Your browser doesn't support javascript.
loading
Show: 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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Immunol ; 184(3): 1589-95, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20042587

RESUMEN

CD4(+) regulatory T cell (Treg) populations are believed to play very important roles in the suppression of immune responses. Overriding Treg inhibition is necessary for initiating primary immune reaction upon inflammatory Ag stimulation. LIGHT, TNF superfamily member 14, has been shown to be a costimulatory molecule for effector T cells. Overexpression of lymphotoxin-related inducible ligand that competes for glycoprotein D binding to herpesvirus entry mediator on T cells (LIGHT) on T cells induces strong T cell-mediated experimental intestinal inflammation. How this process is initiated by LIGHT in suppressive intestinal environments remains incompletely understood. In this study, we assessed the effect of LIGHT on Tregs. Our results indicate that LIGHT can support the expansion and function of Tregs. However, when LIGHT was highly expressed, these abundant Tregs failed to suppress the development of T cell-mediated experimental colitis and antitumor immunity. We showed that this might be, in part, due to an ability of LIGHT to promote effector T cell proliferation and differentiation even in a Treg-abundant environment. Our data collectively suggest that LIGHT might be a critical cytokine involved in the development of autoimmune inflammatory diseases and that LIGHT-targeted immunotherapy might be useful in the treatment of these diseases.


Asunto(s)
Terapia de Inmunosupresión , Linfocitos T Reguladores/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Colitis/genética , Colitis/inmunología , Colitis/patología , Proteínas de Homeodominio/genética , Terapia de Inmunosupresión/métodos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
11.
J Immunol ; 184(10): 5475-84, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20410485

RESUMEN

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.


Asunto(s)
Envejecimiento/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Virus de la Influenza A/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Línea Celular , Epítopos de Linfocito T/genética , Femenino , Vectores Genéticos/inmunología , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/inmunología , Humanos , Epítopos Inmunodominantes/genética , Epítopos Inmunodominantes/inmunología , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Receptores Inmunológicos/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Proteínas Recombinantes de Fusión/inmunología , Transducción de Señal/genética , 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/fisiología , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología
12.
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
13.
Circ Res ; 104(1): 60-8, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-19023130

RESUMEN

The interaction between inflammatory cytokines and endothelial cells is a critical step in atherogenesis leading to endothelial dysfunction and inflammation. We have previously reported that the tumor necrosis factor superfamily member LIGHT could be involved in atherogenesis through its ability to promote vascular inflammation. In the present study we identified proteinase-activated receptor (PAR)-2 as an inflammatory mediator that was markedly enhanced by LIGHT in endothelial cells. We also found that LIGHT acted synergistically with PAR-2 activation to promote enhanced release of the proatherogenic chemokines interleukin-8 and monocyte chemoattractant protein-1, underscoring that the interaction between LIGHT and PAR-2 is biologically active, promoting potent inflammatory effects. We showed that the LIGHT-mediated upregulation of PAR-2 in endothelial cells is mediated through the HVEM receptor, involving Jun N-terminal kinase signaling pathways. A LIGHT-mediated upregulation of PAR-2 mRNA levels was also found in human monocytes when these cells were preactivated by tumor necrosis factor alpha. We have previously demonstrated increased plasma levels of LIGHT in unstable angina patients, and here we show a similar pattern for PAR-2 expression in peripheral blood monocytes. We also found that LIGHT, LIGHT receptors, and PAR-2 showed enhanced expression, and, to some degree, colocalization in endothelial cells and macrophages, in the atherosclerotic plaques of ApoE(-/-) mice, suggesting that the inflammatory interaction between LIGHT and PAR-2 also may be operating in vivo within an atherosclerotic lesion. Our findings suggest that LIGHT/PAR-2-driven inflammation could be a pathogenic loop in atherogenesis potentially representing a target for therapy in this disorder.


Asunto(s)
Aterosclerosis/etiología , Células Endoteliales/metabolismo , Endotelio Vascular/patología , Receptor PAR-2/fisiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Vasculitis/metabolismo , Anciano , Angina de Pecho/metabolismo , Angina de Pecho/patología , Angina Inestable/metabolismo , Angina Inestable/patología , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Cultivadas/metabolismo , Quimiocina CCL2/metabolismo , Células Endoteliales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-8/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptor PAR-2/agonistas , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Vasculitis/complicaciones , Vasculitis/patología
14.
J Immunol ; 183(12): 8157-66, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20007581

RESUMEN

The islet primary nonfunction (PNF) is a serious problem in islet transplantation. In this study, we investigated whether DcR3-secreting transgenic (Tg) islets could reduce PNF. We generated Tg mice expressing human DcR3. The transgenically expressed DcR3 protected islets from IFN-gamma plus IL-1beta- or TNF-alpha plus IL-1beta-induced dysfunction and apoptosis in vitro. The Tg islets presented significantly reduced PNF after transplantation. Mechanistically, in addition to the known FasL apoptotic pathway, components of two other apoptosis pathways, that is, HVEM/LTbetaR for the LIGHT pathway and DR3 for the TL1A pathway, were found to be expressed in islets. Recombinant LIGHT- and TL1A-induced islet apoptosis in the absence of the FasL/Fas pathway, as well as DcR3, could block such induction. These results for the first time demonstrated that LIGHT and TL1A were capable of inducing islet apoptosis in addition to FasL, while DcR3 protected the islets by blocking all three apoptosis pathways. By DNA microarray analysis, we discovered that Adcyap was up-regulated >700-fold and Bank1 was down-regulated 50-fold in the cytokine-assaulted Tg islets, compared with WT islets. Forced overexpression of Adcyap1 by plasmid transfection or knockdown of Bank1 expression by small interfering RNA in insulinoma NIT-1 cells protected them from cytokine-triggered apoptosis, indicating that indeed DcR3 protects beta cells via the action of these two downstream molecules. This study has revealed novel mechanisms by which DcR3 protects islet survival, and it has identified new therapeutic targets of diabetes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Apoptosis/inmunología , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Proteínas de la Membrana/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Miembro 6b de Receptores del Factor de Necrosis Tumoral/fisiología , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/fisiología , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Humanos , Masculino , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/biosíntesis , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Miembro 6b de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
15.
J Neurosci ; 29(6): 1599-607, 2009 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19211867

RESUMEN

In a PCR screen to identify novel cytokine candidates involved in neuronal development, we identified transcripts for the tumor necrosis factor superfamily member 14 (TNFSF14), generally known as LIGHT (lymphotoxin-related inducible ligand that competes for glycoprotein D binding to herpesvirus entry mediator on T cells), together with its receptors, lymphotoxin-beta receptor (LTbetaR) and TNF family receptor herpesvirus entry mediator (HVEM), in the experimentally tractable sensory neurons of the mouse nodose ganglion. Immunocytochemistry revealed coexpression of LIGHT and its receptors in all nodose ganglion neurons in neonates. Enhancing LIGHT signaling in these neurons by overexpressing LIGHT inhibited BDNF-promoted neurite growth during a narrow window of development in the immediate perinatal period without affecting neuronal survival. Overexpressing a LIGHT mutant that selectively activates HVEM, but not one that selectively activates LTbetaR, also inhibited BDNF-promoted growth, suggesting that neurite growth inhibition is mediated via HVEM. Blocking HVEM signaling by a function-blocking anti-HVEM antibody significantly enhanced neurite growth from nodose neurons grown both with and without BDNF. Likewise, neurons from LIGHT-deficient neonates exhibited significantly greater neurite growth than neurons from wild-type littermates in both the presence and absence of BDNF. LIGHT overexpression significantly inhibited NF-kappaB activity, while preventing LIGHT-induced NF-kappaB inhibition by overexpressing the p65 and p50 NF-kappaB subunits prevented LIGHT-mediated growth inhibition. Together, these findings show that LIGHT/HVEM signaling negatively regulates neurite growth from developing sensory neurons via NF-kappaB inhibition.


Asunto(s)
Familia de Multigenes , Neuritas/fisiología , Neurogénesis/fisiología , Células Receptoras Sensoriales/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Regulación hacia Abajo , Inhibidores de Crecimiento/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Familia de Multigenes/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Factores de Necrosis Tumoral/biosíntesis , Factores de Necrosis Tumoral/fisiología
16.
Exp Parasitol ; 126(3): 326-31, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20433831

RESUMEN

Tumor necrosis factor (TNF) has long been recognized to promote malaria parasite killing, but also to contribute to the development of severe malaria disease. The precise molecular mechanisms that influence these different outcomes in malaria patients are not well understood, but the virulence and drug-resistance phenotype of malaria parasites and the genetic background and age of patients are likely to be important determinants. In the past few years, important roles for other TNF family members in host immune responses to malaria parasites and the induction of disease pathology have been discovered. In this review, we will summarize these more recent findings and highlight major gaps in our current knowledge. We will also discuss future research strategies that may allow us to better understand the sometimes subtle and intricate effects of TNF family molecules during malaria infection.


Asunto(s)
Malaria/inmunología , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Evolución Biológica , Humanos , Receptor beta de Linfotoxina/fisiología , Malaria/genética , Malaria Cerebral/etiología , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Selección Genética , Transducción de Señal , Linfocitos T Reguladores/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
17.
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
18.
Immunology ; 128(1 Suppl): e315-24, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19019090

RESUMEN

Formation of osteoclasts and consequent joint destruction are hallmarks of rheumatoid arthritis (RA). Here we show that LIGHT, a member of the tumour necrosis factor (TNF) superfamily, induced the differentiation into tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells (MNCs) of CD14(+) monocytes cocultured with nurse-like cells isolated from RA synovium, but not of freshly isolated CD14(+) monocytes. Receptor activator of nuclear factor-kappaB ligand (RANKL) enhanced this LIGHT-induced generation of TRAP-positive MNCs. The MNCs showed the phenotypical and functional characteristics of osteoclasts; they showed the expression of osteoclast markers such as cathepsin K, actin-ring formation, and the ability to resorb bone. Moreover, the MNCs expressed both matrix metalloproteinase 9 (MMP-9) and MMP-12, but the latter was not expressed in osteoclasts induced from CD14(+) monocytes by RANKL. Immunohistochemical analysis showed that the MMP-12-producing MNCs were present in the erosive areas of joints in RA, but not in the affected joints of osteoarthritic patients. These findings suggested that LIGHT might be involved in the progression of inflammatory bone destruction in RA, and that osteoclast progenitors might become competent for LIGHT-mediated osteoclastogenesis via interactions with synoviocyte-like nurse-like cells.


Asunto(s)
Artritis Reumatoide/inmunología , Monocitos/inmunología , Osteoclastos/inmunología , Membrana Sinovial/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Fosfatasa Ácida/efectos de los fármacos , Fosfatasa Ácida/inmunología , Fosfatasa Ácida/metabolismo , Artritis Reumatoide/metabolismo , Resorción Ósea/inmunología , Resorción Ósea/metabolismo , Huesos/efectos de los fármacos , Huesos/inmunología , Huesos/metabolismo , Huesos/patología , Catepsina K/efectos de los fármacos , Catepsina K/inmunología , Catepsina K/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Humanos , Isoenzimas/efectos de los fármacos , Isoenzimas/inmunología , Isoenzimas/metabolismo , Metaloproteinasa 12 de la Matriz/efectos de los fármacos , Metaloproteinasa 12 de la Matriz/inmunología , Metaloproteinasa 12 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/farmacología , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/metabolismo , Fosfatasa Ácida Tartratorresistente , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología
19.
Am J Respir Crit Care Med ; 177(2): 202-7, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17962639

RESUMEN

RATIONALE: Thrombus formation and inflammation are involved in the pathogenesis of pulmonary arterial hypertension (PAH), and LIGHT (Lymphotoxin-like Inducible protein that competes with Glycoprotein D for Herpesvirus entry mediator on T lymphocytes) has been shown to promote vascular inflammation. OBJECTIVES: We sought to investigate the role of the tumor necrosis factor superfamily ligand LIGHT in the pathogenesis of PAH. METHODS: We studied 73 patients with severe PAH and 10 control subjects. LIGHT and pro- and antithrombotic markers were assessed by enzyme immunoassays. MEASUREMENTS AND MAIN RESULTS: (1) Patients with idiopathic PAH (n = 21), patients with PAH related to risk factors or associated conditions (n = 31), and those with chronic thromboembolic PAH (n = 21) all had raised serum levels of LIGHT compared with control subjects (n = 10). (2) LIGHT levels in femoral artery were significantly related to mortality in the patients with PAH. (3) Immunostaining of LIGHT and its receptors was seen in alveolar macrophages, vascular smooth muscle cells, and endothelial cells in lungs from patients with PAH. (4) Thirteen patients received prostacyclin infusion (3 mo), and all showed hemodynamic improvement, accompanied by decreased LIGHT levels. (5) Prostacyclin abolished the release of LIGHT from activated platelets in vitro, suggesting that the decrease in LIGHT during prostacyclin therapy could involve direct effects on platelets. (6) LIGHT increased tissue factor and plasminogen activator inhibitor type 1 and decreased thrombomodulin levels in endothelial cells, inducing a prothrombotic state in these cells. CONCLUSIONS: Our findings suggest prothrombotic effects of LIGHT in PAH involving endothelium-related mechanisms, potentially contributing to the progression of this disorder.


Asunto(s)
Hipertensión Pulmonar/etiología , Trombosis/etiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Biomarcadores/sangre , Progresión de la Enfermedad , Humanos , Ligandos , 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/sangre , Venas Umbilicales/citología
20.
Biochem Biophys Res Commun ; 371(4): 834-40, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18468509

RESUMEN

Homologous to lymphotoxins, shows inducible expression, and competes with herpes simplex virus (HSV) glycoprotein D (gD) for herpes virus entry mediator (HVEM; TR2) (LIGHT), a ligand of herpes virus entry mediator (HVEM), increased reactive oxygen species (ROS) and enhanced the destruction of bacteria in human monocytes. In this study, rhLIGHT was found to increase the expression of the chemokine receptors, chemokine receptor 1 (CCR1) and CCR2, as well as to accelerate the migration activity of human monocytes. Additionally, rhLIGHT was found to increase ROS via NADPH oxidase p47(phox) phosphorylation, which was found to be required for LIGHT-induced NF-kappaB activation, CCR1 and CCR2 expression, migration and IL-8 and TNF-alpha production. Taken together, these results indicate that NADPH oxidase activation is required for rhLIGHT-induced migration in human monocytes.


Asunto(s)
Movimiento Celular , Monocitos/inmunología , NADPH Oxidasas/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Activación Enzimática , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-8/metabolismo , Monocitos/efectos de los fármacos , Monocitos/enzimología , Inhibidor NF-kappaB alfa , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Receptores CCR1/metabolismo , Receptores CCR2/metabolismo , Proteínas Recombinantes/farmacología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda