Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 162(6): 1365-78, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26359988

RESUMEN

The cytokine TWEAK and its cognate receptor Fn14 are members of the TNF/TNFR superfamily and are upregulated in tumors. We found that Fn14, when expressed in tumors, causes cachexia and that antibodies against Fn14 dramatically extended lifespan by inhibiting tumor-induced weight loss although having only moderate inhibitory effects on tumor growth. Anti-Fn14 antibodies prevented tumor-induced inflammation and loss of fat and muscle mass. Fn14 signaling in the tumor, rather than host, is responsible for inducing this cachexia because tumors in Fn14- and TWEAK-deficient hosts developed cachexia that was comparable to that of wild-type mice. These results extend the role of Fn14 in wound repair and muscle development to involvement in the etiology of cachexia and indicate that Fn14 antibodies may be a promising approach to treat cachexia, thereby extending lifespan and improving quality of life for cancer patients.


Asunto(s)
Caquexia/tratamiento farmacológico , Neoplasias/patología , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Atrofia/tratamiento farmacológico , Caquexia/patología , Muerte Celular , Neoplasias del Colon/tratamiento farmacológico , Citocina TWEAK , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Desarrollo de Músculos , Neoplasias/metabolismo , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/metabolismo , Alineación de Secuencia , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33526652

RESUMEN

Identifying molecular mediators of neural circuit development and/or function that contribute to circuit dysfunction when aberrantly reengaged in neurological disorders is of high importance. The role of the TWEAK/Fn14 pathway, which was recently reported to be a microglial/neuronal axis mediating synaptic refinement in experience-dependent visual development, has not been explored in synaptic function within the mature central nervous system. By combining electrophysiological and phosphoproteomic approaches, we show that TWEAK acutely dampens basal synaptic transmission and plasticity through neuronal Fn14 and impacts the phosphorylation state of pre- and postsynaptic proteins in adult mouse hippocampal slices. Importantly, this is relevant in two models featuring synaptic deficits. Blocking TWEAK/Fn14 signaling augments synaptic function in hippocampal slices from amyloid-beta-overexpressing mice. After stroke, genetic or pharmacological inhibition of TWEAK/Fn14 signaling augments basal synaptic transmission and normalizes plasticity. Our data support a glial/neuronal axis that critically modifies synaptic physiology and pathophysiology in different contexts in the mature brain and may be a therapeutic target for improving neurophysiological outcomes.


Asunto(s)
Degeneración Nerviosa/metabolismo , Transducción de Señal , Accidente Cerebrovascular/metabolismo , Sinapsis/metabolismo , Receptor de TWEAK/metabolismo , Animales , Citocina TWEAK/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipocampo/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/fisiopatología , Plasticidad Neuronal/fisiología , Terminales Presinápticos/metabolismo , Accidente Cerebrovascular/fisiopatología , Transmisión Sináptica/fisiología
3.
Proc Natl Acad Sci U S A ; 116(16): 7941-7950, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30944222

RESUMEN

Studies over the past decade have revealed a central role for innate immune sensors in autoimmune and autoinflammatory diseases. cGAS, a cytosolic DNA sensor, detects both foreign and host DNA and generates a second-messenger cGAMP, which in turn binds and activates stimulator of IFN genes (STING), leading to induction of type I interferons and inflammatory cytokines. Recently, gain-of-function mutations in STING have been identified in patients with STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients present with early-onset systemic inflammation and interstitial lung disease, resulting in pulmonary fibrosis and respiratory failure. Here, we describe two independent SAVI mouse models, harboring the two most common mutations found in patients. A direct comparison of these strains reveals a hierarchy of immune abnormalities, lung inflammation and fibrosis, which do not depend on either IFN-α/ß receptor signaling or mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptotic cell death pathways. Furthermore, radiation chimera experiments reveal how bone marrow from the V154M mutant mice transfer disease to the WT host, whereas the N153S does not, indicating mutation-specific disease outcomes. Moreover, using radiation chimeras we find that T cell lymphopenia depends on T cell-intrinsic expression of the SAVI mutation. Collectively, these mutant mice recapitulate many of the disease features seen in SAVI patients and highlight mutation-specific functions of STING that shed light on the heterogeneity observed in SAVI patients.


Asunto(s)
Modelos Animales de Enfermedad , Interferón Tipo I/metabolismo , Enfermedades Vasculares , Animales , Muerte Celular/inmunología , Citocinas/metabolismo , Mutación con Ganancia de Función , Inflamación/inmunología , Inflamación/fisiopatología , Ratones , Enfermedades Vasculares/genética , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/fisiopatología
4.
J Proteome Res ; 20(7): 3689-3700, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34085531

RESUMEN

Novel therapies and biomarkers are needed for the treatment of acute ischemic stroke (AIS). This study aimed to provide comprehensive insights into the dynamic proteome changes and underlying molecular mechanisms post-ischemic stroke. TMT-coupled proteomic analysis was conducted on mouse brain cortex tissue from five time points up to 4 weeks poststroke in the distal hypoxic-middle cerebral artery occlusion (DH-MCAO) model. We found that nearly half of the detected proteome was altered following stroke, but only ∼8.6% of the changes were at relatively large scales. Clustering on the changed proteome defined four distinct expression patterns characterized by temporal and quantitative changes in innate and adaptive immune response pathways and cytoskeletal and neuronal remodeling. Further analysis on a subset of 309 "top hits", which temporally responded to stroke with relatively large and sustained changes, revealed that they were mostly secreted proteins, highly correlated to different cortical cytokines, and thereby potential pharmacodynamic biomarker candidates for inflammation-targeting therapies. Closer examination of the top enriched neurophysiologic pathways identified 57 proteins potentially associated with poststroke recovery. Altogether, our study generated a rich dataset with candidate proteins worthy of further validation as biomarkers and/or therapeutic targets for stroke. The proteomics data are available in the PRIDE Archive with identifier PXD025077.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Ratones , Proteoma/genética , Proteómica
5.
Am J Transplant ; 18(7): 1636-1645, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29266762

RESUMEN

Calcineurin inhibitor toxicity (CNT) is a frequent occurrence in transplanted renal grafts and autochthone kidneys from patients undergoing long-term treatment with calcineurin inhibitors, notably cyclosporin A (CsA) and tacrolimus. Here, we show an indispensable role of the tumor necrosis factor superfamily (TNFS) molecule TNF-related weak inducer of apoptosis (TWEAK) (TNFSF12) in the pathogenesis of acute CNT lesions in mice. A deficiency in TWEAK resulted in limited tubulotoxicity after CsA exposure, which correlated with diminished expression of inflammatory cytokines and reduced intraparenchymal infiltration with immune cells. We further identified tubular epithelial cells of the kidney as major targets of CsA activity and found that Fn14 (tumor necrosis factor receptor superfamily 12A), the receptor for TWEAK, is a highly CsA-inducible gene in these cells. Correlating with this, CsA pretreatment sensitized tubular epithelial cells specifically to the pro-inflammatory activities of recombinant TWEAK in vitro. Moreover, injection of rTWEAK alone into mice induced moderate disease similar to CsA, and rTWEAK combined with CsA resulted in synergistic nephrotoxicity. These findings support the importance of tubular epithelial cells as cellular targets of CsA toxicity and introduce TWEAK as a critical contributor to CNT pathogenesis.


Asunto(s)
Inhibidores de la Calcineurina/efectos adversos , Citocina TWEAK/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Túbulos Renales/patología , Receptor de TWEAK/metabolismo , Animales , Células Cultivadas , Citocina TWEAK/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor de TWEAK/genética
6.
Bioorg Med Chem Lett ; 28(10): 1964-1971, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29636220

RESUMEN

Germinal center kinase-like kinase (GLK, also known as MAP4K3) has been hypothesized to have an effect on key cellular activities, including inflammatory responses. GLK is required for activation of protein kinase C-θ (PKCθ) in T cells. Controlling the activity of T helper cell responses could be valuable for the treatment of autoimmune diseases. This approach circumvents previous unsuccessful approaches to target PKCθ directly. The use of structure based drug design, aided by the first crystal structure of GLK, led to the discovery of several inhibitors that demonstrate potent inhibition of GLK biochemically and in relevant cell lines.


Asunto(s)
Proteína Quinasa C-theta/metabolismo , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Sitios de Unión , Línea Celular , Humanos , Concentración 50 Inhibidora , Interleucina-2/metabolismo , Ratones , Ratones Noqueados , Simulación del Acoplamiento Molecular , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Estructura Terciaria de Proteína , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacología , Relación Estructura-Actividad , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
7.
J Immunol ; 197(1): 377-86, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27233964

RESUMEN

TNF-like cytokine 1A (TL1A) is expressed on APCs and provides costimulatory signals to activated lymphocytes that bear its functional receptor, death receptor 3 (DR3). TL1A/DR3 signaling is involved in the pathogenesis of human and experimental inflammatory bowel disease. In the current study, we investigated the role of this cytokine/receptor pair in acute intestinal injury/repair pathways. We demonstrate that intact DR3 signaling protected mice from acute dextran sodium sulfate colitis because DR3(-/-) mice showed more severe mucosal inflammation and increased mortality. DR3(-/-) mice were compromised in their ability to maintain adequate numbers of CD4(+)CD25(+)Foxp3(+) regulatory T cells in response to acute mucosal damage. This defect in immune regulation led to a nonspecific upregulation of effector proinflammatory pathways, which was most prominent for the Th17 immunophenotype. TL1A(-/-) mice were similarly more susceptible to dextran sodium sulfate colitis, although without mortality and with delayed kinetics compared with DR3(-/-) mice, and also displayed significantly reduced numbers of regulatory T cells. Infection of DR3(-/-) mice with Salmonella typhimurium was associated with defective microbial clearance and elevated bacterial load. Taken together, our findings indicate a novel protective role for the TL1A/DR3 axis in the regulation of mucosal homeostasis during acute intestinal injury/repair, which contrasts with its known pathogenic function during chronic intestinal inflammation.


Asunto(s)
Colitis/inmunología , Intestinos/patología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Células Cultivadas , Sulfato de Dextran , Factores de Transcripción Forkhead/metabolismo , Humanos , Intestinos/microbiología , Ratones , Ratones Noqueados , Ratones SCID , Miembro 25 de Receptores de Factores de Necrosis Tumoral/genética , Transducción de Señal/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
8.
Semin Immunol ; 26(3): 229-36, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24636536

RESUMEN

TNF-like weak inducer of apoptosis (TWEAK), a TNF family ligand, and its only known signaling receptor, FGF-inducible molecule-14 (Fn14), have emerged as a key molecular pathway regulating tissue responses after acute tissue injury and in contexts of chronic injury and disease, including autoimmunity, chronic inflammation, fibrosis, and malignancy. Usually dormant due to the low level of Fn14 expression in healthy tissues, this axis is specifically activated by the upregulation of Fn14 expression locally within injured tissues, thereby triggering a wide range of activities in tissue parenchymal and stromal cells as well as tissue progenitor cells. Current evidence supports that although transient TWEAK/Fn14 pathway activation may be beneficial for tissue repair after acute injury, excessive or sustained TWEAK/Fn14 activation due to repeated injury or chronic disease mediates significant tissue damage and pathological tissue remodeling. This paradigm for the dichotomous function of the TWEAK/Fn14 pathway is discussed, highlighting emerging findings, complexities, and implications for the treatment of tissue damage-associated pathologies and cancer.


Asunto(s)
Inflamación/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Factores de Necrosis Tumoral/metabolismo , Animales , Citocina TWEAK , Fibrosis/inmunología , Fibrosis/metabolismo , Humanos , Inflamación/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Receptor de TWEAK
9.
Hum Mol Genet ; 24(12): 3440-56, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25765661

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that primarily affects motoneurons in the brain and spinal cord. Astrocyte and microglia activation as well as skeletal muscle atrophy are also typical hallmarks of the disease. However, the functional relationship between astrocytes, microglia and skeletal muscle in the pathogenic process remains unclear. Here, we report that the tumor necrosis factor-like weak inducer of apoptosis (Tweak) and its receptor Fn14 are aberrantly expressed in spinal astrocytes and skeletal muscle of SOD1(G93A) mice. We show that Tweak induces motoneuron death, stimulates astrocytic interleukin-6 release and astrocytic proliferation in vitro. The genetic ablation of Tweak in SOD1(G93A) mice significantly reduces astrocytosis, microgliosis and ameliorates skeletal muscle atrophy. The peripheral neutralization of Tweak through antagonistic anti-Tweak antibody ameliorates muscle pathology and notably, decreases microglial activation in SOD1(G93A) mice. Unexpectedly, none of these approaches improved motor function, lifespan and motoneuron survival. Our work emphasizes the multi-systemic aspect of ALS, and suggests that a combinatorial therapy targeting multiple cell types will be instrumental to halt the neurodegenerative process.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Gliosis/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/genética , Factores de Necrosis Tumoral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Astrocitos/metabolismo , Astrocitos/patología , Muerte Celular , Proliferación Celular , Citocina TWEAK , Modelos Animales de Enfermedad , Eliminación de Gen , Regulación de la Expresión Génica , Interleucina-6/biosíntesis , Esperanza de Vida , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Placa Motora/genética , Placa Motora/metabolismo , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Mutación , Receptores de Superficie Celular/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Médula Espinal/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba
10.
Ann Rheum Dis ; 76(11): 1837-1844, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28780512

RESUMEN

OBJECTIVES: Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease associated with diffuse immune cell dysfunction. CD40-CD40 ligand (CD40L) interaction activates B cells, antigen-presenting cells and platelets. CD40L blockade might provide an innovative treatment for systemic autoimmune disorders. We investigated the safety and clinical activity of dapirolizumab pegol, a polyethylene glycol conjugated anti-CD40L Fab' fragment, in patients with SLE. METHODS: This 32-week randomised, double-blind, multicentre study (NCT01764594) evaluated repeated intravenous administration of dapirolizumab pegol in patients with SLE who were positive for/had history of antidouble stranded DNA/antinuclear antibodies and were on stable doses of immunomodulatory therapies (if applicable). Sixteen patients were randomised to 30 mg/kg dapirolizumab pegol followed by 15 mg/kg every 2 weeks for 10 weeks; eight patients received a matched placebo regimen. Randomisation was stratified by evidence of antiphospholipid antibodies. Patients were followed for 18 weeks after the final dose. RESULTS: No serious treatment-emergent adverse events, thromboembolic events or deaths occurred. Adverse events were mild or moderate, transient and resolved without intervention. One patient withdrew due to infection.Efficacy assessments were conducted only in patients with high disease activity at baseline. Five of 11 (46%) dapirolizumab pegol-treated patients achieved British Isles Lupus Assessment Group-based Composite Lupus Assessment response (vs 1/7; 14% placebo) and 5/12 (42%) evaluable for SLE Responder Index-4 responded by week 12 (vs 1/7; 14% placebo). Mechanism-related gene expression changes were observed in blood RNA samples. CONCLUSIONS: Dapirolizumab pegol could be an effective biological treatment for SLE. Further studies are required to address efficacy and safety. TRIAL REGISTRATION NUMBER: NCT01764594.


Asunto(s)
Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Factores Inmunológicos/administración & dosificación , Lupus Eritematoso Sistémico/tratamiento farmacológico , Polietilenglicoles/administración & dosificación , Transcriptoma/efectos de los fármacos , Administración Intravenosa , Adolescente , Adulto , Anciano , Ligando de CD40/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Esquema de Medicación , Femenino , Humanos , Lupus Eritematoso Sistémico/sangre , Masculino , Persona de Mediana Edad , ARN/sangre , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
11.
Nephrol Dial Transplant ; 32(9): 1468-1477, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28339802

RESUMEN

BACKGROUND: The excessive accumulation of extracellular matrix (ECM) in the renal tubulointerstitium is a key component of chronic renal damage in lupus nephritis (LN) and a critical determinant of the disease progression to renal failure. Detection of fibrosis requires renal biopsy and is therefore limited by high risks associated with an invasive procedure. This study explores whether a unique LN urinary peptidome can be identified and whether LN-specific alteration reflects the underlying fibrogenic process of altered ECM turnover. METHOD: Urinary peptides were analyzed for 36 LN and 35 nonrenal systemic lupus erythematosus (SLE) subjects and 58 healthy volunteers (HVs). RESULTS: In total, 70 collagen and 230 noncollagen peptides were significantly changed between LN and nonrenal SLE and between LN and HV and defined as 'LN peptides'; 14 proteases associated with observed LN collagen peptides were identified and activities in 9 proteases were significantly different between LN and nonrenal SLE; 28 collagen peptides were correlated with at least one parameter of clinical renal dysfunction or histolopathology. CONCLUSION: Urinary peptidomic alterations likely reflect pathogenic pathways involving ECM turnover in LN kidneys and potentially could be developed as biomarkers to monitor renal disease progression.


Asunto(s)
Biomarcadores/orina , Colágeno/orina , Enfermedades Renales/diagnóstico , Lupus Eritematoso Sistémico/complicaciones , Nefritis Lúpica/complicaciones , Fragmentos de Péptidos/orina , Adulto , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Humanos , Enfermedades Renales/etiología , Enfermedades Renales/orina , Masculino
12.
J Pathol ; 239(1): 109-21, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26924336

RESUMEN

Tumour necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) have been associated with liver regeneration in vivo. To further investigate the role of this pathway we examined their expression in human fibrotic liver disease and the effect of pathway deficiency in a murine model of liver fibrosis. The expression of Fn14 and TWEAK in normal and diseased human and mouse liver tissue and primary human hepatic stellate cells (HSCs) were investigated by qPCR, western blotting and immunohistochemistry. In addition, the levels of Fn14 in HSCs following pro-fibrogenic and pro-inflammatory stimuli were assessed and the effects of exogenous TWEAK on HSCs proliferation and activation were studied in vitro. Carbon tetrachloride (CCl4 ) was used to induce acute and chronic liver injury in TWEAK KO mice. Elevated expression of both Fn14 and TWEAK were detected in acute and chronic human liver injury, and co-localized with markers of activated HSCs. Fn14 levels were low in quiescent HSCs but were significantly induced in activated HSCs, which could be further enhanced with the profibrogenic cytokine TGFß in vitro. Stimulation with recombinant TWEAK induced proliferation but not further HSCs activation. Fn14 gene expression was also significantly up-regulated in CCl4 models of hepatic injury whereas TWEAK KO mice showed reduced levels of liver fibrosis following chronic CCl4 injury. In conclusion, TWEAK/Fn14 interaction leads to the progression of fibrotic liver disease via direct modulation of HSCs proliferation, making it a potential therapeutic target for liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/patología , Cirrosis Hepática/etiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/deficiencia , Actinas/metabolismo , Animales , Tetracloruro de Carbono/toxicidad , Proliferación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Citocina TWEAK , Progresión de la Enfermedad , Fibroblastos/metabolismo , Humanos , Cirrosis Hepática/patología , Masculino , Ratones Noqueados , ARN Mensajero/metabolismo , Factor de Transcripción SOX9/metabolismo , Receptor de TWEAK , Factores de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/farmacología , Regulación hacia Arriba/fisiología
13.
J Am Soc Nephrol ; 27(12): 3639-3652, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27026366

RESUMEN

The identification of the cellular origins of myofibroblasts has led to the discovery of novel pathways that potentially drive myofibroblast perpetuation in disease. Here, we further investigated the role of innate immune signaling pathways in this process. In mice, renal injury-induced activation of pericytes, which are myofibroblast precursors attached to endothelial cells, led to upregulated expression of TNF receptor superfamily member 12a, also known as fibroblast growth factor-inducible 14 (Fn14), by these cells. In live rat kidney slices, administration of the Fn14 ligand, TNF-related weak inducer of apoptosis (TWEAK), promoted pericyte-dependent vasoconstriction followed by pericyte detachment from capillaries. In vitro, administration of TWEAK activated and differentiated pericytes into cytokine-producing myofibroblasts, and further activated established myofibroblasts in a manner requiring canonical and noncanonical NF-κB signaling pathways. Deficiency of Fn14 protected mouse kidneys from fibrogenesis, inflammation, and associated vascular instability after in vivo injury, and was associated with loss of NF-κB signaling. In a genetic model of spontaneous CKD, therapeutic delivery of anti-TWEAK blocking antibodies attenuated disease progression, preserved organ function, and increased survival. These results identify the TWEAK-Fn14 signaling pathway as an important factor in myofibroblast perpetuation, fibrogenesis, and chronic disease progression.


Asunto(s)
Enfermedades Renales/etiología , Riñón/patología , Miofibroblastos/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal , Factores de Necrosis Tumoral/fisiología , Animales , Citocina TWEAK , Progresión de la Enfermedad , Fibrosis/etiología , Ratones , Receptor de TWEAK
14.
J Am Soc Nephrol ; 26(5): 1053-70, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25270074

RESUMEN

TNF ligand superfamily member 12, also known as TNF-related weak inducer of apoptosis (TWEAK), acts through its receptor, fibroblast growth factor-inducible 14 (Fn14), to mediate several key pathologic processes involved in tissue injury relating to lupus nephritis. To explore the potential for renal protection in lupus nephritis by targeting this pathway, we introduced the Fn14 null allele into the MRL-lpr/lpr lupus mouse strain. At 26-38 weeks of age, female Fn14-knockout MRL-lpr/lpr mice had significantly lower levels of proteinuria compared with female wild-type MRL-lpr/lpr mice. Furthermore, Fn14-knockout mice had significantly improved renal histopathology accompanied by attenuated glomerular and tubulointerstitial inflammation. There was a significant reduction in glomerular Ig deposition in Fn14-knockout mice, despite no detectable differences in either serum levels of antibodies or splenic immune cell subsets. Notably, we found that the Fn14-knockout mice displayed substantial preservation of podocytes in glomeruli and that TWEAK signaling directly damaged barrier function and increased filtration through podocyte and glomerular endothelial cell monolayers. Our results show that deficiency of the Fn14 receptor significantly improves renal disease in a spontaneous lupus nephritis model through prevention of the direct injurious effects of TWEAK on the filtration barrier and/or modulation of cytokine production by resident kidney cells. Thus, blocking the TWEAK/Fn14 axis may be a novel therapeutic intervention in immune-mediated proliferative GN.


Asunto(s)
Factores de Crecimiento de Fibroblastos/deficiencia , Barrera de Filtración Glomerular/metabolismo , Nefritis Lúpica/etiología , Factores de Necrosis Tumoral/metabolismo , Animales , Citocina TWEAK , Femenino , Inmunoglobulina G/metabolismo , Ratones Noqueados , Proteinuria/metabolismo
15.
Am J Physiol Gastrointest Liver Physiol ; 308(4): G325-34, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25524063

RESUMEN

TNF-like weak inducer of apoptosis (TWEAK) is a growth factor for bipotent liver progenitors that express its receptor, fibroblast growth factor-inducible 14 (Fn14), a TNF receptor superfamily member. Accumulation of Fn14(+) progenitors occurs in severe acute alcoholic steatohepatitis (ASH) and correlates with acute mortality. In patients with severe ASH, inhibition of TNF-α increases acute mortality. The aim of this study was to determine whether deletion of Fn14 improves the outcome of liver injury in alcohol-consuming mice. Wild-type (WT) and Fn14 knockout (KO) mice were fed control high-fat Lieber deCarli diet or high-fat Lieber deCarli diet with 2% alcohol (ETOH) and injected intraperitoneally with CCl4 for 2 wk to induce liver injury. Mice were euthanized 3 or 10 days after CCl4 treatment. Survival was assessed. Liver tissues were analyzed for cell death, inflammation, proliferation, progenitor accumulation, and fibrosis by quantitative RT-PCR, immunoblot, hydroxyproline content, and quantitative immunohistochemistry. During liver injury, Fn14 expression, apoptosis, inflammation, hepatocyte replication, progenitor and myofibroblast accumulation, and fibrosis increased in WT mice fed either diet. Mice fed either diet expressed similar TWEAK/Fn14 levels, but ETOH-fed mice had higher TNF-α expression. The ETOH-fed group developed more apoptosis, inflammation, fibrosis, and regenerative responses. Fn14 deletion did not reduce hepatic TNF-α expression but improved all injury parameters in mice fed the control diet. In ETOH-fed mice, Fn14 deletion inhibited TNF-α induction and increased acute mortality, despite improvement in liver injury. Fn14 mediates wound-healing responses that are necessary to survive acute liver injury during alcohol exposure.


Asunto(s)
Hígado Graso Alcohólico/metabolismo , Hígado/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Enfermedad Aguda , Animales , Apoptosis , Tetracloruro de Carbono , Proliferación Celular , Modelos Animales de Enfermedad , Etanol , Hígado Graso Alcohólico/etiología , Hígado Graso Alcohólico/genética , Hígado Graso Alcohólico/patología , Hidroxiprolina/metabolismo , Mediadores de Inflamación/metabolismo , Hígado/patología , Cirrosis Hepática Alcohólica/etiología , Cirrosis Hepática Alcohólica/metabolismo , Cirrosis Hepática Alcohólica/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal , Receptor de TWEAK , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Cicatrización de Heridas
16.
J Autoimmun ; 60: 40-50, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25911200

RESUMEN

Neuropsychiatric disease is one of the most common manifestations of human systemic lupus erythematosus, but the mechanisms remain poorly understood. In human brain microvascular endothelial cells in vitro, TNF-like weak inducer of apoptosis (TWEAK) decreases tight junction ZO-1 expression and increases the permeability of monolayer cell cultures. Furthermore, knockout (KO) of the TWEAK receptor, Fn14, in the MRL/lpr lupus mouse strain markedly attenuates neuropsychiatric disease, as demonstrated by significant reductions in depressive-like behavior and improved cognitive function. The purpose of the present study was to determine the mechanisms by which TWEAK signaling is instrumental in the pathogenesis of neuropsychiatric lupus (NPSLE). Evaluating brain sections of MRL/lpr Fn14WT and Fn14KO mice, we found that Fn14KO mice displayed significantly decreased cellular infiltrates in the choroid plexus. To evaluate the integrity of the blood brain barrier (BBB) in MRL/lpr mice, Western blot for fibronectin, qPCR for iNOS, and immunohistochemical staining for VCAM-1/ICAM-1 were performed. We found preserved BBB permeability in MRL/lpr Fn14KO mice, attributable to reduced brain expression of VCAM-1/ICAM-1 and iNOS. Additionally, administration of Fc-TWEAK intravenously directly increased the leakage of a tracer (dextran-FITC) into brain tissue. Furthermore, MRL/lpr Fn14KO mice displayed reduced antibody (IgG) and complement (C3, C6, and C4a) deposition in the brain. Finally, we found that MRL/lpr Fn14KO mice manifested reduced neuron degeneration and hippocampal gliosis. Our studies indicate that TWEAK/Fn14 interactions play an important role in the pathogenesis of NPSLE by increasing the accumulation of inflammatory cells in the choroid plexus, disrupting BBB integrity, and increasing neuronal damage, suggesting a novel target for therapy in this disease.


Asunto(s)
Apoptosis/genética , Barrera Hematoencefálica/fisiopatología , Neuronas/patología , Receptores del Factor de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis/inmunología , Plexo Coroideo/fisiopatología , Cognición , Complemento C3/inmunología , Complemento C4a/inmunología , Complemento C6/inmunología , Citocina TWEAK , Depresión/genética , Modelos Animales de Enfermedad , Gliosis/genética , Inmunoglobulina G/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Noqueados , Degeneración Nerviosa/genética , Permeabilidad , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor de TWEAK , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de la Zonula Occludens-1/biosíntesis
17.
FASEB J ; 28(3): 1398-411, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24327607

RESUMEN

Skeletal muscle wasting attributed to inactivity has significant adverse functional consequences. Accumulating evidence suggests that peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and TNF-like weak inducer of apoptosis (TWEAK)-Fn14 system are key regulators of skeletal muscle mass in various catabolic states. While the activation of TWEAK-Fn14 signaling causes muscle wasting, PGC-1α preserves muscle mass in several conditions, including functional denervation and aging. However, it remains unknown whether there is any regulatory interaction between PGC-1α and TWEAK-Fn14 system during muscle atrophy. Here we demonstrate that TWEAK significantly reduces the levels of PGC-1α and mitochondrial content (∼50%) in skeletal muscle. Levels of PGC-1α are significantly increased in skeletal muscle of TWEAK-knockout (KO) and Fn14-KO mice compared to wild-type mice on denervation. Transgenic (Tg) overexpression of PGC-1α inhibited progressive muscle wasting in TWEAK-Tg mice. PGC-1α inhibited the TWEAK-induced activation of NF-κB (∼50%) and dramatically reduced (∼90%) the expression of atrogenes such as MAFbx and MuRF1. Intriguingly, muscle-specific overexpression of PGC-1α also prevented the inducible expression of Fn14 in denervated skeletal muscle. Collectively, our study demonstrates that TWEAK induces muscle atrophy through repressing the levels of PGC-1α. Overexpression of PGC-1α not only blocks the TWEAK-induced atrophy program but also diminishes the expression of Fn14 in denervated skeletal muscle.


Asunto(s)
Músculo Esquelético/patología , Atrofia Muscular/patología , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Transcripción/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Citocina TWEAK , Ensayo de Cambio de Movilidad Electroforética , Ratones , Ratones Transgénicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Receptor de TWEAK
18.
Immunol Rev ; 244(1): 99-114, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22017434

RESUMEN

Our immune system performs the vital function of recognizing and eliminating invading pathogens and malignancies. There is an increasing appreciation that the immune system also actively mediates tissue responses under both physiological and pathological conditions, significantly impacting the inflammatory, fibrogenic, and regenerative components. Likewise, there is a growing understanding of how epithelial, endothelial, and other non-hematopoietic tissue cell types actively contribute to the interplay that shapes tissue responses. While much of the molecular basis underlying the immune regulation of tissue responses remains to be delineated, the tumor necrosis factor (TNF) superfamily ligand/receptor pair of TNF-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor-inducible molecule 14 (Fn14) has now emerged as a key piece of this puzzle. In this review, we first discuss how the usually 'dormant' TWEAK/Fn14 pathway becomes activated specifically in injury and disease contexts. We then summarize how TWEAK-mediated Fn14 signaling triggers a wide range of activities in tissue parenchymal and stromal cells as well as progenitor cells. Finally, we review recent experimental evidence that further supports the functional dichotomy of TWEAK/Fn14 activation in physiological versus pathological tissue responses and its potential therapeutic implications. Whereas transient TWEAK/Fn14 activation promotes productive tissue responses after injury, excessive or persistent TWEAK/Fn14 activation drives pathological tissue responses, leading to progressive damage and degeneration.


Asunto(s)
Inmunidad Innata , Inflamación/inmunología , Músculo Esquelético/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Células Madre/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis/inmunología , Citocina TWEAK , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Expresión Génica/inmunología , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/metabolismo , Receptor de TWEAK , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/metabolismo
19.
J Cell Mol Med ; 18(4): 721-34, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24479820

RESUMEN

Clinical complications associated with atherosclerotic plaques arise from luminal obstruction due to plaque growth or destabilization leading to rupture. Tumour necrosis factor ligand superfamily member 12 (TNFSF12) also known as TNF-related weak inducer of apoptosis (TWEAK) is a proinflammatory cytokine that participates in atherosclerotic plaque development, but its role in plaque stability remains unclear. Using two different approaches, genetic deletion of TNFSF12 and treatment with a TWEAK blocking mAb in atherosclerosis-prone mice, we have analysed the effect of TWEAK inhibition on atherosclerotic plaques progression and stability. Mice lacking both TNFSF12 and Apolipoprotein E (TNFSF12(-/-) ApoE(-/-) ) exhibited a diminished atherosclerotic burden and lesion size in their aorta. Advanced atherosclerotic plaques of TNFSF12(-/-) ApoE(-/-) or anti-TWEAK treated mice exhibited an increase collagen/lipid and vascular smooth muscle cell/macrophage ratios compared with TNFSF12(+/+) ApoE(-/-) control mice, reflecting a more stable plaque phenotype. These changes are related with two different mechanisms, reduction of the inflammatory response (chemokines expression and secretion and nuclear factor kappa B activation) and decrease of metalloproteinase activity in atherosclerotic plaques of TNFSF12(-/-) ApoE(-/-) . A similar phenotype was observed with anti-TWEAK mAb treatment in TNFSF12(+/+) ApoE(-/-) mice. Brachiocephalic arteries were also examined since they exhibit additional features akin to human atherosclerotic plaques associated with instability and rupture. Features of greater plaque stability including augmented collagen/lipid ratio, reduced macrophage content, and less presence of lateral xanthomas, buried caps, medial erosion, intraplaque haemorrhage and calcium content were present in TNFSF12(-/-) ApoE(-/-) or anti-TWEAK treatment in TNFSF12(+/+) ApoE(-/-) mice. Overall, our data indicate that anti-TWEAK treatment has the capacity to diminish proinflamatory response associated with atherosclerotic plaque progression and to alter plaque morphology towards a stable phenotype.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Aterosclerosis/genética , Placa Aterosclerótica/genética , Factores de Necrosis Tumoral/genética , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Apoptosis/genética , Apoptosis/inmunología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Aterosclerosis/patología , Citocina TWEAK , Humanos , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/patología , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/patología , Inhibidores del Factor de Necrosis Tumoral , Factores de Necrosis Tumoral/inmunología
20.
Biochim Biophys Acta ; 1832(10): 1744-55, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23748045

RESUMEN

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) regulates apoptosis, proliferation and inflammation in renal epithelial cells and plays a role in acute kidney injury. However, there is little information on the chronic effects of TWEAK. We hypothesized that TWEAK may influence renal fibrosis and regulate kidney fibroblast biology, in part, through Ras pathway. We studied a chronic model of experimental unilateral ureteral obstruction in wild type and TWEAK deficient mice, and a murine model of systemic TWEAK overexpression. TWEAK actions were also explored in cultured renal and embryonic fibroblasts. TWEAK and TWEAK receptor expression was increased in the obstructed kidneys. The absence of TWEAK decreased early kidney tubular damage, inflammatory infiltrates and myofibroblast number. TWEAK deficient mice had decreased renal fibrosis 21days after obstruction, as assessed by extracellular matrix staining. In mice without prior underlying kidney disease, systemic overexpression of TWEAK induced kidney inflammation and fibrosis. In cultured fibroblasts, TWEAK induced proliferation through activation of the Ras/ERK pathway. TWEAK also activated nuclear factor κB (NFκB)-dependent inflammatory chemokine production in murine renal fibroblasts. In conclusion, lack of TWEAK reduces renal fibrosis in a model of persistent kidney insult and overexpression of TWEAK led to renal fibrosis. TWEAK actions on renal fibroblasts may contribute to the in vivo observations, as TWEAK promotes inflammatory activity and proliferation in fibroblast cultures.


Asunto(s)
Proliferación Celular , Fibrosis/fisiopatología , Enfermedades Renales/fisiopatología , Riñón/patología , Factores de Necrosis Tumoral/fisiología , Proteínas ras/fisiología , Animales , Línea Celular , Citocina TWEAK , Fibroblastos/patología , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA