Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Int J Mol Sci ; 21(13)2020 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-32635185

RESUMEN

Obesity may affect bone health, but literature reports are contradictory about the correlation of body mass index (BMI) and bone markers. LIGHT, one of the immunostimulatory cytokines regulating the homeostasis of bone and adipose tissue, could be involved in obesity. The study involved 111 obese subjects (12.21 ± 3.71 years) and 45 controls. Patients underwent the evaluation of bone status by quantitative ultrasonography (QUS). LIGHT amounts were evaluated in sera by ELISA, whereas its expression on peripheral blood cells was evaluated by flow cytometry. Osteoclastogenesis was performed by culturing peripheral blood mononuclear cells (PBMCs) with or without anti-LIGHT antibodies. Obese patients showed significant high BMI-standard deviation score (SDS), weight-SDS, and Homeostatic model assessment for insulin resistance (HOMA-IR) that negatively correlated with the reduced Amplitude Dependent Speed of Sound (AD-SoS)-Z-score and Bone Transmission Time (BTT-Z)-score. They displayed significantly higher serum levels of LIGHT compared with controls (497.30 ± 363.45 pg/mL vs. 186.06 ± 101.41 pg/mL, p < 0.001). LIGHT expression on monocytes, CD3+-T-cells, and neutrophils was also higher in obese patients than in the controls. Finally, in PBMC cultures, the addition of anti-LIGHT antibodies induced a significant osteoclastogenesis inhibition. Our study highlighted the high serum levels of LIGHT in obese children and adolescents, and its relationship with both the grade of obesity and bone impairment.


Asunto(s)
Obesidad Infantil/sangre , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Adolescente , Biomarcadores/sangre , Índice de Masa Corporal , Densidad Ósea/fisiología , Remodelación Ósea/fisiología , Estudios de Casos y Controles , Niño , Femenino , Humanos , Resistencia a la Insulina/fisiología , Leucocitos Mononucleares/metabolismo , Modelos Lineales , Masculino , Osteogénesis/fisiología , Obesidad Infantil/diagnóstico por imagen , Obesidad Infantil/fisiopatología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
2.
Aging (Albany NY) ; 12(8): 7112-7128, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32310827

RESUMEN

Immune-checkpoint therapy has failed to show significant benefit in glioblastoma (GBM) patients. Immunologic subtypes of GBM are necessary to identify patients who might benefit from immune-checkpoint therapy. This study reviewed 152 GBM samples from The Cancer Genome Atlas (TCGA) and 214 GBM samples from Chinese Glioma Genome Atlas (CGGA). Correlation analysis showed that immune checkpoint genes (ICGs) were mainly positively correlated. The prognostic analysis of the overall survival showed that there was a significant correlation between the overall survival (OS) and the prognosis of ICGs, in which the TNFSF14 gene was a significant adverse prognostic factor. Combined with TMB and neoantigens, we found that TNFSF9 and CD27 were significantly negatively correlated with TMB and neoantigens. The association between adaptive immune pathway genes and ICG expression showed that they were positively correlated with ICGs, indicating that adaptive immune pathway genes have a certain regulatory effect on the expression of ICGs. The analysis of clinical features of the samples showed that the higher the expression of ICGs, the more likely to be correlated with mutant isocitrate dehydrogenase (IDH), while the lower the expression level of IDH, the more likely to be significantly correlated with the primary GBM. Survival analysis showed that low expression of PD-L1, IDO1, or CTLA4 with TNFSF14 in the low expression group had the best prognosis, while high expression of IDO1 or CD274 with TNFSF14 in the high expression group and low expression of CTLA4 with TNFSF14 in the high expression group had the worst prognosis. We conclude that TNFSF14 is a biomarker to identify immunologic subtype and prognosis with other ICGs in GBM and may serve as a potential therapeutic target.


Asunto(s)
Neoplasias Encefálicas/inmunología , Perfilación de la Expresión Génica , Glioblastoma/inmunología , Proteínas de Punto de Control Inmunitario/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Antígeno CTLA-4/genética , Femenino , Glioblastoma/genética , Glioblastoma/mortalidad , Humanos , Masculino , Pronóstico , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/análisis , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
3.
Inflammation ; 41(3): 1021-1031, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29460021

RESUMEN

Interaction between T cells and airway smooth muscle (ASM) cells has been identified as an important factor in the development of asthma. LIGHT (known as TNFSF14) -mediated signaling likely contributes to various inflammatory disorders and airway remodeling. The objective of this study was to investigate the roles of LIGHT-mediated pathways in the interaction between ASM cells and T cells during chronic allergic inflammation. Mice were sensitized and challenged by ovalbumin (OVA) to induce chronic airway allergic inflammation. The control group received PBS only. The histological features and LIGHT expressions in lungs were assessed in vivo. Furthermore, T cells and ASM cells derived from the model mice were co-cultured both in the presence and absence of anti-LIGHT Ab for 72 h. The effects of LIGHT blockade on expressions of downstream signaling molecules, proliferation, and apoptosis of ASM cells, differentiation of T cells, and inflammatory cytokines release were evaluated. We demonstrated that LIGHT blockade strikingly inhibited the mRNA and protein expressions of HVEM, c-JUN, and NFκB. Additionally, LIGHT blockade resulted in decreased proliferation and increased apoptosis of ASM cells. Moreover, depletion of LIGHT dramatically reduced the differentiation of CD4+ T cells into Th1, Th2, and Th17 cells, as well as inhibited inflammatory cytokines release including IL-13, TGF-ß, and IFN-γ, which are associated with CD4+ T cell differentiation and ASM cell proliferation. LIGHT plays an important role in the interaction between T cells and ASM cells in chronic allergic asthma. Blockade of LIGHT markedly suppressed ASM hyperplasia and inflammatory responses, which might be modulated through HVEM-NFκB or c-JUN pathways. Therefore, targeting LIGHT is a promising therapeutic strategy for airway inflammation and remodeling in chronic allergic asthma.


Asunto(s)
Asma/patología , Inflamación/patología , Miocitos del Músculo Liso/metabolismo , Sistema Respiratorio/patología , Linfocitos T/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Comunicación Celular , Técnicas de Cocultivo , Ratones , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
4.
Biochim Biophys Acta ; 1865(2): 204-19, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26923876

RESUMEN

The LTα1ß2 and LIGHT TNF superfamily cytokines exert pleiotropic physiological functions through the activation of their cognate lymphotoxin-ß receptor (LTßR). Interestingly, since the discovery of these proteins, accumulating evidence has pinpointed a role for LTßR signaling in carcinogenesis. Early studies have shown a potential anti-tumoral role in a subset of solid cancers either by triggering apoptosis in malignant cells or by eliciting an anti-tumor immune response. However, more recent studies provided robust evidence that LTßR signaling is also involved in diverse cell-intrinsic and microenvironment-dependent pro-oncogenic mechanisms, affecting several solid and hematological malignancies. Consequently, the usefulness of LTßR signaling axis blockade has been investigated as a potential therapeutic approach for cancer. Considering the seemingly opposite roles of LTßR signaling in diverse cancer types and their key implications for therapy, we here extensively review the different mechanisms by which LTßR activation affects carcinogenesis, focusing on the diverse contexts and different models assessed.


Asunto(s)
Receptor beta de Linfotoxina/fisiología , Neoplasias/etiología , Transducción de Señal/fisiología , Animales , Humanos , Inflamación/etiología , FN-kappa B/fisiología , Microambiente Tumoral , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
5.
Cancer Lett ; 370(1): 85-90, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26477683

RESUMEN

Recent exciting progress in cancer immunotherapy has ushered in a new era of cancer treatment. Immunotherapy can elicit unprecedented durable responses in advanced cancer patients that are much greater than conventional chemotherapy. However, such responses only occur in a relatively small fraction of patients. A positive response to immunotherapy usually relies on dynamic interactions between tumor cells and immunomodulators inside the tumor microenvironment (TME). Depending on the context of these interactions, the TME may play important roles to either dampen or enhance immune responses. Understanding the interactions between immunotherapy and the TME is not only critical to dissect the mechanisms of action but also important to provide new approaches in improving the efficiency of current immunotherapies. In this review, we will highlight recent work on how the TME can influence the efficacy of immunotherapy as well as how manipulating the TME can improve current immunotherapy regimens in some cases.


Asunto(s)
Inmunoterapia , Neoplasias/terapia , Microambiente Tumoral , Antígeno B7-H1/antagonistas & inhibidores , Antígeno CTLA-4/antagonistas & inhibidores , Quimiocina CCL21/análisis , Humanos , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Reconocimiento de Patrones/fisiología , Linfocitos T/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
6.
J Invest Dermatol ; 135(8): 2109-2118, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25789702

RESUMEN

Several inflammatory diseases including scleroderma and atopic dermatitis display dermal thickening, epidermal hypertrophy, or excessive accumulation of collagen. Factors that might promote these features are of interest for clinical therapy. We previously reported that LIGHT, a TNF superfamily molecule, mediated collagen deposition in the lungs in response to allergen. We therefore tested whether LIGHT might similarly promote collagen accumulation and features of skin fibrosis. Strikingly, injection of recombinant soluble LIGHT into naive mice, either subcutaneously or systemically, promoted collagen deposition in the skin and dermal and epidermal thickening. This replicated the activity of bleomycin, an antibiotic that has been previously used in models of scleroderma in mice. Moreover skin fibrosis induced by bleomycin was dependent on endogenous LIGHT activity. The action of LIGHT in vivo was mediated via both of its receptors, HVEM and LTßR, and was dependent on the innate cytokine TSLP and TGF-ß. Furthermore, we found that HVEM and LTßR were expressed on human epidermal keratinocytes and that LIGHT could directly promote TSLP expression in these cells. We reveal an unappreciated activity of LIGHT on keratinocytes and suggest that LIGHT may be an important mediator of skin inflammation and fibrosis in diseases such as scleroderma or atopic dermatitis.


Asunto(s)
Queratinocitos/fisiología , Piel/patología , Piel/fisiopatología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Bleomicina/farmacología , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Fibrosis/fisiopatología , Humanos , Inmunoglobulinas/deficiencia , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Recién Nacido , Queratinocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Citocinas/deficiencia , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/deficiencia , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Piel/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
7.
Mol Immunol ; 62(1): 96-103, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24980868

RESUMEN

Signals mediated by members of the tumor necrosis factor receptor superfamily modulate a network of diverse processes including initiation of inflammatory responses and altering cell fate between pathways favoring survival and death. Although such pathways have been well-described for the TNF-α receptor, less is known about signaling induced by the TNF superfamily member LIGHT and how it is differentially altered by expression of its two receptors LTßR and HVEM in the same cell. We used cell lines with different relative expression of HVEM and LTßR to show that LIGHT-induced signals mediated by these receptors were associated with altered TRAF2 stability and RelA nuclear translocation. Production of the inflammatory chemokine CXCL10 was primarily mediated by LTßR. Higher expression of HVEM was associated with cell survival, while unopposed LTßR signaling favored pathways leading to apoptosis. Importantly, restoring HVEM expression in cells with low endogenous expression recapitulated the phenotype of cells with higher endogenous expression. Together, our data provide evidence that relative expression of HVEM and LTßR modulates canonical NF-κB and pro-apoptotic signals stimulated by LIGHT.


Asunto(s)
Activación de Linfocitos , Receptor beta de Linfotoxina/fisiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/inmunología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Expresión Génica/fisiología , Células HEK293 , Células HT29 , Células HeLa , Humanos , Interferón gamma/farmacología , Ligandos , Activación de Linfocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Células U937
8.
Mediators Inflamm ; 2014: 136463, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24782592

RESUMEN

Allergic asthma can cause airway structural remodeling, involving the accumulation of extracellular matrix and thickening of smooth muscle. Tumor necrosis factor (TNF) family ligand LIGHT (TNFSF14) is a cytokine that binds herpesvirus entry mediator (HVEM)/TNFRSF14 and lymphotoxin ß receptor (LTßR). LIGHT induces asthmatic cytokine IL-13 and fibrogenic cytokine transforming growth factor-ß release from allergic asthma-related eosinophils expressing HVEM and alveolar macrophages expressing LTßR, respectively, thereby playing crucial roles in asthmatic airway remodeling. In this study, we investigated the effects of LIGHT on the coculture of human basophils/eosinophils and bronchial epithelial BEAS-2B cells. The expression of adhesion molecules, cytokines/chemokines, and matrix metalloproteinases (MMP) was measured by flow cytometry, multiplex, assay or ELISA. Results showed that LIGHT could significantly promote intercellular adhesion, cell surface expression of intercellular adhesion molecule-1, release of airway remodeling-related IL-6, CXCL8, and MMP-9 from BEAS-2B cells upon interaction with basophils/eosinophils, probably via the intercellular interaction, cell surface receptors HVEM and LTßR on BEAS-2B cells, and extracellular signal-regulated kinase, p38 mitogen activated protein kinase, and NF-κB signaling pathways. The above results, therefore, enhance our understanding of the immunopathological roles of LIGHT in allergic asthma and shed light on the potential therapeutic targets for airway remodeling.


Asunto(s)
Basófilos/metabolismo , Bronquios/citología , Eosinófilos/metabolismo , Células Epiteliales/citología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Asma/metabolismo , Adhesión Celular , Línea Celular , Membrana Celular/metabolismo , Técnicas de Cocultivo , Humanos , Hipersensibilidad/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Macrófagos/citología , Metaloproteinasa 9 de la Matriz/metabolismo , Microscopía Fluorescente , Proteínas Recombinantes/metabolismo , Transducción de Señal
9.
Arch Virol ; 159(7): 1663-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24435162

RESUMEN

The immunogenicity and efficacy of a DNA vaccine can be greatly enhanced when a gene adjuvant is used. LIGHT, a member of TNF superfamily, can function as a costimulatory molecule for human naïve T cells to proliferate and can be a potential gene adjuvant. In the current study, the eukaryotic expression plasmid pcDNA-nef was constructed by inserting a full-length nef gene into pcDNA3.1(+), and an in vitro transfection experiment suggested that the nef gene could be expressed successfully in mammalian cells. BALB/c mice were immunized with HIV-1 nef DNA vaccine plasmids alone or in combination with LIGHT expression plasmids, and the specific humoral and cellular immune responses were measured. The data showed that HIV-1 nef DNA vaccine plasmids could induce anti-Nef antibodies, Nef-specific lymphocyte proliferation and CTL activity, whereas stronger specific immune responses were induced in mice when co-immunizing with HIV-1 nef DNA vaccine plasmids and LIGHT expression plasmids, suggesting that the eukaryotic expression vector encoding HIV-1 nef is capable of inducing specific immune responses towards HIV-1 Nef and that LIGHT could be considered as a gene adjuvant for HIV-1 DNA vaccination.


Asunto(s)
Vacunas contra el SIDA/inmunología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Plásmidos/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/administración & dosificación , Adyuvantes Inmunológicos/genética , Animales , ADN Viral , Femenino , VIH-1/genética , VIH-1/metabolismo , Células HeLa , Humanos , Inmunidad Celular , Inmunidad Humoral , Ratones , Ratones Endogámicos BALB C , Linfocitos T/fisiología , Vacunas de ADN
10.
J Immunol ; 191(1): 154-63, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23720813

RESUMEN

The TNF superfamily ligand LIGHT (lymphotoxin-like, exhibits inducible expression and competes with HSV glycoprotein D for herpesvirus entry mediator [HVEM], a receptor expressed by T lymphocytes) has been shown to play a role in T cell costimulation and be involved in apoptosis of mononuclear cells. As both T cells and monocytes are key components in the development and progression of experimental autoimmune encephalomyelitis (EAE), we studied the role of LIGHT in EAE. Following immunization with myelin oligodendrocyte glycoprotein peptide (35-55), LIGHT-deficient mice developed severe EAE that resulted in an atypically high mortality rate. Histological examinations revealed intensive activation of microglia/macrophages in the CNS and higher numbers of apoptotic cells within the CNS parenchyma of LIGHT-deficient mice. However, myelin oligodendrocyte glycoprotein peptide-specific CD4(+) T cells from LIGHT-deficient mice showed reduced IFN-γ and IL-17 production and migration. Serum levels of reactive nitrogen intermediates and CNS transcripts of several proinflammatory cytokines and chemokines were also substantially decreased in the absence of LIGHT. EAE adoptive transfer experiments and bone marrow chimeras indicated that expression of LIGHT on donor cells is not required for disease induction. However, its expression on CNS host cells is a decisive factor to limit disease progression and tissue damage. Together, these data show that LIGHT expression is crucially involved in controlling activated macrophages/microglia during autoimmune CNS inflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Recuperación de la Función/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Activación de Macrófagos/genética , Activación de Macrófagos/inmunología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Recuperación de la Función/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia
11.
Biochem Biophys Res Commun ; 428(4): 451-7, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23131560

RESUMEN

Fibrosis is an abnormal response to organ injury, characterized by accumulation of activated fibroblasts at the sites of injury. Fibroblasts arise from several sources, including resident fibroblasts and circulating fibrocytes that infiltrate organ tissue. Recently, epithelial-mesenchymal transition (EMT) has been recognized as a source of mesenchymal cells. EMT is induced by various growth factors, such as transforming growth factor (TGF)-ß1, and enhanced by inflammatory cytokines. Recently the tumor necrosis factor superfamily member LIGHT has been implicated in the pathogenesis of inflammatory disease and airway remodeling in severe asthma. We hypothesized that LIGHT might contribute to the pathogenesis of airway fibrosis via enhancement of EMT. Therefore, we investigated LIGHT's ability to induce EMT. A549 cells were stimulated with LIGHT, TGF-ß1 or both for 48h. To estimate EMT, we evaluated the expression of epithelial and mesenchymal markers using immunocytochemistry, Western blotting and quantitative RT-PCR. Signaling pathways for EMT were characterized by Western analysis to detect phosphorylation of Erk1/2 and smad2. LIGHT enhanced TGF-ß1-induced EMT both morphologically, by suppressing E-cadherin and enhancing vimentin, and functionally, by enhancing cell contractility. Additionally, LIGHT induced EMT without TGF-ß1. Evaluation of the mechanism showed that LIGHT did not induce TGF-ß1 production or affect the smad-snai1 pathway. Inhibition of Erk1/2 phosphorylation reduced LIGHT-induced EMT, indicating the Erk1/2 pathway to be a key pathway in LIGHT-induced EMT. In summary, LIGHT enhanced TGF-ß1-induced EMT but also induced EMT via the Erk1/2 pathway by itself, without TGF-ß1 signaling. LIGHT may contribute to the pathogenesis of airway fibrosis through enhancement of EMT.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Alveolos Pulmonares/fisiología , Mucosa Respiratoria/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibrosis , Humanos , Sistema de Señalización de MAP Quinasas , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Mucosa Respiratoria/efectos de los fármacos , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 28(11): 1133-7, 2012 Nov.
Artículo en Chino | MEDLINE | ID: mdl-23127399

RESUMEN

AIM: To observe the effects of Xinfeng Capsule (XFC) on cardiopulmonary function of knee osteoarthritis (KOA) rats and explore its molecular mechanism. METHODS: Forty rats were randomly divided into normal control (NC) group, model control (MC) group, glucosamine control (GS) group and XFC group, 10 in each group. All rats were induced to KOA by injected papain and L-Cys into the right knee joint except NC group. Fourteen days after modeling, NC and MC groups were given normal saline (0.01 g/kg), GS and XFC groups were given glucosamine (0.098 g/kg) and XFC suspension (0.375 g/kg), respectively. Cardiac and pulmonary function were detected by ultrasonography and animal spirometry, respectively. B and T lymphocyte attenuator (BTLA), herpesvirus entry mediator (HVEM), interleukin (IL)-17, IL-4, transforming growth factor-beta (TGF-ß1) were detected by ELISA; CD4(+);CD25(+);Treg and CD4(+); CD25(+);Foxp3(+); Treg were detected by flow cytometry. RESULTS: Compared with NC group, body mass, levels of early diastolic peak flow velocity (E), early diastolic peak flow velocity/atrial peak flow velocity (E/A), forced vital capacity (FVC), forced expiratory volume in 1 second (FEV1), 25% forced expiratory flow (FEF25), 50% forced expiratory flow (FEF50), 75% forced expiratory flow (FEF75), maximal mid-expiratory flow (MMF), peak expiratory flow (PEF), expressions of BTLA, HVEM, IL-4, CD4(+);CD25(+);Treg and CD4(+);CD25(+);Foxp3(+); Treg significantly decreased, and TGF-ß1, IL-17 significantly increased in MC group (P<0.01 or P<0.05). Compared with MC group, weight, levels of E, E/A, FEV1, FEF50, FEF75, PEF, BTLA, HVEM, IL-4, CD4(+);CD25(+);Treg and CD4(+);CD25(+);Foxp3(+); Treg significantly increased, and Mankin score, cardiacindex (CI), lung index (LI), TGF-ß1, IL-17 significantly decreased in GS and XFC groups (P<0.01 or P<0.05); Compared with GS group, weight and Treg were significantly elevated in XFC group (P<0.01 or P<0.05). CONCLUSION: XFC can decrease Mankin scores of cartilage and improve cardiopulmonary function of KOA rats. Its mechanism may be enhancing BTLA-HVEM negative co-stimulatory signals, inducing Treg immune tolerance, up-regulating IL-4, down-regulating IL-17, TGF-ß1, then to inhibit abnormal inflammatory immune response.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Tolerancia Inmunológica/efectos de los fármacos , Pulmón/efectos de los fármacos , Osteoartritis de la Rodilla/tratamiento farmacológico , Receptores Inmunológicos/fisiología , Linfocitos T Reguladores/efectos de los fármacos , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Cápsulas , Pulmón/fisiopatología , Masculino , Osteoartritis de la Rodilla/inmunología , Osteoartritis de la Rodilla/fisiopatología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/inmunología
13.
Br J Haematol ; 158(6): 788-97, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22775554

RESUMEN

Chronic vascular inflammation and endothelial activation may initiate vaso-occlusion in sickle cell disease (SCD). TNFSF14 (CD258; LIGHT), a recently-identified pro-thrombotic and pro-inflammatory tumour necrosis factor (TNF)-superfamily cytokine, has a potent activating effect on endothelial cells. We evaluated whether TNFSF14 production is altered in SCD and whether platelets contribute to this production. TNFSF14 was measured in platelet-free plasma from healthy-control individuals (CON), steady-state sickle cell anaemia (SCA), SCA on hydroxycarbamide therapy (SCAHC) and haemoglobin SC (HbSC) patients. Mean plasma TNFSF14 was significantly increased in SCA, SCAHC and HbSC, compared to CON individuals. In SCA/SCAHC patients, plasma TNFSF14, showed no correlation with haematological variables, but was significantly correlated with serum lactate dehydrogenase and inflammatory markers (CD40LG , IL8 and ICAM1). Platelet-membrane TNFSF14 expression was significantly augmented on SCA platelets, and correlated with platelet activation; furthermore, measurement of platelet TNFSF14 release indicated that platelets may be a major source of circulating TNFSF14 in SCA. Interestingly, high plasma TNFSF14 was significantly associated with elevated tricuspid regurgitant velocity (≥2·5 m/s) in a population of SCA/SCAHC patients. The pro-inflammatory and atherogenic cytokine, TNFSF14, could contribute to endothelial activation and inflammation in SCA; future investigations may confirm whether this protein contributes to major clinical complications of the disease, such as pulmonary hypertension, and represents a potential therapeutic target.


Asunto(s)
Anemia de Células Falciformes/sangre , Plaquetas/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Adolescente , Adulto , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/patología , Biomarcadores , Endotelio Vascular/patología , Femenino , Genotipo , Hemoglobina C/genética , Enfermedad de la Hemoglobina C/sangre , Enfermedad de la Hemoglobina C/genética , Humanos , Hidroxiurea/uso terapéutico , Mediadores de Inflamación/sangre , Masculino , Persona de Mediana Edad , Activación Plaquetaria , Miembro 14 de Receptores del Factor de Necrosis Tumoral/sangre , Rasgo Drepanocítico/sangre , Rasgo Drepanocítico/genética , Trombofilia/etiología , Trombofilia/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Adulto Joven
15.
J Immunol ; 186(12): 7156-63, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21572030

RESUMEN

Lymph node (LN) hypertrophy, the increased cellularity of LNs, is the major indication of the initiation and expansion of the immune response against infection, vaccination, cancer, or autoimmunity. The mechanisms underlying LN hypertrophy remain poorly defined. In this article, we demonstrate that LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for HVEM, a receptor expressed by lymphocytes) (TNFSF14) is a novel factor essential for LN hypertrophy after CFA immunization. Mechanistically, LIGHT is required for the influx of lymphocytes into but not egress out of LNs. In addition, LIGHT is required for dendritic cell migration from the skin to draining LNs. Compared with wild type mice, LIGHT(-)(/)(-) mice express lower levels of chemokines in skin and addressins in LN vascular endothelial cells after CFA immunization. We unexpectedly observed that LIGHT from radioresistant rather than radiosensitive cells, likely Langerhans cells, is required for LN hypertrophy. Importantly, Ag-specific T cell responses were impaired in draining LNs of LIGHT(-)(/)(-) mice, suggesting the importance of LIGHT regulation of LN hypertrophy in the generation of an adaptive immune response. Collectively, our data reveal a novel cellular and molecular mechanism for the regulation of LN hypertrophy and its potential impact on the generation of an optimal adaptive immune response.


Asunto(s)
Hipertrofia/patología , Inflamación/patología , Ganglios Linfáticos/patología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Inmunidad Adaptativa , Animales , Quimiocinas/metabolismo , Endotelio Vascular/metabolismo , Inmunización , Ratones , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/patología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
16.
Nat Med ; 17(5): 596-603, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21499267

RESUMEN

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.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Asma/fisiopatología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Asma/etiología , Asma/patología , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/fisiología , Interleucina-13/fisiología , Pulmón/patología , Pulmón/fisiopatología , Heterotrímero de Linfotoxina alfa1 y beta2/antagonistas & inhibidores , Heterotrímero de Linfotoxina alfa1 y beta2/fisiología , Ratones , Ratones Noqueados , Transducción de Señal , Factor de Crecimiento Transformador beta/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
17.
J Exp Med ; 208(4): 797-809, 2011 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-21402741

RESUMEN

Memory T helper cells (Th cells) play an important role in host defense against pathogens but also contribute to the pathogenesis of inflammatory disorders. We found that a soluble decoy lymphotoxin ß receptor (LT-ßR)-Fc, which can block tumor necrosis factor (TNF)-related ligands LIGHT (TNFSF14) and LT-αß binding to the herpesvirus entry mediator (HVEM) and the LT-ßR, inhibited the accumulation of memory Th2 cells after antigen encounter and correspondingly reduced inflammatory responses in vivo. Showing that this was a function of the receptor for LIGHT, antigen-specific memory CD4 T cells deficient in HVEM were also unable to persist, despite having a normal immediate response to recall antigen. HVEM(-/-) memory Th2 cells displayed reduced activity of PKB (protein kinase B; Akt), and constitutively active Akt rescued their survival and restored strong inflammation after antigen rechallenge. This was not restricted to Th2 memory cells as HVEM-deficient Th1 memory cells were also impaired in surviving after encounter with recall antigen. Furthermore, the absence of LIGHT on T cells recapitulated the defect seen with the absence of HVEM, suggesting that activated T cells communicate through LIGHT-HVEM interactions. Collectively, our results demonstrate a critical role of HVEM signals in the persistence of large pools of memory CD4 T cells.


Asunto(s)
Memoria Inmunológica , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Supervivencia Celular , Femenino , Inflamación/etiología , Receptor beta de Linfotoxina/farmacología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Linfocitos T Colaboradores-Inductores/citología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
18.
Int J Cancer ; 128(6): 1363-70, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473944

RESUMEN

Lymphotoxin beta receptor (LTßR) activation on mouse fibrosarcoma cells (BFS-1) results in enhanced solid tumor growth paralleled by increased angiogenesis induced by the expression of pro-angiogenic CXCL2. In our study, we demonstrate that both functional ligands of the LTßR, namely LTα(1) ß(2) and LIGHT, are involved in the activation of LTßR in solid fibrosarcomas. To identify whether the lymphocyte population is involved in the activation of LTßR in these fibrosarcoma tumors, we used conditional LTß-deficient mice that specifically lack LTß expression either on T cells (T-LTß(-/-)) or on B cells (B-LTß(-/-)). Solid tumor growth was reduced in both mouse strains when compared to tumor growth in wild-type mice, indicating the participation of both T and B host lymphocytes in the activation of LTßR in these tumors. Tumor growth was also reduced in LIGHT-deficient mice, suggesting a contribution of this ligand to the activation of LTßR in BFS-1 fibrosarcomas. LTßR signaling can involve IκBα and/or NFκB-inducing kinase (NIK) for subsequent NFκB activation in different types of cells. Expression of a dominant negative form of IκBα or of a dominant negative mutant of NIK resulted in decreased activation of NFκB signaling and reduced expression of pro-angiogenic CXCL2 in vitro. Moreover, expression of dominant negative form of NIK or an IκBα repressor in these fibrosarcoma cells resulted in reduced solid tumor growth in vivo, suggesting that both IκBα and NIK are involved in pro-angiogenic signaling after LTßR activation. Our data support the idea that the ablation of LTßR signaling should be considered for cancer treatment.


Asunto(s)
Fibrosarcoma/patología , Heterotrímero de Linfotoxina alfa1 y beta2/metabolismo , Receptor beta de Linfotoxina/metabolismo , Linfotoxina beta/fisiología , FN-kappa B/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Apoptosis , Linfocitos B/metabolismo , Linfocitos B/patología , Western Blotting , Proliferación Celular , Células Cultivadas , Quimiocina CXCL2/metabolismo , Femenino , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología , Factor de Necrosis Tumoral alfa/farmacología
19.
Cell Death Differ ; 18(5): 754-68, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21072055

RESUMEN

Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease that primarily affects motoneurons in the brain and spinal cord. Dominant mutations in superoxide dismutase-1 (SOD1) cause a familial form of ALS. Mutant SOD1-damaged glial cells contribute to ALS pathogenesis by releasing neurotoxic factors, but the mechanistic basis of the motoneuron-specific elimination is poorly understood. Here, we describe a motoneuron-selective death pathway triggered by activation of lymphotoxin-ß receptor (LT-ßR) by LIGHT, and operating by a novel signaling scheme. We show that astrocytes expressing mutant SOD1 mediate the selective death of motoneurons through the proinflammatory cytokine interferon-γ (IFNγ), which activates the LIGHT-LT-ßR death pathway. The expression of LIGHT and LT-ßR by motoneurons in vivo correlates with the preferential expression of IFNγ by motoneurons and astrocytes at disease onset and symptomatic stage in ALS mice. Importantly, the genetic ablation of Light in an ALS mouse model retards progression, but not onset, of the disease and increases lifespan. We propose that IFNγ contributes to a cross-talk between motoneurons and astrocytes causing the selective loss of some motoneurons following activation of the LIGHT-induced death pathway.


Asunto(s)
Muerte Celular/efectos de los fármacos , Interferón gamma/fisiología , Receptor beta de Linfotoxina/metabolismo , Neuronas Motoras/fisiología , Superóxido Dismutasa/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/fisiología , Caspasas/metabolismo , Supervivencia Celular , Células Cultivadas , Activación Enzimática , Eliminación de Gen , Humanos , Interferón gamma/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Missense , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Receptor fas/metabolismo
20.
J Immunol ; 185(3): 1949-58, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20592286

RESUMEN

The TNF superfamily member homologous to lymphotoxins, exhibits inducible expression, and competes with HSV glycoprotein D for herpesvirus entry mediator (HVEM), a receptor expressed by T lymphocytes (LIGHT) [TNF superfamily (SF)-14], is a key cytokine that activates T cells and dendritic cells and is implicated as a mediator of inflammatory, metabolic, and malignant diseases. LIGHT engages the lymphotoxin-beta receptor (LTbetaR) and HVEM (TNFRSF14), but is competitively limited in activating these receptors by soluble decoy receptor-3 (DcR3; TNFRSF6B). Two variants in the human LIGHT alter the protein at E214K (rs344560) in the receptor-binding domain and S32L (rs2291667) in the cytosolic domain; however, the functional impact of these polymorphisms is unknown. A neutralizing Ab failed to bind the LIGHT-214K variant, indicating this position as a part of the receptor-binding region. Relative to the predominant reference variant S32/E214, the other variants showed altered avidity with LTbetaR and less with HVEM. Heterotrimers of the LIGHT variants decreased binding avidity to DcR3 and minimized the inhibitory effect of DcR3 toward LTbetaR-induced activation of NF-kappaB. In patients with immune-mediated inflammatory diseases, such as rheumatoid arthritis, DcR3 protein levels were significantly elevated. Immunohistochemistry revealed synoviocytes as a significant source of DcR3 production, and DcR3 hyperexpression is controlled by posttranscriptional mechanisms. The increased potential for LTbetaR signaling, coupled with increased bioavailability due to lower DcR3 avidity, provides a mechanism of how polymorphic variants in LIGHT could contribute to the pathogenesis of inflammatory diseases.


Asunto(s)
Variación Genética/inmunología , Polimorfismo de Nucleótido Simple/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Secuencia de Aminoácidos , Disponibilidad Biológica , Técnicas de Cocultivo , Células HeLa , Humanos , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/fisiología , Modelos Inmunológicos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , FN-kappa B/antagonistas & inhibidores , Unión Proteica/genética , Unión Proteica/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Miembro 6b de Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...