symposium. Its overall aim was to provide scientific programming at the forefront of research in fields related to the gastrointestinal tract in health and disease. The symposium brought together investigators interested in basic and clinical aspects of gastrointestinal pathobiology in a venue that facilitated meaningful exchanges. This proceeding outlines the 2 days of the symposium and provides insights into recent advances in the field of digestive diseases, as reflected in the speakers' presentations.


Assuntos
Enterócitos/metabolismo , Gastroenterologia/tendências , Gastroenteropatias/terapia , Trato Gastrointestinal/fisiopatologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Microbioma Gastrointestinal , Georgia , Humanos , Imunidade Inata
19.
Eur J Immunol ; 45(8): 2335-42, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26046454

RESUMO

Death receptor 3 (DR3, TNFRSF25) is expressed by activated lymphocytes and signaling by its ligand, TL1A, enhances cytokine expression and proliferation. Recent studies show that DR3 is also present on murine type 2 innate lymphoid cells (ILC2s). Here, we show that DR3 is expressed by IL-22-producing human group 3 innate lymphoid cells (ILC3s). Stimulation of ILC3s with exogenous TL1A alone had no impact on cytokine production or proliferation. Addition of TL1A to IL-1ß + IL-23 significantly enhanced the amount IL-22 produced by ILC3s as well as the percentage IL-22- and IL-8-producing cells. Addition of TL1A to IL-1ß + IL-23 also augmented ILC3 proliferation. Mechanistically, this occurred through the upregulation of CD25 and responsiveness to IL-2 stimulation. The combination of TL1A, IL-1ß+ IL-23, and IL-2 expanded ILC3s while IL-1ß+ IL-23 did not increase proliferation above controls. After 2 weeks of expansion, ILC3s maintained their phenotype, transcription factor expression, and function (IL-22 production). These findings identify DR3 as a costimulatory molecule on ILC3s that could be exploited for ex vivo expansion and clinical use.


Assuntos
Proliferação de Células/efeitos dos fármacos , Imunidade Inata , Interleucina-2/imunologia , Interleucinas/imunologia , Linfócitos/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/farmacologia , Regulação para Cima/efeitos dos fármacos , Células Cultivadas , Humanos , Interleucina-1beta/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade p19 da Interleucina-23/imunologia , Interleucina-8/imunologia , Linfócitos/classificação , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Regulação para Cima/imunologia , Interleucina 22
20.
Rheumatol Int ; 35(8): 1435-7, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25929716

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disease. Cytokine-mediated immunity plays an important role in the pathogenesis of SLE. TNF-like ligand 1A (TL1A) belongs to the TNF superfamily of cytokines and has been found to perform significantly in autoimmune diseases, such as rheumatoid arthritis and inflammatory bowel disease. To date, no study has discussed the expression levels of TL1A in SLE. We found that plasma levels of TL1A were significantly higher in newly diagnosed SLE patients compared with controls. Correlation analysis showed that plasma levels of TL1A were positively associated with SLE disease activity index. These data indicated that TL1A may play a role in SLE and may reflect the disease activity for SLE.


Assuntos
Lúpus Eritematoso Sistêmico/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Adolescente , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...