Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Clin Invest ; 132(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34871182

RESUMO

BACKGROUNDSevere coronavirus disease 2019 (COVID-19) is associated with a dysregulated immune response, which can result in cytokine-release syndrome and acute respiratory distress syndrome (ARDS). Patients with COVID-19-associated ARDS have elevated free serum levels of the cytokine lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells (LIGHT; also known as TNFSF14). Such patients may benefit from LIGHT-neutralization therapy.METHODSThis randomized, double-blind, multicenter, proof-of-concept trial enrolled adults hospitalized with COVID-19-associated pneumonia and mild to moderate ARDS. Patients received standard of care plus a single dose of a human LIGHT-neutralizing antibody (CERC-002) or placebo. The primary endpoint was the proportion of patients receiving CERC-002 who remained alive and free of respiratory failure through day 28. Safety was assessed via adverse event monitoring.RESULTSFor most of the 83 enrolled patients, standard of care included systemic corticosteroids (88.0%) or remdesivir (57.8%). A higher proportion of patients remained alive and free of respiratory failure through day 28 after receiving CERC-002 (83.9%) versus placebo (64.5%; P = 0.044), including in patients 60 years of age or older (76.5% vs. 47.1%, respectively; P = 0.042). Mortality rates were 7.7% (CERC-002) and 14.3% (placebo) on day 28 and 10.8% and 22.5%, respectively, on day 60. Treatment-emergent adverse events were less frequent with CERC-002 than placebo.CONCLUSIONFor patients with COVID-19-associated ARDS, adding CERC-002 to standard-of-care treatment reduces LIGHT levels and might reduce the risk of respiratory failure and death.TRIAL REGISTRATIONClinicalTrials.gov NCT04412057.FUNDINGAvalo Therapeutics.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Tratamento Farmacológico da COVID-19 , Síndrome da Liberação de Citocina/tratamento farmacológico , Síndrome do Desconforto Respiratório/tratamento farmacológico , SARS-CoV-2/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/análogos & derivados , Corticosteroides/administração & dosagem , Adulto , Alanina/administração & dosagem , Alanina/análogos & derivados , COVID-19/sangue , COVID-19/mortalidade , Síndrome da Liberação de Citocina/sangue , Síndrome da Liberação de Citocina/mortalidade , Intervalo Livre de Doença , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Síndrome do Desconforto Respiratório/sangue , Síndrome do Desconforto Respiratório/mortalidade , Taxa de Sobrevida , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/sangue
2.
J Exp Med ; 215(2): 415-422, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29339444

RESUMO

Dermatitis is often associated with an allergic reaction characterized by excessive type 2 responses leading to epidermal acanthosis, hyperkeratosis, and dermal inflammation. Although factors like IL-4, IL-13, and thymic stromal lymphopoietin (TSLP) are thought to be instrumental for the development of this type of skin disorder, other cytokines may be critical. Here, we show that the tumor necrosis factor (TNF) superfamily protein LIGHT (homologous to lymphotoxin, exhibits inducible expression, and competes with HSV glycoprotein D for binding to HVEM, a receptor expressed on T lymphocytes) is required for experimental atopic dermatitis, and LIGHT directly controls keratinocyte hyperplasia, and production of periostin, a matricellular protein that contributes to the clinical features of atopic dermatitis as well as other skin diseases such as scleroderma. Mice with a conditional deletion of the LIGHT receptor HVEM (herpesvirus entry mediator) in keratinocytes phenocopied LIGHT-deficient mice in exhibiting reduced epidermal thickening and dermal collagen deposition in a model of atopic dermatitis driven by house dust mite allergen. LIGHT signaling through HVEM in human epidermal keratinocytes directly induced proliferation and periostin expression, and both keratinocyte-specific deletion of HVEM or antibody blocking of LIGHT-HVEM interactions after disease onset prevented expression of periostin and limited atopic dermatitis symptoms. Developing reagents that neutralize LIGHT-HVEM signaling might be useful for therapeutic intervention in skin diseases where periostin is a central feature.


Assuntos
Dermatite Atópica/metabolismo , Queratinócitos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Antígenos de Dermatophagoides/efeitos adversos , Moléculas de Adesão Celular/metabolismo , Proliferação de Células , Dermatite Atópica/etiologia , Dermatite Atópica/imunologia , Modelos Animais de Doenças , Células HEK293 , Humanos , Queratinócitos/imunologia , Queratinócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Interferente Pequeno/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Membro 14 de Receptores do Fator de Necrose Tumoral/deficiência , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/deficiência , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
3.
Nat Rev Rheumatol ; 13(4): 217-233, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28275260

RESUMO

TNF blockers are highly efficacious at dampening inflammation and reducing symptoms in rheumatic diseases such as rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis, and also in nonrheumatic syndromes such as inflammatory bowel disease. As TNF belongs to a superfamily of 19 structurally related proteins that have both proinflammatory and anti-inflammatory activity, reagents that disrupt the interaction between proinflammatory TNF family cytokines and their receptors, or agonize the anti-inflammatory receptors, are being considered for the treatment of rheumatic diseases. Biologic agents that block B cell activating factor (BAFF) and receptor activator of nuclear factor-κB ligand (RANKL) have been approved for the treatment of systemic lupus erythematosus and osteoporosis, respectively. In this Review, we focus on additional members of the TNF superfamily that could be relevant for the pathogenesis of rheumatic disease, including those that can strongly promote activity of immune cells or increase activity of tissue cells, as well as those that promote death pathways and might limit inflammation. We examine preclinical mouse and human data linking these molecules to the control of damage in the joints, muscle, bone or other tissues, and discuss their potential as targets for future therapy of rheumatic diseases.


Assuntos
Terapia de Alvo Molecular , Doenças Reumáticas/tratamento farmacológico , Doenças Reumáticas/imunologia , Inibidores do Fator de Necrose Tumoral , Fatores de Necrose Tumoral/metabolismo , Ligante 4-1BB/antagonistas & inibidores , Ligante 4-1BB/metabolismo , Animais , Ligante CD27/antagonistas & inibidores , Ligante CD27/metabolismo , Ligante de CD40/antagonistas & inibidores , Ligante de CD40/metabolismo , Morte Celular , Citocina TWEAK , Células Dendríticas/imunologia , Proteína Ligante Fas/antagonistas & inibidores , Proteína Ligante Fas/metabolismo , Humanos , Tolerância Imunológica , Ativação Linfocitária , Linfotoxina-alfa/antagonistas & inibidores , Linfotoxina-alfa/metabolismo , Ligante OX40/antagonistas & inibidores , Ligante OX40/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Fatores de Necrose Tumoral/imunologia
5.
Sci Rep ; 5: 10406, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25993659

RESUMO

Host anti-viral innate immunity plays important roles in the defense against HSV-1 infection. In this study, we find an unexpected role for innate LT/LIGHT signaling in promoting HSV-1 replication and virus induced inflammation in immunocompromised mice. Using a model of footpad HSV-1 infection in Rag1(-/-) mice, we observed that blocking LT/LIGHT signaling with LTßR-Ig could significantly delay disease progression and extend the survival of infected mice. LTßR-Ig treatment reduced late proinflammatory cytokine release in the serum and nervous tissue, and inhibited chemokine expression and inflammatory cells infiltration in the dorsal root ganglia (DRG). Intriguingly, LTßR-Ig treatment restricted HSV-1 replication in the DRG but not the footpad. These findings demonstrate a critical role for LT/LIGHT signaling in modulating innate inflammation and promoting HSV-1 replication in the nervous system, and suggest a new target for treatment of virus-induced adverse immune response and control of severe HSV-1 infection.


Assuntos
Herpesvirus Humano 1/fisiologia , Inflamação , Receptor beta de Linfotoxina/metabolismo , Animais , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Gânglios Espinais/virologia , Herpes Simples/metabolismo , Herpes Simples/mortalidade , Herpes Simples/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imunidade Inata , Receptor beta de Linfotoxina/química , Linfotoxina-alfa/antagonistas & inibidores , Linfotoxina-alfa/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Medula Espinal/metabolismo , Medula Espinal/virologia , Taxa de Sobrevida , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Replicação Viral
6.
Am J Transplant ; 13(3): 541-51, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23356438

RESUMO

The exchange of information during interactions of T cells with dendritic cells, B cells or other T cells regulates the course of T, B and DC-cell activation and their differentiation into effector cells. The tumor necrosis factor superfamily member LIGHT (homologous to lymphotoxin, exhibits inducible expression and competes with HSV glycoprotein D for binding to herpesvirus entry mediator, a receptor expressed on T lymphocytes) is transiently expressed upon T cell activation and modulates CD8 T cell-mediated alloreactive responses upon herpes virus entry mediator (HVEM) and lymphotoxin ß receptor (LTßR) engagement. LIGHT-deficient mice, or WT mice treated with LIGHT-targeting decoy receptors HVEM-Ig, LTßR-Ig or sDcR3-Ig, exhibit prolonged graft survival compared to untreated controls, suggesting that LIGHT modulates the course and severity of graft rejection. Therefore, targeting the interaction of LIGHT with HVEM and/or LTßR using recombinant soluble decoy receptors or monoclonal antibodies represent an innovative therapeutic strategy for the prevention and treatment of allograft rejection and for the promotion of donor-specific tolerance.


Assuntos
Anticorpos Monoclonais/farmacologia , Rejeição de Enxerto/imunologia , Doença Enxerto-Hospedeiro/imunologia , Receptor beta de Linfotoxina/antagonistas & inibidores , Transplante de Órgãos , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Animais , Humanos , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Camundongos , Ligação Proteica , Transplante Homólogo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
7.
Nat Med ; 17(5): 596-603, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21499267

RESUMO

Individuals with chronic asthma show a progressive decline in lung function that is thought to be due to structural remodeling of the airways characterized by subepithelial fibrosis and smooth muscle hyperplasia. Here we show that the tumor necrosis factor (TNF) family member LIGHT is expressed on lung inflammatory cells after allergen exposure. Pharmacological inhibition of LIGHT using a fusion protein between the IgG Fc domain and lymphotoxin ß receptor (LTßR) reduces lung fibrosis, smooth muscle hyperplasia and airway hyperresponsiveness in mouse models of chronic asthma, despite having little effect on airway eosinophilia. LIGHT-deficient mice also show a similar impairment in fibrosis and smooth muscle accumulation. Blockade of LIGHT suppresses expression of lung transforming growth factor-ß (TGF-ß) and interleukin-13 (IL-13), cytokines implicated in remodeling in humans, whereas exogenous administration of LIGHT to the airways induces fibrosis and smooth muscle hyperplasia, Thus, LIGHT may be targeted to prevent asthma-related airway remodeling.


Assuntos
Remodelação das Vias Aéreas/fisiologia , Asma/fisiopatologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Animais , Asma/etiologia , Asma/patologia , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/fisiologia , Interleucina-13/fisiologia , Pulmão/patologia , Pulmão/fisiopatologia , Heterotrímero de Linfotoxina alfa1 e beta2/antagonistas & inibidores , Heterotrímero de Linfotoxina alfa1 e beta2/fisiologia , Camundongos , Camundongos Knockout , Transdução de Sinais , Fator de Crescimento Transformador beta/fisiologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/deficiência , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
8.
J Immunol ; 184(10): 5475-84, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20410485

RESUMO

Immune responses diminish with age resulting in an increased susceptibility of the elderly to infectious agents and an inability to mount protective immune responses to vaccines. Immunosenescence affects multiple aspects of the immune system, including CD8(+) T cells, which control viral infections and are assumed to prevent the development of cancers. In this study, we tested if CD8(+) T cell responses in aged mice could be enhanced through a vaccine that concomitantly expresses Ag and a molecule that blocks an immunoinhibitory pathway. Specifically, we tested a vaccine based on a replication-defective chimpanzee-derived adenovirus vector expressing the nucleoprotein (NP) of influenza A virus as a fusion protein with the HSV type 1 glycoprotein D, which through binding to the herpes virus entry mediator, blocks the immunoinhibitory herpes virus entry mediator B and T lymphocyte attenuator/CD160 pathways. Our results show that the vaccine expressing a fusion protein of NP and glycoprotein D induces significantly higher NP-specific CD8(+) T cell responses in young and aged mice compared with the vaccine expressing NP only.


Assuntos
Envelhecimento/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Vírus da Influenza A/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Transdução de Sinais/imunologia , Regulação para Cima/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Linhagem Celular , Epitopos de Linfócito T/genética , Feminino , Vetores Genéticos/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Humanos , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Nucleocapsídeo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Receptores Imunológicos/genética , Membro 14 de Receptores do Fator de Necrose Tumoral/fisiologia , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/genética , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia
9.
Eur J Immunol ; 39(10): 2765-78, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19757439

RESUMO

Nose-associated lymphoid tissue (NALT) in the rodent upper respiratory tract develops postnatally and is considered to be independent of several factors known to be involved in the organogenesis of LN and Peyer's patches (PP). In this study we demonstrate that at least two different pathways result in NALT development. Following NALT anlage formation the intrinsic pathway relies on a signaling cascade including those mediated through the chemokine receptor CXCR5 and the lymphotoxin beta receptor (LTbetaR). This allows for the formation of high endothelial venules and thereby the recruitment of lymphocytes into NALT. Alternatively, high endothelial venule formation and lymphocyte recruitment can be induced in the NALT anlage by environmental signals, which are independent of LT-betaR and chemokine receptor CXCR5 signaling but in part rely on CD40 ligand. Thus, our study identifies a novel mechanism that facilitates the rescue of NALT development at late stages in adult life independent of the canonical LTbetaR-CXCR5 signaling axis.


Assuntos
Antígenos/imunologia , Tecido Linfoide/crescimento & desenvolvimento , Receptor beta de Linfotoxina/fisiologia , Mucosa Nasal/crescimento & desenvolvimento , Receptores CXCR5/fisiologia , Transdução de Sinais/imunologia , Transferência Adotiva , Envelhecimento/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos de Superfície/metabolismo , Linfócitos B/imunologia , Ligante de CD40/imunologia , Moléculas de Adesão Celular/metabolismo , Contagem de Células , Movimento Celular/imunologia , Vida Livre de Germes/imunologia , Linfonodos/citologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Linfócitos/citologia , Tecido Linfoide/irrigação sanguínea , Tecido Linfoide/patologia , Linfotoxina-alfa/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Mucoproteínas , Mucosa Nasal/irrigação sanguínea , Mucosa Nasal/patologia , Propionibacterium acnes/imunologia , Baço/citologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Vênulas/crescimento & desenvolvimento , Vênulas/metabolismo , Vênulas/patologia
10.
Adv Exp Med Biol ; 647: 146-55, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19760072

RESUMO

Tumor necrosis factor (TNF)-related cytokines function as key communication systems between cells of the immune system and mediate inflammation and tissue destruction. LIGHT (TNFSF14) is a key component of the communication system that controls the responses of T-Cells. LIGHT activates two cell surface receptors, the Herpesvirus Entry Mediator (HVEM) and the Lymphotoxin-beta Receptor and is inhibited by soluble decoy receptor-3. The LIGHT-HVEM pathway is an important cosignaling pathway for T-Cells, whereas LIGHT-LTbetaR modifies the functions of dendritic cells and stromal cells by creating tissue microenvironments, which promote immune responses. HVEM also binds an Ig superfamily member, B and T lymphocyte attenuator (BTLA) that inhibits T-Cell activation. Thus, HVEM serves as a molecular switch between stimulatory and inhibitory signaling. Studies in humans and experimental animal models reveal that LIGHT contributes to inflammation and pathogenesis in mucosal, hepatic, joint and vascular tissues. LIGHT is accessible to biologic-based therapeutics, which can be used to target this molecule during inflammation-driven diseases.


Assuntos
Doenças Autoimunes/terapia , Membro 14 de Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Transdução de Sinais , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Doenças Vasculares/terapia , Animais , Doenças Autoimunes/imunologia , Humanos , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Doenças Vasculares/imunologia
11.
Eur J Immunol ; 39(9): 2502-14, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19701890

RESUMO

The TNF member LIGHT also known as TL4 or TNFSF14) can play a major role in cancer control via its two receptors; it induces tumor cell death through lymphotoxin-beta receptor (LT-betaR) and ligation to the herpes virus entry mediator (HVEM) amplifies the immune response. By studying the effect of LIGHT in the transcriptional profile of a lymphoid malignancy, we found that HVEM, but not LT-betaR, stimulation induces a significant increase in the expression of chemokine genes such as IL-8, and an unexpected upregulation of apoptotic genes. This had functional consequences, since LIGHT, or HVEM mAb, thus far known to costimulate T- and B-cell activation, induced chronic lymphocytic leukemia cell death. Many of the mediators involved were identified here, with an apoptotic pathway as demonstrated by caspases activation, decrease in mitochondrial membrane potential, upregulation of the pro-apoptotic protein Bax, but also a role of TRAIL. Moreover, HVEM induced endogenous TNF-alpha production and TNF-alpha enhanced HVEM-mediated cell death. HVEM function was mainly dependent on LIGHT, since other ligands like HSV-glycoprotein D and B and T lymphocyte attenuator were essentially ineffective. In conclusion, we describe a novel, as yet unknown killing effect of LIGHT through HVEM on a lymphoid malignancy, and combined with induction of chemokine release this may represent an additional tool to boost cancer immunotherapy.


Assuntos
Apoptose , Neoplasias Hematológicas/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Adulto , Idoso , Proteínas Reguladoras de Apoptose/metabolismo , Citocinas/biossíntese , Citocinas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Feminino , Humanos , Receptor beta de Linfotoxina/imunologia , Receptor beta de Linfotoxina/metabolismo , Masculino , Potencial da Membrana Mitocondrial/imunologia , Pessoa de Meia-Idade , Membro 14 de Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
12.
J Immunol ; 182(5): 3139-45, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19234211

RESUMO

Previous studies attempting to influence the severity of collagen-induced arthritis (CIA) by modulating the LIGHT (lymphotoxin-related inducible ligand that competes for glycoprotein D binding to herpesvirus entry mediator (HVEM) on T cells)/lymphotoxin pathway have yielded conflicting results. To further clarify the role of LIGHT in autoimmune arthritis, a HVEM-Ig fusion protein was used. CIA was induced in DBA1 mice, which were injected i.p. with recombinant HVEM-Ig fusion protein and control Ig at different time points. Severity of clinical arthritis and histologic joint destruction were significantly increased in HVEM-Ig-treated mice compared with control-Ig-treated mice. Collagen II-induced in vitro T cell proliferation and IFN-gamma production was augmented in mice treated with HVEM-Ig, as was the production of IgG2a anti-collagen II Ab. Accordingly, serum concentrations of IFN-gamma and IL-6 were higher in mice treated with HVEM-Ig. In conclusion, HVEM-Ig aggravates autoimmunity in collagen-induced arthritis, which is possibly mediated by interaction with B and T lymphocyte attenuator (BTLA) or CD160, despite the blockade of LIGHT. Hence, HVEM-Ig seems not to be a valid therapeutic option in autoimmune arthritis.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Herpesvirus Humano 1/imunologia , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Membro 14 de Receptores do Fator de Necrose Tumoral/imunologia , Animais , Artrite Experimental/patologia , Artrite Reumatoide/patologia , Autoanticorpos/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Colágeno Tipo II/imunologia , Modelos Animais de Doenças , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Receptor beta de Linfotoxina/administração & dosagem , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Camundongos , Camundongos Endogâmicos DBA , Receptores Imunológicos/administração & dosagem , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Membro 14 de Receptores do Fator de Necrose Tumoral/administração & dosagem , Membro 14 de Receptores do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/administração & dosagem , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
13.
Biol Pharm Bull ; 29(10): 2025-30, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17015945

RESUMO

LIGHT is a member of the TNF superfamily, which is transiently expressed on the surface of activated T lymphocytes and immature dendritic cells. Its known receptors are herpesvirus entry mediator (HVEM) prominently in T lymphocytes, and lymphtoxin beta receptor (LTbetaR) in stromal cells or nonlymphoid hematopoietic cells. Previous studies have shown that overexpression of LIGHT on T cells could lead to autoimmune reaction including lymphocytes activation, inflammation, and tissue destruction. To address the role of LIGHT/HVEM signaling in autoimmune hepatitis, an experimental colitis model induced by intravenous administration of concanavalin A (ConA) was given a soluble LTbetaR-Ig fusion protein as a competitive inhibitor of LIGHT/HVEM pathway. Marked elevation of LIGHT expression was detected in isolate intrahepatic leukocytes (IHLs) of the experimental animal. Treatment with LTbetaR-Ig significantly attenuated the progression and histological manifestations of the hepatic inflammation and reduced the production of inflammatory cytokines including TNF-alpha, IFN-gamma. Moreover, LTbetaR-Ig treatment significantly down-regulated LIGHT expression, leading to reduced lymphocytes (particularly CD4+ T cells), infiltrating into the hepatic inflammation and inhibited NF-kappaB activation and expression. We postulated that blockade of LIGHT/HVEM signaling by LTbetaR-Ig may ameliorate hepatitis by down-regulating LIGHT expression, and therefore we envision that LTbetaR-Ig would prove to a promising strategy for the clinical treatment of human autoimmune hepatitis.


Assuntos
Imunoglobulinas/farmacologia , Fígado/patologia , Receptor beta de Linfotoxina/fisiologia , Transdução de Sinais/fisiologia , Linfócitos T/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/antagonistas & inibidores , Animais , Concanavalina A/toxicidade , Hepatite/prevenção & controle , Interferon gama/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA