Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Immunol ; 197(10): 3806-3819, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27815440

RESUMEN

Despite being one of the most common rheumatologic diseases, there is still no disease-modifying drug for primary Sjögren's syndrome (pSS). Advancing our knowledge of the target tissue has been limited by the low dimensionality of histology techniques and the small size of human salivary gland biopsies. In this study, we took advantage of a molecularly validated mouse model of pSS to characterize tissue-infiltrating CD4+ T cells and their regulation by the lymphotoxin/LIGHT signaling axis. Novel cell subsets were identified by combining highly dimensional flow and mass cytometry with transcriptomic analyses. Pharmacologic modulation of the LTßR signaling pathway was achieved by treating mice with LTßR-Ig, a therapeutic intervention currently being tested in pSS patients (Baminercept trial NCT01552681). Using these approaches, we identified two novel CD4+ T cell subsets characterized by high levels of PD1: Prdm1+ effector regulatory T cells expressing immunoregulatory factors, such as Il10, Areg, Fgl2, and Itgb8, and Il21+ effector conventional T cells expressing a pathogenic transcriptional signature. Mirroring these observations in mice, large numbers of CD4+PD1+ T cells were detected in salivary glands from Sjögren's patients but not in normal salivary glands or kidney biopsies from lupus nephritis patients. Unexpectedly, LTßR-Ig selectively halted the recruitment of PD1- naive, but not PD1+, effector T cells to the target tissue, leaving the cells with pathogenic potential unaffected. Altogether, this study revealed new cellular players in pSS pathogenesis, their transcriptional signatures, and differential dependency on the lymphotoxin/LIGHT signaling axis that help to interpret the negative results of the Baminercept trial and will guide future therapeutic interventions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Receptor beta de Linfotoxina/metabolismo , Linfotoxina-alfa/metabolismo , Glándulas Salivales/inmunología , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/fisiopatología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Anfirregulina/genética , Animales , Biopsia , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Interleucina-10/genética , Interleucinas/genética , Riñón/patología , Nefritis Lúpica/inmunología , Linfotoxina-alfa/genética , Ratones , Glándulas Salivales/patología , Transducción de Señal , Síndrome de Sjögren/terapia , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores
2.
J Biol Chem ; 291(38): 19826-34, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27451394

RESUMEN

B cell activating factor of the TNF family (BAFF), also known as B lymphocyte stimulator, is a ligand required for the generation and maintenance of B lymphocytes. In this study, the ability of different monoclonal antibodies to recognize, inhibit, or activate mouse BAFF was investigated. One of them, a mouse IgG1 named Sandy-2, prevented the binding of BAFF to all of its receptors, BAFF receptor, transmembrane activator and calcium modulating ligand interactor, and B cell maturation antigen, at a stoichiometric ratio; blocked the activity of mouse BAFF on a variety of cell-based reporter assays; and antagonized the prosurvival action of BAFF on primary mouse B cells in vitro A single administration of Sandy-2 in mice induced B cell depletion within 2 weeks, down to levels close to those observed in BAFF-deficient mice. This depletion could then be maintained with a chronic treatment. Sandy-2 and a previously described rat IgG1 antibody, 5A8, also formed a pair suitable for the sensitive detection of endogenous circulating BAFF by ELISA or using a homogenous assay. Interestingly, 5A8 and Sandy-5 displayed activities opposite to that of Sandy-2 by stimulating recombinant BAFF in vitro and endogenous BAFF in vivo These tools will prove useful for the detection and functional manipulation of endogenous mouse BAFF and provide an alternative to the widely used BAFF receptor-Fc decoy receptor for the specific depletion of BAFF in mice.


Asunto(s)
Anticuerpos/farmacología , Factor Activador de Células B/antagonistas & inhibidores , Linfocitos B/inmunología , Inmunoglobulina G/farmacología , Animales , Anticuerpos/inmunología , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Linfocitos B/patología , Supervivencia Celular/efectos de los fármacos , Hiperplasia , Inmunoglobulina G/inmunología , Depleción Linfocítica/métodos , Ratones , Ratones Noqueados
3.
Hum Mol Genet ; 24(7): 2035-48, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25504044

RESUMEN

Myotonic dystrophy type 1 (DM1), the most prevalent muscular dystrophy in adults, is characterized by progressive muscle wasting and multi-systemic complications. DM1 is the prototype for disorders caused by RNA toxicity. Currently, no therapies exist. Here, we identify that fibroblast growth factor-inducible 14 (Fn14), a member of the tumor necrosis factor receptor super-family, is induced in skeletal muscles and hearts of mouse models of RNA toxicity and in tissues from DM1 patients, and that its expression correlates with severity of muscle pathology. This is associated with downstream signaling through the NF-κB pathways. In mice with RNA toxicity, genetic deletion of Fn14 results in reduced muscle pathology and better function. Importantly, blocking TWEAK/Fn14 signaling with an anti-TWEAK antibody likewise improves muscle histopathology and functional outcomes in affected mice. These results reveal new avenues for therapeutic development and provide proof of concept for a novel therapeutic target for which clinically available therapy exists to potentially treat muscular dystrophy in DM1.


Asunto(s)
Distrofia Miotónica/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Adulto , Animales , Anticuerpos/administración & dosificación , Citocina TWEAK , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal/efectos de los fármacos , Receptor de TWEAK , Inhibidores del Factor de Necrosis Tumoral , Factores de Necrosis Tumoral/genética
4.
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
5.
J Biol Chem ; 290(26): 16330-42, 2015 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-25953898

RESUMEN

The closely related TNF family ligands B cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL) serve in the generation and maintenance of mature B-lymphocytes. Both BAFF and APRIL assemble as homotrimers that bind and activate several receptors that they partially share. However, heteromers of BAFF and APRIL that occur in patients with autoimmune diseases are incompletely characterized. The N and C termini of adjacent BAFF or APRIL monomers are spatially close and can be linked to create single-chain homo- or hetero-ligands of defined stoichiometry. Similar to APRIL, heteromers consisting of one BAFF and two APRILs (BAA) bind to the receptors B cell maturation antigen (BCMA), transmembrane activator and CAML interactor (TACI) but not to the BAFF receptor (BAFFR). Heteromers consisting of one APRIL and two BAFF (ABB) bind to TACI and BCMA and weakly to BAFFR in accordance with the analysis of the receptor interaction sites in the crystallographic structure of ABB. Receptor binding correlated with activity in reporter cell line assays specific for BAFFR, TACI, or BCMA. Single-chain BAFF (BBB) and to a lesser extent single-chain ABB, but not APRIL or single-chain BAA, rescued BAFFR-dependent B cell maturation in BAFF-deficient mice. In conclusion, BAFF-APRIL heteromers of different stoichiometries have distinct receptor-binding properties and activities. Based on the observation that heteromers are less active than BAFF, we speculate that their physiological role might be to down-regulate BAFF activity.


Asunto(s)
Factor Activador de Células B/metabolismo , Antígeno de Maduración de Linfocitos B/metabolismo , Proteína Activadora Transmembrana y Interactiva del CAML/metabolismo , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Factor Activador de Células B/química , Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/genética , Receptor del Factor Activador de Células B/metabolismo , Antígeno de Maduración de Linfocitos B/genética , Dimerización , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Unión Proteica , Transducción de Señal , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/química , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
6.
Clin Immunol ; 169: 69-79, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27352977

RESUMEN

B-cell activating factor (BAFF) levels are increased in rheumatoid arthritis, lupus and primary Sjögren's syndrome (pSS). However, BAFF contribution to pathogenesis is not completely understood. In pSS, immune infiltration of the salivary and lacrimal glands leads to xerostomia and xerophtalmia. Glandular B cell hyperactivation, differentiation into germinal center (GC)-like structures and plasma cell accumulation are histopathological hallmarks that were attributed to increased BAFF. Here, we experimentally tested this hypothesis by overexpressing BAFF in a mouse model of pSS. BAFF overexpression enhanced lymphocytic infiltration and MHCII expression on B cells. Increased BAFF also induced B cell differentiation into GC B cells within the autoimmune target tissue. However, even in these conditions, GC B cells only accounted for <1% of glandular B cells, demonstrating that BAFF is not efficiently promoting ectopic GC formation in pSS and warranting further investigation of therapeutics targeting both BAFF and the related TNF-family member APRIL.


Asunto(s)
Factor Activador de Células B/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Síndrome de Sjögren/inmunología , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Linfocitos B/metabolismo , Linfocitos B/patología , Diferenciación Celular/genética , Células Cultivadas , Citometría de Flujo , Perfilación de la Expresión Génica/métodos , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inmunohistoquímica , Aparato Lagrimal/inmunología , Aparato Lagrimal/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Análisis de Secuencia por Matrices de Oligonucleótidos , Síndrome de Sjögren/genética , Síndrome de Sjögren/metabolismo , Xeroftalmia/genética , Xeroftalmia/inmunología , Xeroftalmia/metabolismo , Xerostomía/genética , Xerostomía/inmunología , Xerostomía/metabolismo
7.
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
8.
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
9.
Biochem Biophys Res Commun ; 446(4): 1219-1224, 2014 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-24680686

RESUMEN

Progressive loss of skeletal muscle mass and strength (sarcopenia) is a major clinical problem in the elderly. Recently, proinflammatory cytokine TWEAK and its receptor Fn14 were identified as key mediators of muscle wasting in various catabolic states. However, the role of the TWEAK-Fn14 pathway in pathological changes in skeletal muscle during aging remains unknown. In this study, we demonstrate that the levels of Fn14 are increased in skeletal muscle of 18-month old (aged) mice compared with adult mice. Genetic ablation of Fn14 significantly increased the levels of specific muscle proteins and blunted the age-associated fiber atrophy in mice. While gene expression of two prominent muscle-specific E3 ubiquitin ligases MAFBx and MuRF1 remained comparable, levels of ubiquitinated proteins and the expression of autophagy-related molecule Atg12 were significantly reduced in Fn14-knockout (KO) mice compared with wild-type mice during aging. Ablation of Fn14 significantly diminished the DNA-binding activity of transcription factor nuclear factor-kappa B (NF-κB), gene expression of various inflammatory molecules, and interstitial fibrosis in skeletal muscle of aged mice. Collectively, our study suggests that the TWEAK-Fn14 signaling axis contributes to age-associated muscle atrophy and fibrosis potentially through its local activation of proteolytic systems and inflammatory pathways.


Asunto(s)
Envejecimiento , Músculo Esquelético/patología , Atrofia Muscular/patología , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Colágeno/análisis , Colágeno/metabolismo , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Eliminación de Gen , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , FN-kappa B/metabolismo , Proteolisis , Transducción de Señal , Receptor de TWEAK
10.
Basic Res Cardiol ; 108(2): 325, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23325387

RESUMEN

Pulmonary arterial hypertension (PAH) is a fatal disease for which no cure is yet available. The leading cause of death in PAH is right ventricular (RV) failure. Previously, the TNF receptor superfamily member fibroblast growth factor-inducible molecule 14 (Fn14) has been associated with different fibrotic diseases. However, so far there is no study demonstrating a causal role for endogenous Fn14 signaling in RV or LV heart disease. The purpose of this study was to determine whether global ablation of Fn14 prevents RV fibrosis and remodeling improving heart function. Here, we provide evidence for a causative role of Fn14 in pulmonary artery banding (PAB)-induced RV fibrosis and dysfunction in mice. Fn14 expression was increased in the RV after PAB. Mice lacking Fn14 (Fn14(-/-)) displayed substantially reduced RV fibrosis and dysfunction following PAB compared to wild-type littermates. Cell culture experiments demonstrated that activation of Fn14 induces collagen expression via RhoA-dependent nuclear translocation of myocardin-related transcription factor-A (MRTF-A)/MAL. Furthermore, activation of Fn14 in vitro caused fibroblast proliferation and myofibroblast differentiation, which corresponds to suppression of PAB-induced RV fibrosis in Fn14(-/-) mice. Moreover, our findings suggest that Fn14 expression is regulated by endothelin-1 (ET-1) in cardiac fibroblasts. We conclude that Fn14 is an endogenous key regulator in cardiac fibrosis and suggest this receptor as potential new target for therapeutic interventions in heart failure.


Asunto(s)
Hipertrofia Ventricular Derecha/prevención & control , Miocardio/patología , Receptores del Factor de Necrosis Tumoral/fisiología , Disfunción Ventricular Derecha/prevención & control , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Western Blotting , Diferenciación Celular , Proliferación Celular , Colágeno/metabolismo , Citocina TWEAK , Endotelina-1/fisiología , Hipertensión Pulmonar Primaria Familiar , Fibrosis/prevención & control , Técnica del Anticuerpo Fluorescente , Hipertensión Pulmonar/complicaciones , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Inmunohistoquímica , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Miofibroblastos , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Receptor de TWEAK , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba , Disfunción Ventricular Derecha/metabolismo , Disfunción Ventricular Derecha/fisiopatología
11.
Mol Pharmacol ; 82(5): 948-57, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22909796

RESUMEN

Reactive astrogliosis is beneficial in many aspects; however, it is also detrimental in some pathological states such as the development of lethal brain tumors. It is therefore crucial to understand the mechanisms regulating astrocyte proliferation. Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor family, was shown to stimulate astrocyte proliferation in vitro. Herein, we further characterize the mitogenic potential of TWEAK on central nervous system cells. Among these cells, astrocytes express the highest level of TWEAK and Fn14 transcripts, suggesting that they are particularly sensitive to TWEAK stimulation. Using in vitro model systems, we found that TWEAK was as potent as epidermal growth factor (EGF) (a prototypical astrocyte mitogen) in mediating astrocyte proliferation. However, its mitogenic activity was delayed compared with that of EGF, suggesting distinct mechanisms of action. Using cell signaling pathway inhibitors, neutralizing antibodies, and protein assays, we further show that the mitogenic activity of TWEAK on primary astrocytes requires stimulation of the transforming growth factor-α (TGF-α) and of the epidermal growth factor receptor (EGFR) signaling pathway through extracellular signal-regulated kinase and p38 mitogen-activated protein kinase activation. In aggregates, our data demonstrate that TWEAK acts as a potent astrocyte mitogen through the induction of a TGF-α/EGFR signaling pathway. We anticipate that description of such a mechanism may allow novel approaches to human pathologies associated with astrocyte proliferation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Astrocitos/citología , Receptores ErbB/fisiología , Proteínas de la Membrana/metabolismo , Factor de Crecimiento Transformador alfa/fisiología , Factores de Necrosis Tumoral/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/farmacología , Astrocitos/metabolismo , Proliferación Celular , Citocina TWEAK , Embrión de Mamíferos , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Proteínas de la Membrana/farmacología , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Cultivo Primario de Células , Ratas , Ratas Wistar , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Circulation ; 119(15): 2058-68, 2009 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-19349318

RESUMEN

BACKGROUND: Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor superfamily, is a multifunctional cytokine known to regulate cellular functions in contexts of injury and disease through its receptor, fibroblast growth factor-inducible molecule 14 (Fn14). Although many of the processes and downstream signals regulated by the TWEAK/Fn14 pathway have been implicated in the development of cardiac dysfunction, the role of TWEAK in the cardiovascular system is completely unknown. METHODS AND RESULTS: Herein, we demonstrate that mouse and human cardiomyocytes express the TWEAK receptor Fn14. Furthermore, we determine that elevated circulating levels of TWEAK, induced via transgenic or adenoviral-mediated gene expression in mice, result in dilated cardiomyopathy with subsequent severe cardiac dysfunction. This phenotype was mediated exclusively by the Fn14 receptor, independent of tumor necrosis factor-alpha, and was associated with cardiomyocyte elongation and cardiac fibrosis but not cardiomyocyte apoptosis. Moreover, we find that circulating TWEAK levels were differentially upregulated in patients with idiopathic dilated cardiomyopathy compared with other forms of heart disease and normal control subjects. CONCLUSIONS: Our data suggest that TWEAK/Fn14 may be important in regulating myocardial structural remodeling and function and may play a role in the pathogenesis of dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Apoptosis , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Tamaño de la Célula , Enfermedad Coronaria/metabolismo , Enfermedad Coronaria/patología , Citocina TWEAK , Femenino , Fibrosis , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Humanos , Hipertensión/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Persona de Mediana Edad , Fenotipo , Proteínas Recombinantes de Fusión/fisiología , Receptor de TWEAK , Transducción Genética , Factores de Necrosis Tumoral/sangre , Factores de Necrosis Tumoral/genética
13.
Arterioscler Thromb Vasc Biol ; 29(12): 2021-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19762780

RESUMEN

OBJECTIVE: TWEAK is a multifunctional cytokine belonging to the tumor necrosis factor superfamily and binds to the receptor Fn14. TWEAK and Fn14 are expressed in atherosclerotic plaques in areas rich in macrophages and foam cells. We investigated the role of TWEAK/Fn14 interactions in ApoE(-/-) mice and bone marrow-derived macrophages in vitro. METHODS AND RESULTS: ApoE(-/-) mice were treated with TWEAK-inhibiting fusion protein, Fn14-Fc, in an early (5 to 17 weeks of age) or delayed (17 to 29 weeks of age) setting. In the aortic arch, Fn14-Fc as compared to control treatment resulted in advanced plaques which were smaller (early treatment), fewer (delayed treatment), lower in fibrotic content (early and delayed treatment), and exhibited an increased macrophage content and smaller macrophage size (delayed treatment). There were no differences in apoptosis in atherosclerotic plaques after Fn14-Fc versus control Ab treatment. However, blocking TWEAK resulted in less macrophage uptake of modified lipids in vitro. CONCLUSIONS: Fn14-Fc fusion protein treatment did not prevent lesion initiation but inhibited some features of plaque progression and induced a unique advanced plaque phenotype with increased macrophage content and smaller macrophage size, which may be attributable to reduced lipid uptake. These findings indicate that TWEAK/Fn14 interactions regulate atherosclerosis and mediate lipid uptake in macrophages.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/etiología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Inhibidores del Factor de Necrosis Tumoral , Animales , Apolipoproteínas E/genética , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Transporte Biológico Activo/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Citocina TWEAK , Citocinas/biosíntesis , Técnicas In Vitro , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Ratones Noqueados , Proteínas Recombinantes de Fusión/farmacología , Receptor de TWEAK , Factores de Necrosis Tumoral/fisiología
14.
J Immunol ; 181(10): 6810-9, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18981099

RESUMEN

Caspase (Casp) family proteases regulate not only lymphocyte apoptosis but also lymphocyte activation and development. In this study, we show that Casp6 regulates B cell activation and differentiation into plasma cells by modifying cell cycle entry. B cells from Casp6 knockout (Casp6 KO) mice examined ex vivo have more cells in G(1) than wild-type B cells, and mitogen-induced G(1) entry of Casp6 KO B cells is much faster than that of wild-type B cells. Even so, S phase entry and proliferation are not increased in Casp6 KO B cells. Rather than proliferating, activated Casp6 KO B cells preferentially differentiate into syndecan-1(+) plasma cells and produce Abs. In Casp6 KO mice compared with WT mice, serum levels of IgG1, IgG2a, and IgG2b are increased and Ag-specific Ab responses are also enhanced along with increased percentages of syndecan-1(+) plasma cells. Casp6 may regulate both B cell activation and differentiation by modifying requirements for G(0) B cells to enter G(1).


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Caspasa 6/metabolismo , Activación de Linfocitos/inmunología , Células Plasmáticas/citología , Células Plasmáticas/inmunología , Animales , Linfocitos B/metabolismo , Western Blotting , Caspasa 6/inmunología , Ciclo Celular/inmunología , Diferenciación Celular/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Ratones , Ratones Noqueados , Células Plasmáticas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Leukoc Biol ; 84(2): 338-47, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18483204

RESUMEN

Growing evidence suggests that the tumor necrosis factor superfamily (TNFSF) member TWEAK and its cognate receptor Fn14 play an important role in both physiological and pathological tissue remodeling. Herein, we review the various lines of experimental evidence that support the involvement of this ligand/receptor pair in triggering a wide range of cellular responses crucial to tissue remodeling, including angiogenic, proliferative, and inflammatory responses, and discuss the molecular mechanisms by which TWEAK/Fn14-induced tissue responses can lead to desired vs. undesired consequences in a context-dependent manner. We explore the key features of TWEAK-induced end-organ pathologies in various autoimmune disorders and the potential therapeutic benefits of TWEAK inhibition therein. We submit the viewpoint that TWEAK/Fn14-mediated pathogenic tissue remodeling represents an important, universal mechanism leading to various end-organ pathologies associated with autoimmune and inflammatory disorders. The highly specific and localized nature of its pathogenic contribution, therefore, makes the TWEAK/Fn14 pathway a unique and promising therapeutic target.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Necrosis Tumoral/fisiología , Citocina TWEAK , Humanos , Hiperplasia , Inflamación/fisiopatología , Neovascularización Patológica , Neovascularización Fisiológica , Regeneración/fisiología , Células Madre/fisiología , Receptor de TWEAK , Cicatrización de Heridas/fisiología
16.
Sci Rep ; 9(1): 4521, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30872777

RESUMEN

Systemic sclerosis (SSc or scleroderma) is an auto-immune disease characterized by skin fibrosis. While primary cells from patients are considered as a unique resource to better understand human disease biology, the effect of in vitro culture on these cells and their evaluation as a platform to identify disease regulators remain poorly characterized. The goal of our studies was to provide insights into the utility of SSc dermal fibroblast primary cells for therapeutic target discovery. The disease phenotypes of freshly isolated and in vitro cultured SSc dermal fibroblasts were characterized using whole transcriptome profiling, alpha smooth muscle actin (ASMA) expression and cell impedance. SSc dermal fibroblasts retained most of the molecular disease phenotype upon in vitro culture for at least four cell culture passages (approximatively 10 cell doublings). We validated an RNA interference high throughput assay that successfully identified genes affecting the myofibroblast phenotype of SSc skin fibroblasts. These genes included MKL1, RHOA and LOXL2 that were previously proposed as therapeutic anti-fibrotic target, and ITGA5, that has been less studied in fibrosis biology and may be a novel potential modifier of SSc fibroblast biology. Together our results demonstrated the value of carefully-phenotyped SSc dermal fibroblasts as a platform for SSc target and drug discovery.


Asunto(s)
Fibroblastos/metabolismo , Esclerodermia Sistémica/patología , Actinas/antagonistas & inhibidores , Actinas/genética , Actinas/metabolismo , Adulto , Anciano de 80 o más Años , Estudios de Casos y Controles , Células Cultivadas , Femenino , Fibroblastos/citología , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Análisis de Componente Principal , ARN Interferente Pequeño/metabolismo , Esclerodermia Sistémica/metabolismo , Índice de Severidad de la Enfermedad , Transactivadores/antagonistas & inhibidores , Transactivadores/metabolismo , Transcriptoma
17.
J Neuropathol Exp Neurol ; 67(12): 1137-48, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19018248

RESUMEN

The expression patterns of tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a pleiotropic cytokine with proinflammatory and cell death-inducing activities, and its receptor, fibroblast growth factor-inducible 14 (Fn14), were examined in postmortem brain tissue samples from patients with multiple sclerosis (MS) and controls. Immunohistochemical analysis and real-time reverse transcription-polymerase chain reaction demonstrated that both TWEAK and Fn14 were upregulated in the MS compared with control unaffected brain samples. Perivascular and meningeal macrophages and astrocytes and microglia associated with lesions were identified as the main sources of TWEAK in the MS brains. The highest frequency of TWEAK+ cells was found at edges of chronic active white matter lesions and in subpial cortical lesions inMS cases with abundant meningeal inflammation and ectopic B-cell follicles. Neurons and reactive astrocytes expressing Fn14 were mainly localized in the cerebral cortex in highly infiltrated MS brains. Numerous TWEAK-expressing microglia were associated with the extensive loss of myelin and astrocytosis, neuronal damage, and vascular abnormalities in subpial cortical lesions; this suggests that TWEAK could synergize with other cytotoxic factors diffusing from the inflamed meninges to promote cortical injury. Taken together, these findings indicate that the TWEAK/Fn14 pathway contributes to inflammation and tissue injury and is, therefore, a potential therapeutic target in MS.


Asunto(s)
Encéfalo/inmunología , Encefalitis/inmunología , Microglía/inmunología , Esclerosis Múltiple/inmunología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/inmunología , Astrocitos/inmunología , Linfocitos B/inmunología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Encéfalo/patología , Encéfalo/fisiopatología , Arterias Cerebrales/inmunología , Arterias Cerebrales/patología , Arterias Cerebrales/fisiopatología , Corteza Cerebral/inmunología , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Quimiotaxis de Leucocito/inmunología , Citocina TWEAK , Encefalitis/patología , Encefalitis/fisiopatología , Gliosis/inmunología , Gliosis/patología , Gliosis/fisiopatología , Humanos , Macrófagos/inmunología , Meninges/inmunología , Meninges/patología , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Receptores del Factor de Necrosis Tumoral/análisis , Receptores del Factor de Necrosis Tumoral/genética , Receptor de TWEAK , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/metabolismo , Regulación hacia Arriba/inmunología
18.
J Clin Invest ; 115(9): 2330-40, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16110324

RESUMEN

Progenitor ("oval") cell expansion accompanies many forms of liver injury, including alcohol toxicity and submassive parenchymal necrosis as well as experimental injury models featuring blocked hepatocyte replication. Oval cells can potentially become either hepatocytes or biliary epithelial cells and may be critical to liver regeneration, particularly when hepatocyte replication is impaired. The regulation of oval cell proliferation is incompletely understood. Herein we present evidence that a TNF family member called TWEAK (TNF-like weak inducer of apoptosis) stimulates oval cell proliferation in mouse liver through its receptor Fn14. TWEAK has no effect on mature hepatocytes and thus appears to be selective for oval cells. Transgenic mice overexpressing TWEAK in hepatocytes exhibit periportal oval cell hyperplasia. A similar phenotype was obtained in adult wild-type mice, but not Fn14-null mice, by administering TWEAK-expressing adenovirus. Oval cell expansion induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) was significantly reduced in Fn14-null mice as well as in adult wild-type mice with a blocking anti-TWEAK mAb. Importantly, TWEAK stimulated the proliferation of an oval cell culture model. Finally, we show increased Fn14 expression in chronic hepatitis C and other human liver diseases relative to its expression in normal liver, which suggests a role for the TWEAK/Fn14 pathway in human liver injury. We conclude that TWEAK has a selective mitogenic effect for liver oval cells that distinguishes it from other previously described growth factors.


Asunto(s)
Proliferación Celular , Hígado/citología , Hígado/metabolismo , Células Madre/fisiología , Factores de Necrosis Tumoral/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Citocina TWEAK , Femenino , Humanos , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/citología , Receptor de TWEAK , Distribución Tisular , Transgenes , Factores de Necrosis Tumoral/genética
19.
Anal Biochem ; 377(1): 95-104, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18358819

RESUMEN

We have deduced the disulfide bond linkage patterns, at very low protein levels (<0.5 nmol), in two cysteine-rich polypeptide domains using a new strategy involving partial reduction/alkylation of the protein, followed by peptide mapping and tanden mass spectrometry (MS/MS) sequencing on a nanoflow liquid chromatography-MS/MS system. The substrates for our work were the cysteine-rich ectodomain of human Fn14, a member of the tumor necrosis factor receptor family, and the IgV domain of murine TIM-1 (T-cell, Ig domain, and mucin domain-1). We have successfully determined the disulfide linkages for Fn14 and independently confirmed those of the IgV domain of TIM-1, whose crystal structure was published recently. The procedures that we describe here can be used to determine the disulfide structures for proteins with complex characteristics. They will also provide a means to obtain important information for structure-function studies and to ensure correct protein folding and batch-to-batch consistency in commercially produced recombinant proteins.


Asunto(s)
Disulfuros/química , Mapeo Peptídico/métodos , Proteínas/química , Proteínas/metabolismo , Espectrometría de Masas en Tándem/métodos , Alquilación , Secuencia de Aminoácidos , Animales , Cromatografía Liquida , Cricetinae , Humanos , Ratones , Datos de Secuencia Molecular , Nanotecnología , Oxidación-Reducción , Estructura Terciaria de Proteína , Proteínas/aislamiento & purificación , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/aislamiento & purificación , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Sensibilidad y Especificidad , Receptor de TWEAK
20.
Genes (Basel) ; 9(12)2018 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-30501089

RESUMEN

Fibroblasts/myofibroblasts are the key effector cells responsible for excessive extracellular matrix (ECM) deposition and fibrosis progression in both idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc) patient lungs, thus it is critical to understand the transcriptomic and proteomic programs underlying their fibrogenic activity. We conducted the first integrative analysis of the fibrotic programming in these cells at the levels of gene and microRNA (miRNA) expression, as well as deposited ECM protein to gain insights into how fibrotic transcriptional programs culminate in aberrant ECM protein production/deposition. We identified messenger RNA (mRNA), miRNA, and deposited matrisome protein signatures for IPF and SSc fibroblasts obtained from lung transplants using next-generation sequencing and mass spectrometry. SSc and IPF fibroblast transcriptional signatures were remarkably similar, with enrichment of WNT, TGF-ß, and ECM genes. miRNA-seq identified differentially regulated miRNAs, including downregulation of miR-29b-3p, miR-138-5p and miR-146b-5p in disease fibroblasts and transfection of their mimics decreased expression of distinct sets of fibrotic signature genes as assessed using a Nanostring fibrosis panel. Finally, proteomic analyses uncovered a distinct "fibrotic" matrisome profile deposited by IPF and SSc fibroblasts compared to controls that highlights the dysregulated ECM production underlying their fibrogenic activities. Our comprehensive analyses of mRNA, miRNA, and matrisome proteomic profiles in IPF and SSc lung fibroblasts revealed robust fibrotic signatures at both the gene and protein expression levels and identified novel fibrogenesis-associated miRNAs whose aberrant downregulation in disease fibroblasts likely contributes to their fibrotic and ECM gene expression.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda