Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
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 ; 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
4.
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
5.
Nat Commun ; 10(1): 3371, 2019 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-31358760

RESUMO

TNF-like ligand 1 A (TL1A) and death receptor 3 (DR3) are a ligand-receptor pair involved in the pathogenesis of inflammatory bowel disease. Group 3 innate lymphoid cells (ILC3s) regulate intestinal immunity and highly express DR3. Here, we report that activation of DR3 signaling by an agonistic anti-DR3 antibody increases GM-CSF production from ILC3s through the p38 MAPK pathway. GM-CSF causes accumulation of eosinophils, neutrophils and CD11b+CD11c+ myeloid cells, resulting in loss of ILC3s from the intestine in an IL-23-dependent manner and exacerbating colitis. Blockade of GM-CSF or IL-23 reverses anti-DR3 antibody-driven ILC3 loss, whereas overexpression of IL-23 induces loss of ILC3s in the absence of GM-CSF. Neutralization of TL1A by soluble DR3 ameliorates both DSS and anti-CD40 antibody-induced colitis. Moreover, ILC3s are required for the deleterious effect of anti-DR3 antibodies on innate colitis. These findings clarify the process and consequences of DR3 signaling-induced intestinal inflammation through regulation of ILC3s.


Assuntos
Colite/imunologia , Doenças Inflamatórias Intestinais/imunologia , Linfócitos/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais/imunologia , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Células Cultivadas , Colite/genética , Colite/metabolismo , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/metabolismo , Interleucina-23/farmacologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/genética
6.
Front Immunol ; 9: 362, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29545797

RESUMO

Death receptor 3 (DR3), a member of the tumor necrosis factor receptor (TNFR) superfamily, has been implicated in regulating T-helper type-1 (TH1), type-2 (TH2), and type-17 (TH17) responses as well as regulatory T cell (Treg) and innate lymphoid cell (ILC) functions during immune-mediated diseases. However, the role of DR3 in controlling lymphocyte functions in inflammatory bowel disease (IBD) is not fully understood. Recent studies have shown that activation of DR3 signaling modulates Treg expansion suggesting that stimulation of DR3 represents a potential therapeutic target in human inflammatory diseases, including Crohn's disease (CD). In this study, we tested a specific DR3 agonistic antibody (4C12) in SAMP1/YitFc (SAMP) mice with CD-like ileitis. Interestingly, treatment with 4C12 prior to disease manifestation markedly worsened the severity of ileitis in SAMP mice despite an increase in FoxP3+ lymphocytes in mesenteric lymph node (MLN) and small-intestinal lamina propria (LP) cells. Disease exacerbation was dominated by overproduction of both TH1 and TH2 cytokines and associated with expansion of dysfunctional CD25-FoxP3+ and ILC group 1 (ILC1) cells. These effects were accompanied by a reduction in CD25+FoxP3+ and ILC group 3 (ILC3) cells. By comparison, genetic deletion of DR3 effectively reversed the inflammatory phenotype in SAMP mice by promoting the expansion of CD25+FoxP3+ over CD25-FoxP3+ cells and the production of IL-10 protein. Collectively, our data demonstrate that DR3 signaling modulates a multicellular network, encompassing Tregs, T effectors, and ILCs, governing disease development and progression in SAMP mice with CD-like ileitis. Manipulating DR3 signaling toward the restoration of the balance between protective and inflammatory lymphocytes may represent a novel and targeted therapeutic modality for patients with CD.


Assuntos
Doença de Crohn/imunologia , Ileíte/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/agonistas , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Ileíte/genética , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos AKR , Camundongos Knockout , Camundongos Transgênicos , Proteínas Nucleares/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais
7.
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
8.
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
9.
J Immunol ; 199(7): 2279-2290, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28827283

RESUMO

Sarcoidosis is a granulomatous disease that primarily affects the lungs and is characterized by an accumulation of CD4+ T cells in the bronchoalveolar lavage (BAL). Previous work has indicated that HLA-DRB1*03:01+ (DR3+) patients diagnosed with the acute form of the disease, Löfgren's syndrome (LS), have an accumulation of CD4+ T cells bearing TCRs using TRAV12-1 (formerly AV2S3). However, the importance of these α-chains in disease pathogenesis and the paired TCRß-chain remains unknown. This study aimed to identify expanded αßTCR pairs expressed on CD4+ T cells derived from the BAL of DR3+ LS patients. Using a deep-sequencing approach, we determined TCRα- and TCRß-chain usage, as well as αßTCR pairs expressed on BAL CD4+ T cells from LS patients. TRAV12-1 and TRBV2 (formerly BV22) were the most expanded V region gene segments in DR3+ LS patients relative to control subjects, and TRAV12-1 and TRBV2 CDR3 motifs were shared among multiple DR3+ LS patients. When assessing αßTCR pairing, TRAV12-1 preferentially paired with TRBV2, and these TRAV12-1/TRBV2 TCRs displayed CDR3 homology. These findings suggest that public CD4+ TCR repertoires exist among LS patients and that these T cells are recognizing the putative sarcoidosis-associated Ag(s) in the context of DR3.


Assuntos
Líquido da Lavagem Broncoalveolar/citologia , Pulmão/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Sarcoidose Pulmonar/imunologia , Doença Aguda , Adulto , Idoso , Líquido da Lavagem Broncoalveolar/imunologia , Linfócitos T CD4-Positivos/imunologia , Feminino , Cadeias HLA-DRB1/genética , Cadeias HLA-DRB1/imunologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia
10.
PLoS Pathog ; 13(8): e1006566, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28817719

RESUMO

Th1 cells can be activated by TCR-independent stimuli, but the importance of this pathway in vivo and the precise mechanisms involved require further investigation. Here, we used a simple model of non-cognate Th1 cell stimulation in Salmonella-infected mice to examine these issues. CD4 Th1 cell expression of both IL-18R and DR3 was required for optimal IFN-γ induction in response to non-cognate stimulation, while IL-15R expression was dispensable. Interestingly, effector Th1 cells generated by immunization rather than live infection had lower non-cognate activity despite comparable IL-18R and DR3 expression. Mice lacking T cell intrinsic expression of MyD88, an important adapter molecule in non-cognate T cell stimulation, exhibited higher bacterial burdens upon infection with Salmonella, Chlamydia or Brucella, suggesting that non-cognate Th1 stimulation is a critical element of efficient bacterial clearance. Thus, IL-18R and DR3 are critical players in non-cognate stimulation of Th1 cells and this response plays an important role in protection against intracellular bacteria.


Assuntos
Infecções Bacterianas/imunologia , Ativação Linfocitária/imunologia , Receptores de Interleucina-18/biossíntese , Membro 25 de Receptores de Fatores de Necrose Tumoral/biossíntese , Células Th1/imunologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interleucina-18/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Receptores de Interleucina-18/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Células Th1/metabolismo
11.
Immunology ; 150(4): 418-431, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27859243

RESUMO

Significant barriers to transplantation exist for individuals who are pre-sensitized to donor antigen and have high titres of donor-reactive antibody. We report the effect of autologous bone marrow transplantation (BMTx) after myeloablation in pre-sensitized mice along with the use of monoclonal antibodies (mAbs) to tumour necrosis factor-receptor super family 25 (TNFRSF25), expressed on regulatory T (Treg) cells. C57BL/6 mice, which had been sensitized earlier with BALB/c skin allografts, received secondary BALB/c grafts after the primary grafts had been rejected. Subsequently, recipient mice underwent myeloablation with cyclophosphamide and busulphan and were injected with T-cell-depleted bone marrow from CD45.1 congenic donors (BMTx). Recipient mice underwent immunosuppressive treatment with rapamycin. A subgroup of mice was also treated with mAbs to TNFRSF25. Control mice were pre-sensitized mice that received cyclophosphamide and busulphan followed by rapamycin. BMTx-treated mice had significantly prolonged skin graft survival versus control mice. These mice also showed attenuated donor-specific mixed lymphocyte co-culture responses relative to controls, increased splenic Treg cells and markedly diminished serum anti-donor IgG. Infusion of anti-TNFRSF25 mAbs further augmented graft survival and increased graft-infiltrating Treg cells. These mAbs also expanded murine and human Treg cells in vitro with the capacity to attenuate mixed lymphocyte co-cultures using fresh peripheral blood mononuclear cells. Overall, this study delineates the roles of autologous BMTx and anti-TNFRSF25 mAbs in expanding Treg cells and attenuating alloimmune responses in pre-sensitized mice.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Transplante de Medula Óssea , Rejeição de Enxerto/prevenção & controle , Imunoterapia/métodos , Linfócitos T Reguladores/imunologia , Aloenxertos/imunologia , Animais , Células Cultivadas , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto , Humanos , Tolerância Imunológica , Isoanticorpos/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Transplante Homólogo
12.
Immunopharmacol Immunotoxicol ; 38(1): 29-38, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26810853

RESUMO

A number of anti-tumor necrosis factor alpha (TNF-α) biologics have been developed in recent years, such as adalimumab, etanercept, and infliximab for the treatment of chronic inflammatory disorders like rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and psoriasis and several other novel drugs that target TNF-α signaling are still being developed. Indeed, blockade of this pathway seems so important amongst immune-targets that TNF-α targeted therapies will continue to have a significant role in the treatment of chronic inflammation. However, up to 40% of RA and IBD patients do not respond to anti-TNF-α treatment and one possible explanation may be the heterogeneity of chronic inflammatory diseases and a dominance of other significant TNF family members. Indeed, polymorphisms in the TNF family member, TL1A gene, is associated with the development of IBD and increased serum concentrations of TL1A has been demonstrated in patients with various chronic inflammatory disorders. Here, we describe the current knowledge of TL1As immunobiology and present results from human disease, animal models, and pre-clinical intervention studies that point toward development of anti-TL1A therapy as a highly promising strategy for treatment of chronic inflammatory disorders.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Reumatoide , Doenças Inflamatórias Intestinais , Psoríase , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Doença Crônica , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Psoríase/tratamento farmacológico , Psoríase/imunologia , Psoríase/patologia , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia
13.
Blood ; 126(4): 546-57, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-26063163

RESUMO

The paucity of regulatory T cells (Tregs) limits clinical translation to control aberrant immune reactions including graft-versus-host disease (GVHD). Recent studies showed that the agonistic antibody to DR3 (αDR3) expanded CD4(+)FoxP3(+) Tregs in vivo. We investigated whether treating donor mice with a single dose of αDR3 could alleviate acute GVHD in a MHC-mismatched bone marrow transplantation model. αDR3 induced selective proliferation of functional Tregs. CD4(+) T cells isolated from αDR3-treated mice contained higher numbers of Tregs and were less proliferative to allogeneic stimuli. In vivo GVHD studies confirmed that Tregs from αDR3-treated donors expanded robustly and higher frequencies of Tregs within donor CD4(+) T cells were maintained, resulting in improved survival. Conventional T cells derived from αDR3-treated donors showed reduced activation and proliferation. Serum levels of proinflammatory cytokines (IFNγ, IL-1ß, and TNFα) and infiltration of donor T cells into GVHD target tissues (gastrointestinal tract and liver) were decreased. T cells from αDR3-treated donors retained graft-vs-tumor (GVT) effects. In conclusion, a single dose of αDR3 alleviates acute GVHD while preserving GVT effects by selectively expanding and maintaining donor Tregs. This novel strategy will facilitate the clinical application of Treg-based therapies.


Assuntos
Anticorpos/administração & dosagem , Linfócitos T CD4-Positivos/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Efeito Enxerto vs Tumor/imunologia , Linfoma de Células B/prevenção & controle , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Linfócitos T Reguladores/imunologia , Animais , Transplante de Medula Óssea , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Doença Enxerto-Hospedeiro/imunologia , Imunoglobulinas/administração & dosagem , Linfoma de Células B/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Doadores de Tecidos , Transplante Homólogo , Células Tumorais Cultivadas
14.
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
15.
Mucosal Immunol ; 8(4): 731-4, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26059005

RESUMO

Memory CD4 T cells are strategically positioned at mucosal surfaces to initiate a robust adaptive immune response. The detection of specific antigen via the T-cell receptor causes these memory T cells to unleash a potent antimicrobial response that includes rousing local innate immune populations for tissue-specific defense. Paradoxically, these same memory T cells can also be stimulated by nonantigen-specific signals that are generated by the activity of local innate immune cells. This versatility of mucosal memory T cells in both the initiation and the sensing of local innate immunity could be a vitally important asset during pathogen defense but alternatively could be responsible for initiating and maintaining chronic inflammation in sensitive mucosal tissues.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Doença de Crohn/imunologia , Imunidade Inata , Pólipos Nasais/imunologia , Receptores de Interleucina-18/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Humanos
16.
J Autoimmun ; 61: 17-28, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26005049

RESUMO

Recently we identified in a wide spectrum of autoimmune diseases frequently occurring proinflammatory autoantibodies directed against progranulin, a direct inhibitor of TNFR1 & 2 and of DR3. In the present study we investigated the mechanisms for the breakdown of self-tolerance against progranulin. Isoelectric focusing identified a second, differentially electrically charged progranulin isoform exclusively present in progranulin-antibody-positive patients. Alkaline phosphatase treatment revealed this additional progranulin isoform to be hyperphosphorylated. Subsequently Ser81, which is located within the epitope region of progranulin-antibodies, was identified as hyperphosphorylated serine residue by site directed mutagenesis of candidate phosphorylation sites. Hyperphosphorylated progranulin was detected exclusively in progranulin-antibody-positive patients during the courses of their diseases. The occurrence of hyperphosphorylated progranulin preceded seroconversions of progranulin-antibodies, indicating adaptive immune response. Utilizing panels of kinase and phosphatase inhibitors, PKCß1 was identified as the relevant kinase and PP1 as the relevant phosphatase for phosphorylation and dephosphorylation of Ser81. In contrast to normal progranulin, hyperphosphorylated progranulin interacted exclusively with inactivated (pThr320) PP1, suggesting inactivated PP1 to cause the detectable occurrence of phosphorylated Ser81 PGRN. Investigation of possible functional alterations of PGRN due to Ser81 phosphorylation revealed, that hyperphosphorylation prevents the interaction and thus direct inhibition of TNFR1, TNFR2 and DR3, representing an additional direct proinflammatory effect. Finally phosphorylation of Ser81 PGRN alters the conversion pattern of PGRN. In conclusion, inactivated PP1 induces hyperphosphorylation of progranulin in a wide spectrum of autoimmune diseases. This hyperphosphorylation prevents direct inhibition of TNFR1, TNFR2 and DR3 by PGRN, alters the conversion of PGRN, and is strongly associated with the occurrence of neutralizing, proinflammatory PGRN-antibodies, indicating immunogenicity of this alternative secondary modification.


Assuntos
Autoanticorpos/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Precursores de Proteínas/imunologia , Serina/imunologia , Animais , Autoanticorpos/genética , Autoanticorpos/metabolismo , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Citometria de Fluxo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Progranulinas , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Proteína Quinase C beta/genética , Proteína Quinase C beta/imunologia , Proteína Quinase C beta/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Serina/genética , Serina/metabolismo
17.
J Immunol ; 194(8): 3567-82, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25786692

RESUMO

The TNF family cytokine TL1A (Tnfsf15) costimulates T cells and type 2 innate lymphocytes (ILC2) through its receptor DR3 (Tnfrsf25). DR3-deficient mice have reduced T cell accumulation at the site of inflammation and reduced ILC2-dependent immune responses in a number of models of autoimmune and allergic diseases. In allergic lung disease models, immunopathology and local Th2 and ILC2 accumulation is reduced in DR3-deficient mice despite normal systemic priming of Th2 responses and generation of T cells secreting IL-13 and IL-4, prompting the question of whether TL1A promotes the development of other T cell subsets that secrete cytokines to drive allergic disease. In this study, we find that TL1A potently promotes generation of murine T cells producing IL-9 (Th9) by signaling through DR3 in a cell-intrinsic manner. TL1A enhances Th9 differentiation through an IL-2 and STAT5-dependent mechanism, unlike the TNF-family member OX40, which promotes Th9 through IL-4 and STAT6. Th9 differentiated in the presence of TL1A are more pathogenic, and endogenous TL1A signaling through DR3 on T cells is required for maximal pathology and IL-9 production in allergic lung inflammation. Taken together, these data identify TL1A-DR3 interactions as a novel pathway that promotes Th9 differentiation and pathogenicity. TL1A may be a potential therapeutic target in diseases dependent on IL-9.


Assuntos
Asma/imunologia , Diferenciação Celular/imunologia , Interleucina-9/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Animais , Asma/genética , Asma/patologia , Diferenciação Celular/genética , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-4/genética , Interleucina-4/imunologia , Interleucina-9/genética , Camundongos , Camundongos Knockout , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T Auxiliares-Indutores/patologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
18.
J Neuroimmunol ; 278: 90-9, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25595257

RESUMO

The study was designed to test DNA Aß42 immunization in mice as alternative approach for possible active immunotherapy in Alzheimer patients. As results, we found polarized Th2 immune responses, efficient Aß42 antibody levels, and disappearance of antigen specific T cells. In-vivo TNFRSF4/25 antibody co-stimulation enhanced Aß42 specific T cell responses with initial Th2 expansion and subsequent development of Aß42 specific CD4+CD25+Foxp3+ cells. It showed that Th2 biased responses due to gene gun immunizations propagate the development of regulatory T cells. In conclusion, full-length DNA Aß42 immunization into skin results in a regulatory response with minimal risk of inflammation and autoimmunity.


Assuntos
Peptídeos beta-Amiloides/genética , Anticorpos/farmacologia , Citocinas/metabolismo , DNA/imunologia , Fragmentos de Peptídeos/genética , Receptores OX40/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos/sangue , Células Cultivadas , DNA/química , Proteínas de Ligação a DNA/metabolismo , Feminino , Fatores de Transcrição Forkhead/genética , Fator de Transcrição GATA3/metabolismo , Humanos , Imunoterapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Fragmentos de Peptídeos/imunologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Linfócitos T Reguladores/citologia , Fatores de Transcrição/metabolismo
19.
Mucosal Immunol ; 8(3): 545-58, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25269704

RESUMO

Mucosal tissues contain large numbers of memory CD4(+) T cells that, through T-cell receptor-dependent interactions with antigen-presenting cells, are believed to have a key role in barrier defense and maintenance of tissue integrity. Here we identify a major subset of memory CD4(+) T cells at barrier surfaces that coexpress interleukin-18 receptor alpha (IL-18Rα) and death receptor-3 (DR3), and display innate lymphocyte functionality. The cytokines IL-15 or the DR3 ligand tumor necrosis factor (TNF)-like cytokine 1A (TL1a) induced memory IL-18Rα(+)DR3(+)CD4(+) T cells to produce interferon-γ, TNF-α, IL-6, IL-5, IL-13, granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL-22 in the presence of IL-12/IL-18. TL1a synergized with IL-15 to enhance this response, while suppressing IL-15-induced IL-10 production. TL1a- and IL-15-mediated cytokine induction required the presence of IL-18, whereas induction of IL-5, IL-13, GM-CSF, and IL-22 was IL-12 independent. IL-18Rα(+)DR3(+)CD4(+) T cells with similar functionality were present in human skin, nasal polyps, and, in particular, the intestine, where in chronic inflammation they localized with IL-18-producing cells in lymphoid aggregates. Collectively, these results suggest that human memory IL-18Rα(+)DR3(+) CD4(+) T cells may contribute to antigen-independent innate responses at barrier surfaces.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Doença de Crohn/imunologia , Imunidade Inata , Pólipos Nasais/imunologia , Receptores de Interleucina-18/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Linfócitos T CD4-Positivos/patologia , Doença de Crohn/genética , Doença de Crohn/patologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunidade nas Mucosas , Memória Imunológica , Interferon gama/genética , Interferon gama/imunologia , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-15/genética , Interleucina-15/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucinas/genética , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Pólipos Nasais/genética , Pólipos Nasais/patologia , Cultura Primária de Células , Receptores de Interleucina-18/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Transdução de Sinais , Pele/citologia , Pele/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Interleucina 22
20.
Transplantation ; 98(12): 1271-8, 2014 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-25340612

RESUMO

BACKGROUND: CD200 overexpression in transgenic recipients, or by tissue allografts, increases skin and cardiac graft survival in mice receiving rapamycin, in association with increased Foxp3(+)Treg graft infiltration. However, less than 50% of skin grafts persist beyond 25 days. We investigated whether immune ablation using busulphan and cyclophosphamide, followed by autologous marrow reconstitution, would permit long-term survival in a greater percentage of recipients and induce tolerance, allowing for withdrawal of immunosuppressive drugs. METHODS: C57BL/6 wild type (BL/6 WT) or CD200(tg) mice received BALB/c skin grafts with rapamycin (1 mg/kg/36 hr) for 7 days. Thereafter, subgroups received busulphan or cyclophospamide for 6 days, followed by BL/6 marrow transplantation (BMTx), whereas controls were maintained on rapamycin. Beginning 7 days after marrow transplantation, mice receiving BMTx also received rapamycin for a further 7, 14, or 21 days (to a maximum of 42 days after transplantation) after which times, all immunosuppressions were withdrawn. Graft survival was monitored throughout, and mixed leukocyte cultures (MLCs) (using peripheral blood leukocytes) were performed for all groups at 60 days after transplantation. Gene expression was performed in grafts harvested at 80 days. RESULTS: Although all WT mice rejected grafts by 16 days, survival in CD200(tg) was approximately 40% at 60 days, with antigen-specific decreased MLC responses to BALB/c. After BMTx, survival in WT mice was greater than 40% at 60 days, and greater than 90% at 60 days in CD200(tg), with corresponding attenuated MLC responses. Long-term surviving grafts were infiltrated by Tregs of both host and donor marrow origins, as defined using CD45 congeneic donors or recipients. Survival was increased by infusion of an antibody directed to TNFRSF25 on Tregs. CONCLUSION: Autologous BMTx improves graft survival and promotes graft tolerance.


Assuntos
Antígenos CD/genética , Transplante de Medula Óssea , Sobrevivência de Enxerto , Tolerância Imunológica , Transplante de Pele , Pele/imunologia , Aloenxertos , Animais , Anticorpos/imunologia , Antígenos CD/imunologia , Separação Celular , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Imunossupressores/química , Imunossupressores/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Membro 25 de Receptores de Fatores de Necrose Tumoral/imunologia , Tolerância ao Transplante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA