Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 35(11): e22008, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34679191

RESUMEN

An impaired blood-retinal barrier (BRB) leads to diabetic macular edema (DME), which is a major complication of Diabetic retinopathy (DR). Mediators such as inflammation cause BRB breakdown. However, the explicit mechanism of its disruption is largely unknown. In this study, we identified tumor necrosis factor ligand-related molecule 1A (TL1A) as a crucial factor which protect retinal endothelial cells integrity in DR. By providing both human and mouse data, we show that TL1A is significantly decreased in the retinas of DME patients and diabetic rodents. We further demonstrate that the loss of TL1A accelerated diabetes-induced retinal barrier breakdown. TL1A supplementation protects the diabetic retina against BRB breakdown. Mechanistically, TL1A stabilize intracellular junctions and protect vascular integrity by blocking SHP1-Src-regulated VE-cadherin phosphorylation. Collectively, our findings reveal that loss of TL1A in the retina leads to increased vascular permeability in DR, and that TL1A treatment is of potential therapeutic interest for the treatment of DME.


Asunto(s)
Barrera Hematorretinal/metabolismo , Permeabilidad Capilar , Retinopatía Diabética/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Células Endoteliales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Vasos Retinianos
2.
Proc Natl Acad Sci U S A ; 110(34): 13863-8, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23918400

RESUMEN

Mouse bone marrow-derived Lin(-)-Sca-1(+) endothelial progenitor cell (EPC) has pluripotent abilities such as supporting neovascularization. Vascular endothelial growth factor (VEGF) receptor 1 (VEGFR1) (Flt1) recognizes various VEGF isoforms and is critically implicated in a wide range of physiological and pathological settings, including vasculogenesis. Mouse EPC expresses two isoforms of VEGFR1: mFlt1, which transmits ligand-induced signals; and sFlt1, which acts as a negative regulator by sequestering ligands of VEGF receptors. How the relative levels of mFlt1 and sFlt1 are regulated is not yet clear. We report here that tumor necrosis factor superfamily 15 (TNFSF15) (also known as VEGI or TL1A), an endothelial cell-secreted cytokine, simultaneously promotes mFlt1 degradation and up-regulates sFlt1 expression in EPC, giving rise to disruption of VEGF- or PlGF-induced activation of eNOS and MAPK p38 and effective inhibition of VEGF-driven, EPC-supported vasculogenesis in a murine Matrigel implant model. TNFSF15 treatment of EPC cultures facilitates Akt deactivation-dependent, ubiquitin-assisted degradation of mFlt1 and stimulates sFlt1 expression by activating the PKC, Src, and Erk1/2 signaling pathway. Additionally, TNFSF15 promotes alternative splicing of the Flt1 gene in favor of sFlt1 production by down-regulating nuclear protein Jumonji domain-containing protein 6 (Jmjd6), thus alleviating Jmjd6-inhibited sFlt1 expression. These findings indicate that TNFSF15 is a key component of a molecular mechanism that negatively modulates EPC-supported vasculogenesis through regulation of the relative levels of mFlt1 and sFlt1 in EPC.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Neovascularización Fisiológica/fisiología , Proteolisis , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Empalme Alternativo/fisiología , Análisis de Varianza , Animales , Western Blotting , Colágeno , Combinación de Medicamentos , Células Endoteliales/metabolismo , Laminina , Ligandos , Ratones , Microscopía Fluorescente , Isoformas de Proteínas/metabolismo , Proteoglicanos , Células Madre/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
3.
J Immunol ; 190(4): 1540-50, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23319737

RESUMEN

TNF superfamily member 15 (TL1A) is the ligand for TNFR superfamily (TNFRSF)25. We previously reported that TNFRSF25 stimulation with an agonist Ab, 4C12, expands pre-existing CD4(+)Foxp3(+) regulatory T cells (Tregs) in vivo. To determine how the physiological ligand differs from the Ab, we generated a soluble mouse TL1A-Ig fusion protein that forms a dimer of TL1A trimers in solution with an apparent molecular mass of 516 kDa. In vitro, TL1A-Ig mediated rapid proliferation of Foxp3(+) Tregs and a population of CD4(+)Foxp3(-) conventional T cells. TL1A-Ig also blocked de novo biogenesis of inducible Tregs and it attenuated the suppressive function of Tregs. TNFRSF25 stimulation by TL1A-Ig in vivo induced expansion of Tregs such that they increased to 30-35% of all CD4(+) T cells in the peripheral blood within 5 d of treatment. Treg proliferation in vivo was dependent on TCR engagement with MHC class II. Elevated Treg levels can be maintained for at least 20 d with daily injections of TL1A-Ig. TL1A-Ig-expanded Tregs expressed high levels of activation/memory markers KLRG1 and CD103 and were highly suppressive ex vivo. TL1A-Ig-mediated Treg expansion in vivo was protective against allergic lung inflammation, a mouse model for asthma, by reversing the ratio of conventional T cells to Tregs in the lung and blocking eosinophil exudation into the bronchoalveolar fluid. Thus, TL1A-Ig fusion proteins are highly active and tightly controllable agents to stimulate Treg proliferation in vivo, and they are uniquely able to maintain high levels of expanded Tregs by repeated administration.


Asunto(s)
Proteínas Recombinantes de Fusión/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/aislamiento & purificación , Animales , Células CHO , Línea Celular Tumoral , Clonación Molecular , Cricetinae , Citometría de Flujo , Genes Reporteros , Cadenas Pesadas de Inmunoglobulina/biosíntesis , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Ratones , Ratones Endogámicos C57BL , Mutagénesis Insercional , Células 3T3 NIH , Plásmidos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/inmunología , Hipersensibilidad Respiratoria/metabolismo , Transfección , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
4.
BMC Nephrol ; 15: 178, 2014 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-25399326

RESUMEN

BACKGROUND: Death receptors (DRs) play an important role in renal pathology. We have shown that DR3 is inducibly expressed on renal tubular epithelial cells in the setting of inflammatory injuries. In this study we investigate the expression of DR3 in renal endothelial cells and their response to TL1A, the only known ligand of DR3. METHODS: We did RT-PCR, flow cytometry and subcellular immunoblotting to examine the expression and function of DR3 in cells in vitro. We did organ culture of human and mouse tissue to examine expression and signal of DR3 in vivo. RESULTS: DR3 is expressed in some interstitial vascular endothelial cells (EC) in human kidney in situ; these EC also respond to its ligand TL1A by activating NF-κB. Very low levels of DR3 can be detected on the cell surface of cultured human umbilical vein (HUV) EC, which do not respond to TL1A. HUVEC transfected to overexpress DR3 become responsive to TL1A, assessed by IκBα degradation and E-selectin induction, indicating that the signaling components needed for DR3 responsiveness are expressed. TL1A induces NF-κB activation in EC in renal and cardiac tissue from wild type but not DR3 knock-out mice. CONCLUSION: TL1A and DR3 activate NF-κB in vascular endothelial cells, and can be an important regulator of renal interstitial vascular injury.


Asunto(s)
Células Endoteliales/metabolismo , Riñón/citología , FN-kappa B/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas I-kappa B/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/química , Inhibidor NF-kappaB alfa , Técnicas de Cultivo de Órganos , Miembro 25 de Receptores de Factores de Necrosis Tumoral/biosíntesis , Miembro 25 de Receptores de Factores de Necrosis Tumoral/deficiencia , Proteínas Recombinantes/farmacología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/farmacología
5.
FASEB J ; 26(8): 3575-86, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593543

RESUMEN

Death receptor 3 (DR3, TNFRSF25), the closest family relative to tumor necrosis factor receptor 1, promotes CD4(+) T-cell-driven inflammatory disease. We investigated the in vivo role of DR3 and its ligand TL1A in viral infection, by challenging DR3-deficient (DR3(KO)) mice and their DR3(WT) littermates with the ß-herpesvirus murine cytomegalovirus or the poxvirus vaccinia virus. The phenotype and function of splenic T-cells were analyzed using flow cytometry and molecular biological techniques. We report surface expression of DR3 by naive CD8(+) T cells, with TCR activation increasing its levels 4-fold and altering the ratio of DR3 splice variants. T-cell responses were reduced up to 90% in DR3(KO) mice during acute infection. Adoptive transfer experiments indicated this was dependent on T-cell-restricted expression of DR3. DR3-dependent CD8(+) T-cell expansion was NK and CD4 independent and due to proliferation, not decreased cell death. Notably, impaired immunity in DR3(KO) hosts on a C57BL/6 background was associated with 4- to 7-fold increases in viral loads during the acute phase of infection, and in mice with suboptimal NK responses was essential for survival (37.5%). This is the first description of DR3 regulating virus-specific T-cell function in vivo and uncovers a critical role for DR3 in mediating antiviral immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Herpesviridae/inmunología , Muromegalovirus/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Traslado Adoptivo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ratones , Ratones Noqueados , Carga Viral
6.
Biogerontology ; 14(5): 543-56, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24030243

RESUMEN

Aging of the bladder detrusor muscle plays an important role in lower urinary tract symptoms in elderly people. Our previous work demonstrated that elderly patients have increased levels of vascular endothelial growth inhibitor (VEGI) in bladder tissue. Therefore, we hypothesized that VEGI may play a role in aging of the bladder detrusor muscle cells. This study aims to develop and characterize primary cultures of aged porcine bladder detrusor muscle cells in order to explore the expression and function of VEGI. Bladder samples from female pigs were divided into two groups: the aged group (Model) and the young group (Control). We confirmed ß-galactosidase expression, a marker for senescence, in aged muscle cells (identified by α-smooth muscle actin (α-SMA) staining), but not in the young group. mRNA levels of VEGI-251 and death receptor 3 (DR3) were up-regulated (P < 0.05) and total cell protein levels of VEGI-251, DR3 and nuclear factor-kappa B [NF-κB (p65)], membrane protein levels of DR3, and nuclear protein levels of NF-κB (p65) were significantly higher (P < 0.01) in the Model cells compared to Control cells. In conclusion, we have established a method to culture aged detrusor muscle cells derived from porcine bladder. Higher levels of VEGI-251, DR3 and NF-κB (p65) were observed in the aged cells. VEGI-251 may function by increasing DR3 on cellular membranes and promoting the transfer of NF-κB into the nucleus. This suggests that VEGI may be a target for reversing the aging process of bladder detrusor muscle cells.


Asunto(s)
Músculo Liso/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Vejiga Urinaria/metabolismo , Factores de Edad , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN , Femenino , Técnica del Anticuerpo Fluorescente , Músculo Liso/citología , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Vejiga Urinaria/citología
7.
J Immunol ; 186(7): 4175-82, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21368235

RESUMEN

The nervous system influences immune responses through the release of neural factors such as neuropeptides. Among them, the tachykinin substance P (SP) signals via the neurokinin 1 receptor (NK-1R), which is expressed by various immune cells. We thereby analyzed in this paper whether tachykinins may participate in human CD4(+) Th cell polarization. We report that SP and hemokinin-1 (HK-1) upregulate IL-17A and IFN-γ production by human memory CD4(+) T cells without affecting IL-4 and IL-10 production. SP and HK-1 switch non-Th17-committed CD4(+) memory T cells into bona fide Th17 cells and Th1/Th17 cells. In contrast, SP and HK-1 do not modulate the polarization of naive CD4(+) T cells. SP- and HK-1-induced Th17 cell generation is mediated through NK-1R and requires the presence of monocytes. SP and HK-1 trigger IL-1ß, IL-6, and TNF-α production, upregulate IL-23 production, and enhance TNF-like 1A expression on monocyte surface. Neutralization experiments demonstrated that IL-1ß, IL-23, and TNF-like 1A are involved in the SP- and HK-1-induced Th17 cell. The other members of the tachykinin family, neurokinins A and B, have no effect on the differentiation of naive and memory T cells. These results thereby show that SP and HK-1 are novel Th17 cell-inducing factors that may act locally on memory T cells to amplify inflammatory responses.


Asunto(s)
Diferenciación Celular/inmunología , Memoria Inmunológica , Interleucina-1beta/biosíntesis , Interleucina-23/biosíntesis , Monocitos/inmunología , Sustancia P/fisiología , Taquicininas/fisiología , Células Th17/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Comunicación Celular/genética , Comunicación Celular/inmunología , Diferenciación Celular/genética , Polaridad Celular/genética , Polaridad Celular/inmunología , Células Cultivadas , Humanos , Memoria Inmunológica/genética , Mediadores de Inflamación/fisiología , Interleucina-1beta/genética , Interleucina-1beta/fisiología , Interleucina-23/genética , Interleucina-23/fisiología , Monocitos/metabolismo , Monocitos/patología , Células Th17/metabolismo , Células Th17/patología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
8.
Mediators Inflamm ; 2013: 258164, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24453414

RESUMEN

TNF-like ligand 1A (TL1A), which binds its cognate receptor DR3 and the decoy receptor DcR3, is an identified member of the TNF superfamily. TL1A exerts pleiotropic effects on cell proliferation, activation, and differentiation of immune cells, including helper T cells and regulatory T cells. TL1A and its two receptors expression is increased in both serum and inflamed tissues in autoimmune diseases such as inflammatory bowel disease (IBD), rheumatoid arthritis (RA), and ankylosing spondylitis (AS). Polymorphisms of the TNFSF15 gene that encodes TL1A are associated with the pathogenesis of irritable bowel syndrome, leprosy, and autoimmune diseases, including IBD, AS, and primary biliary cirrhosis (PBC). In mice, blocking of TL1A-DR3 interaction by either antagonistic antibodies or deletion of the DR3 gene attenuates the severity of multiple autoimmune diseases, whereas sustained TL1A expression on T cells or dendritic cells induces IL-13-dependent small intestinal inflammation. This suggests that modulation of TL1A-DR3 interaction may be a potential therapeutic target in several autoimmune diseases, including IBD, RA, AS, and PBC.


Asunto(s)
Enfermedades Autoinmunes/etiología , Inflamación/etiología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Artritis Reumatoide/etiología , Humanos , Enfermedades Inflamatorias del Intestino/etiología , Psoriasis/etiología , Transducción de Señal , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
9.
J Immunol ; 184(10): 5827-34, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20410491

RESUMEN

TNF-like protein 1A (TL1A), a TNF superfamily cytokine that binds to death receptor 3 (DR3), is highly expressed in macrophage foam cell-rich regions of atherosclerotic plaques, although its role in foam cell formation has yet to be elucidated. We investigated whether TL1A can directly stimulate macrophage foam cell formation in both THP-1 and primary human monocyte-derived macrophages with the underlying mechanisms involved. We demonstrated that TL1A promotes foam cell formation in human macrophages in vitro by increasing both acetylated and oxidized low-density lipoprotein uptake, by enhancing intracellular total and esterified cholesterol levels and reducing cholesterol efflux. This imbalance in cholesterol homeostasis is orchestrated by TL1A-mediated changes in the mRNA and protein expression of several genes implicated in the uptake and efflux of cholesterol, such as scavenger receptor A and ATP-binding cassette transporter A1. Furthermore, through the use of virally delivered DR3 short-hairpin RNA and bone marrow-derived macrophages from DR3 knockout mice, we demonstrate that DR3 can regulate foam cell formation and contributes significantly to the action of TL1A in this process in vitro. We show, for the first time, a novel proatherogenic role for both TL1A and DR3 that implicates this pathway as a target for the therapeutic intervention of atherosclerosis.


Asunto(s)
Diferenciación Celular/inmunología , Células Espumosas/citología , Células Espumosas/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Transducción de Señal/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Aterosclerosis/inmunología , Aterosclerosis/patología , Transporte Biológico/inmunología , Línea Celular Tumoral , Células Cultivadas , Proteínas de Transferencia de Ésteres de Colesterol/antagonistas & inhibidores , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Femenino , Células Espumosas/patología , Humanos , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Lipoproteínas LDL/metabolismo , Ratones , Ratones Noqueados , Miembro 25 de Receptores de Factores de Necrosis Tumoral/deficiencia , Regulación hacia Arriba/inmunología
10.
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
11.
Inflamm Bowel Dis ; 25(1): 14-26, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30295722

RESUMEN

Background: TNF-like cytokine 1A (TL1A) and its functional receptor, death-domain-receptor-3 (DR3), are multifunctional mediators of effector and regulatory immunity. We aimed to evaluate the functional role and therapeutic potential of TL1A/DR3 signaling in Crohn's disease-like ileitis. Methods: Ileitis-prone SAMP1/YitFc (SAMP) and TNFΔARE/+ mice were rendered deficient for DR3 or TL1A by microsatellite marker-assisted backcrossing. Pathological and immunological characteristics were compared between control and knockout mice, and mucosal immunophenotype was analyzed by Nanostring microarray assay. The therapeutic effect of pharmacological TL1A neutralization was also investigated. Results: DR3 deficiency was associated with restoration of a homeostatic mucosal immunostat in SAMP mice through the regulation of several pro- and anti-inflammatory genes. This led to suppression of effector immunity, amelioration of ileitis severity, and compromised ability of either unfractionated CD4+ or CD4+CD45RBhi mucosal lymphocytes to transfer ileitis to severe combined immunodeficient mice recipients. TNF-driven ileitis was also prevented in TNFΔARE/+xDR3-/- mice, in association with decreased expression of the pro-inflammatory cytokines TNF and IFN-γ. In contrast to DR3, TL1A was dispensable for the development of ileitis although it affected the kinetics of inflammation, as TNFΔARE/+xTL1A-/- demonstrated delayed onset of inflammation, whereas administration of a neutralizing, anti-TL1A antibody ameliorated early but not late TNFΔARE/+ ileitis. Conclusion: We found a prominent pro-inflammatory role of DR3 in chronic ileitis, which is only partially mediated via interaction with TL1A, raising the possibility for additional DR3 ligands. Death-domain-receptor-3 appears to be a master regulator of mucosal homeostasis and inflammation and may represent a candidate therapeutic target for chronic inflammatory conditions of the bowel.


Asunto(s)
Enfermedad de Crohn/complicaciones , Regulación de la Expresión Génica , Ileítis/prevención & control , Inflamación/prevención & control , Miembro 25 de Receptores de Factores de Necrosis Tumoral/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Ileítis/etiología , Inflamación/etiología , Mediadores de Inflamación/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
12.
Inflamm Bowel Dis ; 25(3): 510-523, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30462201

RESUMEN

BACKGROUND: The tumor necrosis factor alpha (TNFα)-homologous cytokine TL1A is emerging as a major player in intestinal inflammation. From in vitro experiments on human lymphocytes, TNF-like molecule 1A (TL1A) is known to activate a highly inflammatory lymphoid response in synergy with interleukin (IL)-12 and IL-18. Carriers of specific genetic polymorphisms associated with IL-12, IL-18, or TL1A signaling have increased Crohn's disease risk, and all 3 cytokines are upregulated during active disease. The study aim was to investigate whether the type 1-polarizing cytokines IL-12 and IL-18 could directly initiate intestinal pathology in mice and how TL1A would influence the resulting inflammatory response. METHODS: Conventional barrier-bred and germ-free mice were randomly allocated to different groups and injected twice with different combinations of IL-12, IL-18, and TL1A, and killed 3 days after the first injection. All treatment groups were co-housed and fed a piroxicam-supplemented chow diet. RESULTS: Intestinal pathology was evident in IL-12- and IL-18-treated mice and highly exacerbated by TL1A in both the colon and ileum. The cytokine-induced intestinal inflammation was characterized by epithelial damage, increased colonic levels of TNFα, IL-1ß, IFN-γ, and IL-6, and various chemokines along with gut microbiota alterations exhibiting high abundance of Enterobacteriaceae. Furthermore, the inflamed ileum and colon exhibited a TL1A-specific increased infiltration of intraepithelial natural killer cells co-expressing NKG2D and IL-18Ra and a higher frequency of unconventional T cells in the colonic epithelium. Upon cytokine injection, germ-free mice exhibited similar intraepithelial lymphoid infiltration and increased colonic levels of IFNγ and TNFα. CONCLUSIONS: This study demonstrates that TL1A aggravates IL-12- and IL-18-induced intestinal inflammation in the presence and absence of microbiota.


Asunto(s)
Células Epiteliales/inmunología , Tracto Gastrointestinal/inmunología , Inflamación/etiología , Subunidad p35 de la Interleucina-12/administración & dosificación , Interleucina-18/administración & dosificación , Células Asesinas Naturales/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
13.
Clin Immunol ; 129(2): 249-55, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18757243

RESUMEN

TL1A is a novel TNF-like cytokine, which provides co-stimulatory and Th1-polarizing signals to activated lymphocytes, via binding to death-domain receptor 3 (DR3). These functions are inhibited when TL1A associates to decoy receptor 3 (DcR3). We investigated the serum expression of TL1A and DcR3 in 81 patients with RA and 51 healthy controls. TL1A concentrations were elevated in patients by 5-fold (P<0.00001). This increase was more prominent in RFactor-positive patients and correlated with clinical activity in this subgroup. DcR3 was detected more frequently and in significantly higher values in RA-derived sera, correlated strongly with TL1A, and was present in inflammatory synovial fluid. Severe RA stage was associated with highly elevated TL1A and DcR3 serum levels. Treatment with an anti-TNF agent significantly decreased TL1A serum levels. We conclude that TL1A may serve as an inflammatory marker in RA. Interactions between TL1A and its receptors may be important in the pathogenesis of RA.


Asunto(s)
Artritis Reumatoide/inmunología , Miembro 6b de Receptores del Factor de Necrosis Tumoral/sangre , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Adulto , Anciano , Artritis Reumatoide/etiología , Biomarcadores , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
14.
Anticancer Res ; 33(9): 3815-22, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24023314

RESUMEN

Pituitary ademonas are benign tumours from the pituitary gland but may have an invasive and destructive growth pattern. There is little understanding of the growth and progression control of pituitary tumours. In the present study, we investigated the expression of vascular endothelial growth inhibitor (VEGI), a vascular endothelial growth and apoptosis regulator and VEGI receptor Death Receptor-3 (DR3), in clinical pituitary tumours. Pituitary tumours from 95 patients were included in the study. Fresh pituitary tumours were obtained immediately after surgery and processed for histological and molecular-based analyses. Histopathological and clinical information including tumour size, tumour invasion and endocrine status were analyzed against the gene transcript expression of VEGI, DR3 and VEGF. VEGI and VEGF family and VEGF receptors were quantitatively determined for their gene transcript expression. The expression levels of VEGI were significantly lower in pituitary tumours which invaded the sella floor, and with suprasellar extension than in non-invasive tumours (p=0.0073). VEGI levels were also negatively correlated with cavernous sinus invasion stage (p<0.0001), in that a high level of VEGI was associated with low tumour grade. Multivariate analysis indicated that VEGI is an independent factor predictive of invasion (p=0.05). It was further demonstrated that the relationship between VEGI and pituitary tumour invasion were independent of the expression of VEGF and its receptors. Low levels of VEGI transcripts were associated with the intratumoural haemorrhage (p=0.05). Out of all the pituitary tumours, 59 were non-functional. Out of the functional tumours, it was found that follicle stimulating hormone (FSH)-expressing and gonadotrophic tumours tended to have markedly low levels of VEGI transcripts, compared with non-functional tumours (p=0.0026 and p=0.003, respectively). The opposite was seen with thyroid-stimulating hormone (TSH)-secreting tumours. Levels of DR3 in tumours with sella destruction were also lower than in those without destruction. VEGI, possibly via DR3, suppresses the aggressive nature of pituitary tumours and its expression level is closely linked to the invasion and destruction of the suprasellar and sella regions. It also has implications for the endocrine nature of these tumours. VEGI thus has an important predictive and prognostic value in patients with pituitary tumours.


Asunto(s)
Adenoma/patología , Invasividad Neoplásica , Neoplasias Hipofisarias/patología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Secuencia de Bases , Cartilla de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Hipofisarias/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Oncogene ; 30(16): 1892-900, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21217782

RESUMEN

Lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF) α factor (LITAF) is a multiple functional molecule whose sequence is identical to the small integral membrane protein of the lysosome/late endosome. LITAF was initially identified as a transcription factor that activates transcription of proinflammatory cytokine in macrophages in response to LPS. Mutations of the LITAF gene are associated with a genetic disease, called Charcot-Marie-Tooth syndrome. Recently, we have reported that mRNA levels of LITAF and TNF superfamily member 15 (TNFSF15) are upregulated by 5' adenosine monophosphate (AMP)-activated protein kinase (AMPK). The present study further assesses their biological functions. Thus, we show that 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), a pharmacological activator of AMPK, increases the abundance of LITAF and TNFSF15 in LNCaP and C4-2 prostate cancer cells, which is abrogated by small hairpin RNA (shRNA) or the dominant-negative mutant of AMPK α1 subunit. Our data further demonstrate that AMPK activation upregulates the transcription of LITAF. Intriguingly, silencing LITAF by shRNA enhances proliferation, anchorage-independent growth of these cancer cells and tumor growth in the xenograft model. In addition, our study reveals that LITAF mediates the effect of AMPK by binding to a specific sequence in the promoter region. Furthermore, we show that TNFSF15 remarkably inhibits the growth of prostate cancer cells and bovine aortic endothelial cells in vitro, with a more potent effect toward the latter. In conjuncture, intratumoral injection of TNFSF15 significantly reduces the size of tumors and number of blood vessels and induces changes that are characteristic of tumor cell differentiation. Therefore, our studies for the first time establish the regulatory axis of AMPK-LITAF-TNFSF15 and also suggest that LITAF may function as a tumor suppressor.


Asunto(s)
Adenilato Quinasa/metabolismo , Neoplasias/patología , Proteínas Nucleares/fisiología , Factores de Transcripción/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Activación Enzimática , Humanos , Neovascularización Patológica , Proteínas Nucleares/genética , Factores de Transcripción/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
16.
Inflamm Bowel Dis ; 16(4): 568-75, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19834969

RESUMEN

BACKGROUND: Tumor necrosis factor (TNF)-like protein 1A (TL1A) is a member of the TNF superfamily and contributes to the pathogenesis of Crohn's disease (CD) by stimulating T-helper (Th) 1 cells. In addition to Th1, recent studies have focused on the role of Th17 cells in the pathogenesis of CD. Here we tried to clarify the role of TL1A in Th1 and Th17 immunity in CD. METHODS: TL1A expression was assessed by quantitative reverse-transcription polymerase chain reaction (RT-PCR) in lamina propria (LP) macrophages (LP-M phi s) from normal controls (NC) and patients with CD or ulcerative colitis (UC). Purified LP CD4(+) T cells were stimulated with TL1A and/or IL-23 and interferon gamma (IFN-gamma) and interleukin (IL)-17 levels were analyzed. We also examined the effect of TL1A on naïve CD4(+) T-cell differentiation. RESULTS: We found that LP-M phi s are a major producer of TL1A. TL1A expression was markedly enhanced in LP-M phi s from CD patients compared with NC or UC patients. IL-23, in addition to TL1A, was induced in LP-M phi s by commensal bacteria stimulation. TL1A and IL-23 synergistically promoted the production of IFN-gamma and IL-17 by LP T cells, while TL1A alone did not induce cytokine production. Furthermore, TL1A promoted Th17 differentiation from naïve T cells by LP-M phi s; however, IL-23 did not show any synergistic effects on Th17 differentiation. CONCLUSIONS: TL1A expressed in LP-M phi s might play an important role in the pathogenesis of CD by inducing Th1 and Th17 immunity. IL-23 differentially regulated these functions of TL1A on memory and naïve T cells.


Asunto(s)
Enfermedad de Crohn/inmunología , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Células TH1/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Células Cultivadas , Células Dendríticas/inmunología , Humanos , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Activación de Linfocitos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Gerontol A Biol Sci Med Sci ; 65(11): 1165-80, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20675618

RESUMEN

Tumor necrosis factor-like cytokine 1A (TL1A) is expressed in endothelial cells and contributes to T-cell activation, via an extracellular fragment TL1A(L72-L251), generated by ectodomain shedding. Fragments of TL1A, referred to as vascular endothelial growth inhibitor, were found to induce growth arrest and apoptosis in endothelial cells; however, the underlying mechanisms remained obscure. Here, we show that full-length TL1A is the major detectable gene product in both human umbilical vein endothelial cells and circulating endothelial progenitor cells. TL1A expression was significantly enhanced in senescent circulating endothelial progenitor cells, and knockdown of TL1A partially reverted senescence. TL1A overexpression induced premature senescence in both circulating endothelial progenitor cells and human umbilical vein endothelial cells. We also identified a novel extracellular fragment of TL1A, TL1A(V84-L251), resulting from differential ectodomain shedding, which induced growth arrest and apoptosis in human umbilical vein endothelial cells. These findings suggest that TL1A is involved in the regulation of endothelial cell senescence, via a novel fragment produced by differential ectodomain shedding.


Asunto(s)
Senescencia Celular/fisiología , Células Endoteliales/fisiología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Apoptosis , Western Blotting , Proliferación Celular , Células Cultivadas , Dipéptidos/farmacología , Electroporación/métodos , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Escherichia coli , Humanos , Ácidos Hidroxámicos/farmacología , Isoformas de Proteínas/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Venas Umbilicales/citología , Venas Umbilicales/metabolismo , beta-Galactosidasa/metabolismo
18.
J Exp Med ; 205(5): 1049-62, 2008 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-18411337

RESUMEN

T helper type 17 (Th17) cells play an important pathogenic function in autoimmune diseases; their regulation, however, is not well understood. We show that the expression of a tumor necrosis factor receptor family member, death receptor 3 (DR3; also known as TNFRSF25), is selectively elevated in Th17 cells, and that TL1A, its cognate ligand, can promote the proliferation of effector Th17 cells. To further investigate the role of the TL1A-DR3 pathway in Th17 regulation, we generated a TL1A-deficient mouse and found that TL1A(-/-) dendritic cells exhibited a reduced capacity in supporting Th17 differentiation and proliferation. Consistent with these data, TL1A(-/-) animals displayed decreased clinical severity in experimental autoimmune encephalomyelitis (EAE). Finally, we demonstrated that during EAE disease progression, TL1A was required for the optimal differentiation as well as effector function of Th17 cells. These observations thus establish an important role of the TL1A-DR3 pathway in promoting Th17 cell function and Th17-mediated autoimmune disease.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Antígeno HLA-DR3/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/deficiencia , Animales , Encéfalo/inmunología , Encéfalo/patología , Diferenciación Celular , División Celular , Citocinas/metabolismo , Cartilla de ADN , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Médula Espinal/inmunología , Médula Espinal/patología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología
19.
J Immunol ; 180(12): 8327-32, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18523299

RESUMEN

The role of chicken TNF-like ligand 1A (ChTL1A) on inflammation and its receptor candidates was investigated to further understand its function as a proinflammatory cytokine. ChTL1A decreased the viability of CHO-K1 cells transfected with chicken TNFR2 or decoy receptor 3 and bound to TNFR2 and decoy receptor 3. ChTL1A was detected in chicken blood samples taken 4 h after LPS injection. Increased mRNA for inflammatory response-related factors such as IL-1beta, IL-6, ChTL1A, IFN-gamma, inducible NO synthase, and cyclooxygenase 2 were found in spleen samples following LPS injection. Ceruloplasmin and alpha(1) acid glycoprotein (as positive acute phase proteins) were increased in chicken plasma 12 h after ChTL1A injection. The injection of anti-ChTL1A Ab was able to prevent typical increases in plasma nitrite plus nitrate, ceruloplasmin, and alpha(1) acid glycoprotein concentrations following LPS injection. These results indicate that ChTL1A is a proinflammatory cytokine in chickens, animals that do not have TNF-alpha and lymphotoxin alpha orthologous genes, and that its proinflammatory action is, at least in part, expressed through binding to TNFR2.


Asunto(s)
Proteínas Aviares/fisiología , Pollos/inmunología , Mediadores de Inflamación/fisiología , Receptores Tipo II del Factor de Necrosis Tumoral/química , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Animales , Proteínas Aviares/sangre , Proteínas Aviares/genética , Proteínas Aviares/inmunología , Células CHO , Ceruloplasmina/antagonistas & inhibidores , Ceruloplasmina/metabolismo , Pollos/genética , Cricetinae , Cricetulus , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Sueros Inmunes/administración & dosificación , Mediadores de Inflamación/sangre , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/antagonistas & inhibidores , Masculino , Unión Proteica/genética , Unión Proteica/inmunología , Receptores del Factor de Necrosis Tumoral , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/sangre , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/sangre , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/fisiología
20.
Int Wound J ; 4(1): 55-64, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17425548

RESUMEN

Vascular endothelial growth inhibitor (VEGI) is an endothelial cell-specific cytokine and a potent inhibitor of endothelial cell proliferation and angiogenesis. The role of VEGI in angiogenesis related to tissue repair has previously not been investigated. Biopsies from different wound types were analysed by immunohistochemistry and quantitative polymerase chain reaction for the presence of VEGI protein and transcript, respectively. Human vascular endothelial cell line was transfected with VEGI expression plasmid and tested for their in vitro angiogenesis properties. Immunohistochemical staining for VEGI showed reduced expression in the dermal layer of the acute wounds compared with the chronic wound or normal skin. The ability of VEGI to prevent angiogenesis by in vitro assays showed that VEGI acts as a suppressor to the proliferation and microtubule formation of endothelial cells, and the addition of Hepatocyte Growth Factor had little effect on the ability of cell lines expressing the VEGI gene to increase microtubule formation. The aberrant expression of VEGI in different wound types appears to be linked to the outcome of the healing in these wounds. The altered expression of VEGI in chronic wounds constitutes an important target of future therapies.


Asunto(s)
Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/fisiología , Cicatrización de Heridas/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Línea Celular , Endotelio Vascular/citología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica/fisiología , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Piel/irrigación sanguínea , Piel/metabolismo , Piel/patología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA