Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(2): 648-53, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23267072

RESUMEN

The coagulation protease activated protein C (aPC) confers cytoprotective effects in various in vitro and in vivo disease models, including diabetic nephropathy. The nephroprotective effect may be related to antioxidant effects of aPC. However, the mechanism through which aPC may convey these antioxidant effects and the functional relevance of these properties remain unknown. Here, we show that endogenous and exogenous aPC prevents glomerular accumulation of oxidative stress markers and of the redox-regulating protein p66(Shc) in experimental diabetic nephropathy. These effects were predominately observed in podocytes. In vitro, aPC inhibited glucose-induced expression of p66(Shc) mRNA and protein in podocytes (via PAR-1 and PAR-3) and various endothelial cell lines, but not in glomerular endothelial cells. Treatment with aPC reversed glucose-induced hypomethylation and hyperacetylation of the p66(Shc) promoter in podocytes. The hyperacetylating agent sodium butyrate abolished the suppressive effect of aPC on p66(Shc) expression both in vitro and in vivo. Moreover, sodium butyrate abolished the beneficial effects of aPC in experimental diabetic nephropathy. Inhibition of p66(Shc) expression and mitochondrial translocation by aPC normalized mitochondrial ROS production and the mitochondrial membrane potential in glucose-treated podocytes. Genetic ablation of p66(Shc) compensated for the loss of protein C activation in vivo, normalizing markers of diabetic nephropathy and oxidative stress. These studies identify a unique mechanism underlying the cytoprotective effect of aPC. Activated PC epigenetically controls expression of the redox-regulating protein p66(Shc), thus linking the extracellular protease aPC to mitochondrial function in diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/tratamiento farmacológico , Represión Epigenética/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Proteína C/farmacología , Proteínas Adaptadoras de la Señalización Shc/antagonistas & inhibidores , Análisis de Varianza , Animales , Butiratos/farmacología , Inmunoprecipitación de Cromatina , Metilación de ADN/efectos de los fármacos , Cartilla de ADN/genética , Nefropatías Diabéticas/etiología , Técnicas de Silenciamiento del Gen , Immunoblotting , Inmunohistoquímica , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Podocitos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Estadísticas no Paramétricas , Fracciones Subcelulares
2.
J Am Soc Nephrol ; 26(11): 2789-99, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26015455

RESUMEN

Ischemia-reperfusion injury (IRI) is the leading cause of ARF. A pathophysiologic role of the coagulation system in renal IRI has been established, but the functional relevance of thrombomodulin (TM)-dependent activated protein C (aPC) generation and the intracellular targets of aPC remain undefined. Here, we investigated the role of TM-dependent aPC generation and therapeutic aPC application in a murine renal IRI model and in an in vitro hypoxia and reoxygenation (HR) model using proximal tubular cells. In renal IRI, endogenous aPC levels were reduced. Genetic or therapeutic reconstitution of aPC efficiently ameliorated renal IRI independently of its anticoagulant properties. In tubular cells, cytoprotective aPC signaling was mediated through protease activated receptor-1- and endothelial protein C receptor-dependent regulation of the cold-shock protein Y-box binding protein-1 (YB-1). The mature 50 kD form of YB-1 was required for the nephro- and cytoprotective effects of aPC in vivo and in vitro, respectively. Reduction of mature YB-1 and K48-linked ubiquitination of YB-1 was prevented by aPC after renal IRI or tubular HR injury. aPC preserved the interaction of YB-1 with the deubiquitinating enzyme otubain-1 and maintained expression of otubain-1, which was required to reduce K48-linked YB-1 ubiquitination and to stabilize the 50 kD form of YB-1 after renal IRI and tubular HR injury. These data link the cyto- and nephroprotective effects of aPC with the ubiquitin-proteasome system and identify YB-1 as a novel intracellular target of aPC. These insights may provide new impetus for translational efforts aiming to restrict renal IRI.


Asunto(s)
Riñón/patología , Proteína C/metabolismo , Daño por Reperfusión/patología , Factores de Transcripción/metabolismo , Ubiquitinación , Alelos , Animales , Anticoagulantes/química , Cruzamientos Genéticos , Cisteína Endopeptidasas/genética , Modelos Animales de Enfermedad , Exones , Hipoxia/patología , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxígeno/química , Transducción de Señal , Trombosis/metabolismo
3.
Kidney Int ; 87(1): 74-84, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25075770

RESUMEN

Diabetic nephropathy is a growing health concern with characteristic sterile inflammation. As the underlying mechanisms of this inflammation remain poorly defined, specific therapies targeting sterile inflammation in diabetic nephropathy are lacking. Intriguingly, an association of diabetic nephropathy with inflammasome activation has recently been shown, but the pathophysiological relevance of this finding remains unknown. Within glomeruli, inflammasome activation was detected in endothelial cells and podocytes in diabetic humans and mice and in glucose-stressed glomerular endothelial cells and podocytes in vitro. Abolishing Nlrp3 or caspase-1 expression in bone marrow-derived cells fails to protect mice against diabetic nephropathy. Conversely, Nlrp3-deficient mice are protected against diabetic nephropathy despite transplantation of wild-type bone marrow. Pharmacological IL-1R antagonism prevented or even reversed diabetic nephropathy in mice. Mitochondrial reactive oxygen species (ROS) activate the Nlrp3 inflammasome in glucose or advanced glycation end product stressed podocytes. Inhibition of mitochondrial ROS prevents glomerular inflammasome activation and nephropathy in diabetic mice. Thus, mitochondrial ROS and Nlrp3-inflammasome activation in non-myeloid-derived cells aggravate diabetic nephropathy. Targeting the inflammasome may be a potential therapeutic approach to diabetic nephropathy.


Asunto(s)
Proteínas Portadoras/inmunología , Nefropatías Diabéticas/inmunología , Inflamasomas/inmunología , Glomérulos Renales/citología , Animales , Células Endoteliales/inmunología , Humanos , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Podocitos/inmunología , Índice de Severidad de la Enfermedad
4.
Development ; 138(11): 2235-47, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21558372

RESUMEN

Absence of the leucine zipper transcription factor p45NF-E2 results in thrombocytopenia, impaired placental vascularization and intrauterine growth restriction (IUGR) in mice. The mechanism underlying the p45NF-E2-dependent placental defect and IUGR remains unknown. Here, we show that the placental defect and IUGR of p45NF-E2 (Nfe2) null mouse embryos is unrelated to thrombocytopenia, establishing that embryonic platelets and platelet-released mediators are dispensable for placentation. Rather, p45NF-E2, which was hitherto thought to be specific to hematopoietic cells, is expressed in trophoblast cells, where it is required for normal syncytiotrophoblast formation, placental vascularization and embryonic growth. Expression of p45NF-E2 in labyrinthine trophoblast cells colocalizes with that of Gcm1, a transcription factor crucial for syncytiotrophoblast formation. In the absence of p45NF-E2, the width of syncytiotrophoblast layer 2 and the expression of Gcm1 and Gcm1-dependent genes (Synb and Cebpa) are increased. In vitro, p45NF-E2 deficiency results in spontaneous syncytiotrophoblast formation, which can be reversed by Gcm1 knockdown. Increased Gcm1 expression in the absence of p45NF-E2 is dependent on enhanced protein acetylation, including post-translational modification of Gcm1. Increasing and inhibiting acetylation in the placenta of wild-type control embryos phenocopies and corrects, respectively, the changes observed in p45NF-E2-deficient embryos. These studies identify a novel function of p45NF-E2 during placental development: in trophoblast cells, p45NF-E2 represses Gcm1 and syncytiotrophoblast formation via acetylation.


Asunto(s)
Desarrollo Embrionario , Subunidad p45 del Factor de Transcripción NF-E2/metabolismo , Neovascularización Fisiológica , Neuropéptidos/metabolismo , Placenta/irrigación sanguínea , Trofoblastos/metabolismo , Acetilación , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Femenino , Retardo del Crecimiento Fetal , Técnicas de Sustitución del Gen , Células Gigantes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Subunidad p45 del Factor de Transcripción NF-E2/genética , Neuropéptidos/genética , Placenta/metabolismo , Placentación , Reacción en Cadena de la Polimerasa , Embarazo , Procesamiento Proteico-Postraduccional , Trombocitopenia , Factores de Transcripción
5.
J Biol Chem ; 287(8): 5400-11, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22174410

RESUMEN

We recently demonstrated that the bZip transcription factor nuclear factor erythroid-derived 2 (Nfe2) represses protein acetylation and expression of the transcription factor glial cell missing 1 (Gcm1) in trophoblast cells, preventing excess syncytiotrophoblast formation and permitting normal placental vascularization and embryonic growth. However, the Gcm1 promoter lacks a Nfe2-binding site and hence the mechanisms linking Nfe2 and Gcm1 expression remained unknown. Here we show that Nfe2 represses JunD DNA-binding activity to the Gcm1 promoter during syncytiotrophoblast differentiation. Interventional studies using knockdown and knockin approaches show that enhanced JunD DNA-binding activity is required for increased expression of Gcm1 and syncytiotrophoblast formation as well as impaired placental vascularization and reduced growth of Nfe2(-/-) embryos. Induction of Gcm1 expression requires binding of JunD to the -1441 site within the Gcm1 promoter, which is distinct from the -1314 site previously shown to induce Gcm1 expression by other bZip transcription factors. Nfe2 modulates JunD binding to the Gcm1 promoter via acetylation, as reducing JunD acetylation using the histone acetyltransferase inhibitor curcumin reverses the increased JunD DNA-binding activity observed in the absence of Nfe2. This identifies a novel mechanism through which bZip transcription factors interact. Within the placenta this interaction regulates Gcm1 expression, syncytiotrophoblast formation, placental vascularization, and embryonic growth.


Asunto(s)
Diferenciación Celular , ADN/metabolismo , Subunidad p45 del Factor de Transcripción NF-E2/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Trofoblastos/citología , Trofoblastos/metabolismo , Acetilación , Animales , Proteínas de Unión al ADN , Femenino , Células HEK293 , Humanos , Ratones , Neovascularización Fisiológica , Neuropéptidos/genética , Placenta/citología , Placenta/embriología , Placenta/metabolismo , Embarazo , Regiones Promotoras Genéticas/genética , Unión Proteica , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción
6.
Sci Total Environ ; 581-582: 434-447, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28088543

RESUMEN

The changing climate in the Arctic has a profound impact on permafrost coasts, which are subject to intensified thermokarst formation and erosion. Consequently, terrestrial organic matter (OM) is mobilized and transported into the nearshore zone. Yet, little is known about the fate of mobilized OM before and after entering the ocean. In this study we investigated a retrogressive thaw slump (RTS) on Qikiqtaruk - Herschel Island (Yukon coast, Canada). The RTS was classified into an undisturbed, a disturbed (thermokarst-affected) and a nearshore zone and sampled systematically along transects. Samples were analyzed for total and dissolved organic carbon and nitrogen (TOC, DOC, TN, DN), stable carbon isotopes (δ13C-TOC, δ13C-DOC), and dissolved inorganic nitrogen (DIN), which were compared between the zones. C/N-ratios, δ13C signatures, and ammonium (NH4-N) concentrations were used as indicators for OM degradation along with biomarkers (n-alkanes, n-fatty acids, n-alcohols). Our results show that OM significantly decreases after disturbance with a TOC and DOC loss of 77 and 55% and a TN and DN loss of 53 and 48%, respectively. C/N-ratios decrease significantly, whereas NH4-N concentrations slightly increase in freshly thawed material. In the nearshore zone, OM contents are comparable to the disturbed zone. We suggest that the strong decrease in OM is caused by initial dilution with melted massive ice and immediate offshore transport via the thaw stream. In the mudpool and thaw stream, OM is subject to degradation, whereas in the slump floor the nitrogen decrease is caused by recolonizing vegetation. Within the nearshore zone of the ocean, heavier portions of OM are directly buried in marine sediments close to shore. We conclude that RTS have profound impacts on coastal environments in the Arctic. They mobilize nutrients from permafrost, substantially decrease OM contents and provide fresh water and nutrients at a point source.

7.
Nat Commun ; 6: 6496, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25754093

RESUMEN

Endoplasmic reticulum (ER) stress is associated with diabetic nephropathy (DN), but its pathophysiological relevance and the mechanisms that compromise adaptive ER signalling in podocytes remain unknown. Here we show that nuclear translocation of the transcription factor spliced X-box binding protein-1 (sXBP1) is selectively impaired in DN, inducing activating transcription factor-6 (ATF6) and C/EBP homology protein (CHOP). Podocyte-specific genetic ablation of XBP1 or inducible expression of ATF6 in mice aggravates DN. sXBP1 lies downstream of insulin signalling and attenuating podocyte insulin signalling by genetic ablation of the insulin receptor or the regulatory subunits phosphatidylinositol 3-kinase (PI3K) p85α or p85ß impairs sXBP1 nuclear translocation and exacerbates DN. Corroborating our findings from murine DN, the interaction of sXBP1 with p85α and p85ß is markedly impaired in the glomerular compartment of human DN. Thus, signalling via the insulin receptor, p85, and XBP1 maintains podocyte homeostasis, while disruption of this pathway impairs podocyte function in DN.


Asunto(s)
Factor de Transcripción Activador 6/genética , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Proteínas de Unión al ADN/genética , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/genética , Estrés del Retículo Endoplásmico/genética , Podocitos/metabolismo , Factores de Transcripción/genética , Factor de Transcripción Activador 6/deficiencia , Animales , Fosfatidilinositol 3-Quinasa Clase Ia/deficiencia , Proteínas de Unión al ADN/deficiencia , Bases de Datos Factuales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/inducido químicamente , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Regulación de la Expresión Génica , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/patología , Receptor de Insulina/deficiencia , Receptor de Insulina/genética , Factores de Transcripción del Factor Regulador X , Transducción de Señal , Estreptozocina , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Factores de Transcripción/deficiencia , Proteína 1 de Unión a la X-Box
8.
Thromb Haemost ; 108(6): 1141-53, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23014597

RESUMEN

Coagulation and complement regulators belong to two interactive systems constituting emerging mechanisms of diabetic nephropathy. Thrombomodulin (TM) regulates both coagulation and complement activation, in part through discrete domains. TM's lectin like domain dampens complement activation, while its EGF-like domains independently enhance activation of the anti-coagulant and cytoprotective serine protease protein C (PC). A protective effect of activated PC in diabetic nephropathy is established. We hypothesised that TM controls diabetic nephropathy independent of PC through its lectin-like domain by regulating complement. Diabetic nephropathy was analysed in mice lacking TM's lectin-like domain (TMLeD/LeD) and controls (TMwt/wt). Albuminuria (290 µg/mg vs. 166 µg/mg, p=0.03) and other indices of experimental diabetic nephropathy were aggravated in diabetic TMLeD/LeD mice. Complement deposition (C3 and C5b-9) was markedly increased in glomeruli of diabetic TMLeD/LeD mice. Complement inhibition with enoxaparin ameliorated diabetic nephropathy in TMLeD/LeD mice (e.g. albuminuria 85 µg/mg vs. 290 µg/mg, p<0.001). In vitro TM's lectin-like domain cell-autonomously prevented glucose-induced complement activation on endothelial cells and - notably - on podocytes. Podocyte injury, which was enhanced in diabetic TMLeD/LeD mice, was reduced following complement inhibition with enoxaparin. The current study identifies a novel mechanism regulating complement activation in diabetic nephropathy. TM's lectin-like domain constrains glucose-induced complement activation on endothelial cells and podocytes and ameliorates albuminuria and glomerular damage in mice.


Asunto(s)
Nefropatías Diabéticas/etiología , Trombomodulina/química , Trombomodulina/fisiología , Animales , Línea Celular , Activación de Complemento/fisiología , Proteínas Inactivadoras de Complemento/química , Proteínas Inactivadoras de Complemento/deficiencia , Proteínas Inactivadoras de Complemento/genética , Proteínas Inactivadoras de Complemento/fisiología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/fisiopatología , Nefropatías Diabéticas/prevención & control , Células Endoteliales/inmunología , Células Endoteliales/patología , Células Endoteliales/fisiología , Glomérulos Renales/inmunología , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/fisiología , Podocitos/inmunología , Podocitos/patología , Podocitos/fisiología , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombomodulina/deficiencia , Trombomodulina/genética
9.
Atherosclerosis ; 219(1): 74-83, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21719015

RESUMEN

OBJECTIVE: Minocycline, a tetracycline derivate, mediates vasculoprotective effects independent of its antimicrobial properties. Thus, minocycline protects against diabetic nephropathy and reduces neointima formation following vascular injury through inhibition of apoptosis or migration, respectively. Whether minocycline has an effect on primary atherogenesis remains unknown. METHODS: Using morphological and immunohistochemical analyses we determined de novo atherogenesis in ApoE-/- mice receiving a high fat diet (HFD) with or without minocycline treatment. The effect of minocycline on proliferation, expression of p27(Kip1) or PARP-1 (Poly [ADP-ribose] polymerase 1), or on PAR (poly ADP-ribosylation) modification in vascular smooth muscle cells (VSMC) was analyzed in ex vivo and in vitro (primary human and mouse VSMC). RESULTS AND CONCLUSION: Minocycline reduced plaque size and stenosis in ApoE-/- HFD mice. This was associated with a lower number and less proliferation of VSMC, reduced PAR (poly ADP-ribosylation) modification and increased p27(Kip1) expression within the plaques. In agreement with the ex vivo data minocycline reduced proliferation, PARP-1 expression, PAR modification while inducing p27 expression in human and mouse VSMC in vitro. These effects were observed at a low minocycline concentration (10 µM), which had no effect on VSMC migration or apoptosis. Minocycline inhibited PARP-1 and induced p27(Kip1) expression in VSMC as efficiently as the specific PARP-1 inhibitor PJ 34. Knock down of p27(Kip1) abolished the antiproliferative effect of minocycline. These data establish a novel antiatherosclerotic mechanism of minocycline during de novo atherogenesis, which depends on p27(Kip1) mediated inhibition of VSMC proliferation.


Asunto(s)
Apolipoproteínas E/deficiencia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Minociclina/farmacología , Miocitos del Músculo Liso/efectos de los fármacos , Placa Aterosclerótica/prevención & control , Animales , Estenosis de la Válvula Aórtica/prevención & control , Aterosclerosis/metabolismo , Proliferación Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Dieta Alta en Grasa , Humanos , Ratones , Músculo Liso Vascular/efectos de los fármacos , Fenantrenos/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Poli(ADP-Ribosa) Polimerasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA