Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 111(37): 13493-8, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25197062

RESUMEN

It is poorly understood why there is greater cardiovascular disease risk associated with the apolipoprotein E4 (apoE) allele vs. apoE3, and also greater risk with the LRP8/apolipoprotein E receptor 2 (ApoER2) variant ApoER2-R952Q. Little is known about the function of the apoE-ApoER2 tandem outside of the central nervous system. We now report that in endothelial cells apoE3 binding to ApoER2 stimulates endothelial NO synthase (eNOS) and endothelial cell migration, and it also attenuates monocyte-endothelial cell adhesion. However, apoE4 does not stimulate eNOS or endothelial cell migration or dampen cell adhesion, and alternatively it selectively antagonizes apoE3/ApoER2 actions. The contrasting endothelial actions of apoE4 vs. apoE3 require the N-terminal to C-terminal interaction in apoE4 that distinguishes it structurally from apoE3. Reconstitution experiments further reveal that ApoER2-R952Q is a loss-of-function variant of the receptor in endothelium. Carotid artery reendothelialization is decreased in ApoER2(-/-) mice, and whereas adenoviral-driven apoE3 expression in wild-type mice has no effect, apoE4 impairs reendothelialization. Moreover, in a model of neointima formation invoked by carotid artery endothelial denudation, ApoER2(-/-) mice display exaggerated neointima development. Thus, the apoE3/ApoER2 tandem promotes endothelial NO production, endothelial repair, and endothelial anti-inflammatory properties, and it prevents neointima formation. In contrast, apoE4 and ApoER2-R952Q display dominant-negative action and loss of function, respectively. Thus, genetic variants of apoE and ApoER2 impact cardiovascular health by differentially modulating endothelial function.


Asunto(s)
Apolipoproteínas E/genética , Células Endoteliales/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Células 3T3 , Animales , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Arterias Carótidas/metabolismo , Bovinos , Adhesión Celular , Movimiento Celular , Células Endoteliales/citología , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ratones , Monocitos/citología , Proteínas Mutantes/metabolismo , Neointima/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo
2.
Proc Natl Acad Sci U S A ; 111(1): E129-38, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24367119

RESUMEN

Mammalian skeletal muscle can remodel, repair, and regenerate itself by mobilizing satellite cells, a resident population of myogenic progenitor cells. Muscle injury and subsequent activation of myogenic progenitor cells is associated with oxidative stress. Cytoglobin is a hemoprotein expressed in response to oxidative stress in a variety of tissues, including striated muscle. In this study, we demonstrate that cytoglobin is up-regulated in activated myogenic progenitor cells, where it localizes to the nucleus and contributes to cell viability. siRNA-mediated depletion of cytoglobin from C2C12 myoblasts increased levels of reactive oxygen species and apoptotic cell death both at baseline and in response to stress stimuli. Conversely, overexpression of cytoglobin reduced reactive oxygen species levels, caspase activity, and cell death. Mice in which cytoglobin was knocked out specifically in skeletal muscle were generated to examine the role of cytoglobin in vivo. Myogenic progenitor cells isolated from these mice were severely deficient in their ability to form myotubes as compared with myogenic progenitor cells from wild-type littermates. Consistent with this finding, the capacity for muscle regeneration was severely impaired in mice deficient for skeletal-muscle cytoglobin. Collectively, these data demonstrate that cytoglobin serves an important role in muscle repair and regeneration.


Asunto(s)
Regulación de la Expresión Génica , Globinas/metabolismo , Músculos/fisiología , Regeneración/fisiología , Células Satélite del Músculo Esquelético/citología , Animales , Apoptosis , Diferenciación Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Citoglobina , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno , Células Satélite del Músculo Esquelético/patología , Células Madre/citología , Factores de Tiempo
3.
J Biol Chem ; 290(17): 10703-16, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25733667

RESUMEN

In beating hearts, phosphorylation of myosin regulatory light chain (RLC) at a single site to 0.45 mol of phosphate/mol by cardiac myosin light chain kinase (cMLCK) increases Ca(2+) sensitivity of myofilament contraction necessary for normal cardiac performance. Reduction of RLC phosphorylation in conditional cMLCK knock-out mice caused cardiac dilation and loss of cardiac performance by 1 week, as shown by increased left ventricular internal diameter at end-diastole and decreased fractional shortening. Decreased RLC phosphorylation by conventional or conditional cMLCK gene ablation did not affect troponin-I or myosin-binding protein-C phosphorylation in vivo. The extent of RLC phosphorylation was not changed by prolonged infusion of dobutamine or treatment with a ß-adrenergic antagonist, suggesting that RLC is constitutively phosphorylated to maintain cardiac performance. Biochemical studies with myofilaments showed that RLC phosphorylation up to 90% was a random process. RLC is slowly dephosphorylated in both noncontracting hearts and isolated cardiac myocytes from adult mice. Electrically paced ventricular trabeculae restored RLC phosphorylation, which was increased to 0.91 mol of phosphate/mol of RLC with inhibition of myosin light chain phosphatase (MLCP). The two RLCs in each myosin appear to be readily available for phosphorylation by a soluble cMLCK, but MLCP activity limits the amount of constitutive RLC phosphorylation. MLCP with its regulatory subunit MYPT2 bound tightly to myofilaments was constitutively phosphorylated in beating hearts at a site that inhibits MLCP activity. Thus, the constitutive RLC phosphorylation is limited physiologically by low cMLCK activity in balance with low MLCP activity.


Asunto(s)
Miocardio/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Animales , Calcio/metabolismo , Calmodulina/metabolismo , Ventrículos Cardíacos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Cadenas Ligeras de Miosina/deficiencia , Cadenas Ligeras de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley
4.
Circ Res ; 112(1): 140-51, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23023567

RESUMEN

RATIONALE: Signal initiation by the high-density lipoprotein (HDL) receptor scavenger receptor class B, type I (SR-BI), which is important to actions of HDL on endothelium and other processes, requires cholesterol efflux and the C-terminal transmembrane domain. The C-terminal transmembrane domain uniquely interacts with plasma membrane (PM) cholesterol. OBJECTIVE: The molecular basis and functional significance of SR-BI interaction with PM cholesterol are unknown. We tested the hypotheses that the interaction is required for SR-BI signaling, and that it enables SR-BI to serve as a PM cholesterol sensor. METHODS AND RESULTS: In studies performed in COS-M6 cells, mutation of a highly conserved C-terminal transmembrane domain glutamine to alanine (SR-BI-Q445A) decreased PM cholesterol interaction with the receptor by 71% without altering HDL binding or cholesterol uptake or efflux, and it yielded a receptor incapable of HDL-induced signaling. Signaling prompted by cholesterol efflux to methyl-ß-cyclodextrin also was prevented, indicating that PM cholesterol interaction with the receptor enables it to serve as a PM cholesterol sensor. Using SR-BI-Q445A, we further demonstrated that PM cholesterol sensing by SR-BI does not influence SR-BI-mediated reverse cholesterol transport to the liver in mice. However, the PM cholesterol sensing does underlie apolipoprotein B intracellular trafficking in response to postprandial micelles or methyl-ß-cyclodextrin in cultured enterocytes, and it is required for HDL activation of endothelial NO synthase and migration in cultured endothelial cells and HDL-induced angiogenesis in vivo. CONCLUSIONS: Through interaction with PM cholesterol, SR-BI serves as a PM cholesterol sensor, and the resulting intracellular signaling governs processes in both enterocytes and endothelial cells.


Asunto(s)
Membrana Celular/metabolismo , Colesterol/metabolismo , Células Endoteliales/metabolismo , Enterocitos/metabolismo , Receptores Depuradores de Clase B/metabolismo , Transducción de Señal , Alanina , Animales , Apolipoproteínas B/metabolismo , Células CACO-2 , Bovinos , Membrana Celular/efectos de los fármacos , HDL-Colesterol/metabolismo , Células Endoteliales/efectos de los fármacos , Enterocitos/efectos de los fármacos , Glutamina , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Mutación , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Receptores Depuradores de Clase B/química , Receptores Depuradores de Clase B/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , beta-Ciclodextrinas/farmacología
5.
Proc Natl Acad Sci U S A ; 109(19): 7529-34, 2012 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-22529366

RESUMEN

During pregnancy, uterine quiescence is maintained by increased progesterone receptor (PR) activity, but labor is facilitated by a series of events that impair PR function. Previously, we discovered that miR-200 family members serve as progesterone (P(4))-modulated activators of contraction-associated genes in the pregnant uterus. In this study, we identified a unique role for miR-200a to enhance the local metabolism of P(4) in myometrium and, thus, decrease PR function during the progression toward labor. miR-200a exerts this action by direct repression of STAT5b, a transcriptional repressor of the P(4)-metabolizing enzyme 20α-hydroxysteroid dehydrogenase (20α-HSD). We observed that miR-200a expression increased and STAT5b expression coordinately decreased in myometrium of mice as they progressed to labor and in laboring myometrium from pregnant women. These changes were associated with a dramatic increase in expression and activity of 20α-HSD in laboring myometrium from mouse and human. Notably, overexpression of miR-200a in cultured human myometrial cells (hTERT-HM) suppressed STAT5b and increased 20α-HSD mRNA levels. In uterine tissues of ovariectomized mice injected with P(4), miR-200 expression was significantly decreased, STAT5b expression was up-regulated, and 20α-HSD mRNA was decreased, but in 15 d postcoitum pregnant mice injected with the PR antagonist RU486, preterm labor was associated with increased miR-200a, decreased STAT5b, and enhanced 20α-HSD expression. Taken together, these findings implicate miR-200a as an important regulator of increased local P(4) metabolism in the pregnant uterus near term and provide insight into the importance of miR-200s in the decline in PR function leading to labor.


Asunto(s)
Trabajo de Parto/genética , MicroARNs/genética , Trabajo de Parto Prematuro/genética , Receptores de Progesterona/genética , 20-alfa-Hidroxiesteroide Deshidrogenasa/genética , 20-alfa-Hidroxiesteroide Deshidrogenasa/metabolismo , Animales , Células Cultivadas , Femenino , Expresión Génica/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Humanos , Immunoblotting , Trabajo de Parto/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Mifepristona/farmacología , Miometrio/citología , Miometrio/metabolismo , Trabajo de Parto Prematuro/metabolismo , Ovariectomía , Embarazo , Progesterona/metabolismo , Progesterona/farmacología , Receptores de Progesterona/antagonistas & inhibidores , Receptores de Progesterona/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Útero/efectos de los fármacos , Útero/metabolismo
6.
Proc Natl Acad Sci U S A ; 109(8): 3143-8, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22315431

RESUMEN

The endocrine hormone fibroblast growth factor 21 (FGF21) is a powerful modulator of glucose and lipid metabolism and a promising drug for type 2 diabetes. Here we identify FGF21 as a potent regulator of skeletal homeostasis. Both genetic and pharmacologic FGF21 gain of function lead to a striking decrease in bone mass. In contrast, FGF21 loss of function leads to a reciprocal high-bone-mass phenotype. Mechanistically, FGF21 inhibits osteoblastogenesis and stimulates adipogenesis from bone marrow mesenchymal stem cells by potentiating the activity of peroxisome proliferator-activated receptor γ (PPAR-γ). Consequently, FGF21 deletion prevents the deleterious bone loss side effect of the PPAR-γ agonist rosiglitazone. Therefore, FGF21 is a critical rheostat for bone turnover and a key integrator of bone and energy metabolism. These results reveal that skeletal fragility may be an undesirable consequence of chronic FGF21 administration.


Asunto(s)
Resorción Ósea/patología , Factores de Crecimiento de Fibroblastos/metabolismo , PPAR gamma/metabolismo , Adipogénesis/efectos de los fármacos , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Resorción Ósea/metabolismo , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Resistencia a Medicamentos/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/administración & dosificación , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/patología , Osteogénesis/efectos de los fármacos , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacología
7.
J Cell Sci ; 125(Pt 22): 5329-37, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22956541

RESUMEN

In response to severe injury, adult skeletal muscle exhibits a remarkable regenerative capacity due to a resident muscle stem/progenitor cell population. While a number of factors are expressed in the muscle progenitor cell (MPC) population, the molecular networks that govern this cell population remain an area of active investigation. In this study, utilizing knockdown techniques and overexpression of Foxk1 in the myogenic lineage, we observed dysregulation of Foxo and Mef2 downstream targets. Utilizing an array of technologies, we establish that Foxk1 represses the transcriptional activity of Foxo4 and Mef2 and physically interacts with Foxo4 and Mef2, thus promoting MPC proliferation and antagonizing the myogenic lineage differentiation program, respectively. Correspondingly, knockdown of Foxk1 in C2C12 myoblasts results in cell cycle arrest, and Foxk1 overexpression in C2C12CAR myoblasts retards muscle differentiation. Collectively, we have established that Foxk1 promotes MPC proliferation by repressing Foxo4 transcriptional activity and inhibits myogenic differentiation by repressing Mef2 activity. These studies enhance our understanding of the transcriptional networks that regulate the MPC population and muscle regeneration.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/metabolismo , Desarrollo de Músculos , Factores Reguladores Miogénicos/metabolismo , Animales , Ciclo Celular , Proteínas de Ciclo Celular , Proliferación Celular , ADN/metabolismo , Factores de Transcripción MEF2 , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Unión Proteica , Regeneración , Proteínas Represoras/metabolismo , Transcripción Genética
8.
J Biol Chem ; 287(36): 30800-11, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22807441

RESUMEN

Hypoxia-inducible factors (HIFs) are oxygen-sensitive transcription factors. HIF-1α plays a prominent role in hypoxic gene induction. HIF-2α target genes are more restricted but include erythropoietin (Epo), one of the most highly hypoxia-inducible genes in mammals. We previously reported that HIF-2α is acetylated during hypoxia but is rapidly deacetylated by the stress-responsive deacetylase Sirtuin 1. We now demonstrate that the lysine acetyltransferases cAMP-response element-binding protein-binding protein (CBP) and p300 are required for efficient Epo induction during hypoxia. However, despite close structural similarity, the roles of CBP and p300 differ in HIF signaling. CBP acetylates HIF-2α, is a major coactivator for HIF-2-mediated Epo induction, and is required for Sirt1 augmentation of HIF-2 signaling during hypoxia in Hep3B cells. In comparison, p300 is a major contributor for HIF-1 signaling as indicated by induction of Pgk1. Whereas CBP can bind with HIF-2α independent of the HIF-2α C-terminal activation domain via enzyme/substrate interactions, p300 only complexes with HIF-2α through the C-terminal activation domain. Maximal CBP/HIF-2 signaling requires intact CBP acetyltransferase activity in both Hep3B cells as well as in mice.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína de Unión a CREB/metabolismo , Fragmentos de Péptidos/metabolismo , Sialoglicoproteínas/metabolismo , Transducción de Señal/fisiología , Sirtuina 1/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteína de Unión a CREB/genética , Línea Celular , Eritropoyetina/biosíntesis , Eritropoyetina/genética , Humanos , Ratones , Fragmentos de Péptidos/genética , Fosfoglicerato Quinasa/genética , Fosfoglicerato Quinasa/metabolismo , Estructura Terciaria de Proteína , Sialoglicoproteínas/genética , Sirtuina 1/genética , Factores de Transcripción p300-CBP/genética
9.
J Biol Chem ; 287(49): 41334-41, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23038264

RESUMEN

Fibroblast growth factor 19 (FGF19) is a postprandial enterokine induced by the nuclear bile acid receptor, FXR, in ileum. FGF19 inhibits bile acid synthesis in liver through transcriptional repression of cholesterol 7α-hydroxylase (CYP7A1) via a mechanism involving the nuclear receptor SHP. Here, in a series of loss-of-function studies, we show that the nuclear receptors HNF4α and LRH-1 have dual roles in regulating Cyp7a1 in vivo. First, they cooperate in maintaining basal Cyp7a1 expression. Second, they enable SHP binding to the Cyp7a1 promoter and facilitate FGF19-mediated repression of bile acid synthesis. HNF4α and LRH-1 promote active transcription histone marks on the Cyp7a1 promoter that are reversed by FGF19 in a SHP-dependent manner. These findings demonstrate that both HNF4α and LRH-1 are important regulators of Cyp7a1 transcription in vivo.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/biosíntesis , Regulación de la Expresión Génica , Factor Nuclear 4 del Hepatocito/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Ácidos y Sales Biliares/metabolismo , Sitios de Unión , Factores de Crecimiento de Fibroblastos/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Hígado/metabolismo , Ratones , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transcripción Genética
10.
Nat Med ; 12(11): 1253-5, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17072310

RESUMEN

The cycle of gallbladder filling and emptying controls the flow of bile into the intestine for digestion. Here we show that fibroblast growth factor-15, a hormone made by the distal small intestine in response to bile acids, is required for gallbladder filling. These studies demonstrate that gallbladder filling is actively regulated by an endocrine pathway and suggest a postprandial timing mechanism that controls gallbladder motility.


Asunto(s)
Factores de Crecimiento de Fibroblastos/fisiología , Vesícula Biliar/fisiología , Animales , Colecistoquinina/sangre , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/genética , Vesícula Biliar/metabolismo , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética
11.
Proc Natl Acad Sci U S A ; 107(48): 20828-33, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21079000

RESUMEN

Throughout most of pregnancy, uterine quiescence is maintained by increased progesterone receptor (PR) transcriptional activity, whereas spontaneous labor is initiated/facilitated by a concerted series of biochemical events that activate inflammatory pathways and have a negative impact on PR function. In this study, we uncovered a previously undescribed regulatory pathway whereby micro-RNAs (miRNAs) serve as hormonally modulated and conserved mediators of contraction-associated genes in the pregnant uterus in the mouse and human. Using miRNA and gene expression microarray analyses of uterine tissues, we identified a conserved family of miRNAs, the miR-200 family, that is highly induced at term in both mice and humans as well as two coordinately down-regulated targets, zinc finger E-box binding homeobox proteins ZEB1 and ZEB2, which act as transcriptional repressors. We also observed up-regulation of the miR-200 family and down-regulation of ZEB1 and ZEB2 in two different mouse models of preterm labor. We further demonstrated that ZEB1 is directly up-regulated by the action of progesterone (P(4))/PR at the ZEB1 promoter. Excitingly, we observed that ZEB1 and ZEB2 inhibit expression of the contraction-associated genes, oxytocin receptor and connexin-43, and block oxytocin-induced contractility in human myometrial cells. Together, these findings implicate the miR-200 family and their targets, ZEB1 and ZEB2, as unique P(4)/PR-mediated regulators of uterine quiescence and contractility during pregnancy and labor and shed light on the molecular mechanisms involved in preterm birth.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Trabajo de Parto/genética , MicroARNs/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Contracción Uterina/genética , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Trabajo de Parto/efectos de los fármacos , Ratones , MicroARNs/genética , Modelos Animales , Miometrio/efectos de los fármacos , Miometrio/metabolismo , Embarazo , Progesterona/farmacología , Receptores de Progesterona/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Contracción Uterina/efectos de los fármacos , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
12.
Proc Natl Acad Sci U S A ; 107(9): 4218-23, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20142475

RESUMEN

microRNAs (miRNAs) play key roles in modulating a variety of cellular processes through repression of mRNA targets. In a screen for miRNAs regulated by myocardin-related transcription factor-A (MRTF-A), a coactivator of serum response factor (SRF), we discovered a muscle-enriched miRNA, miR-486, controlled by an alternative promoter within intron 40 of the Ankyrin-1 gene. Transcription of miR-486 is directly controlled by SRF and MRTF-A, as well as by MyoD. Among the most strongly predicted targets of miR-486 are phosphatase and tensin homolog (PTEN) and Foxo1a, which negatively affect phosphoinositide-3-kinase (PI3K)/Akt signaling. Accordingly, PTEN and Foxo1a protein levels are reduced by miR-486 overexpression, which, in turn, enhances PI3K/Akt signaling. Similarly, we show that MRTF-A promotes PI3K/Akt signaling by up-regulating miR-486 expression. Conversely, inhibition of miR-486 expression enhances the expression of PTEN and Foxo1a and dampens signaling through the PI3K/Akt-signaling pathway. Our findings implicate miR-486 as a downstream mediator of the actions of SRF/MRTF-A and MyoD in muscle cells and as a potential modulator of PI3K/Akt signaling.


Asunto(s)
MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Northern Blotting , Ensayo de Cambio de Movilidad Electroforética , Hibridación in Situ , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Transducción de Señal , Transactivadores/metabolismo
13.
J Biol Chem ; 286(16): 13869-78, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21345792

RESUMEN

Hypoxia-inducible factors (HIFs) are stress-responsive transcriptional regulators of cellular and physiological processes involved in oxygen metabolism. Although much is understood about the molecular machinery that confers HIF responsiveness to oxygen, far less is known about HIF isoform-specific mechanisms of regulation, despite the fact that HIF-1 and HIF-2 exhibit distinct biological roles. We recently determined that the stress-responsive genetic regulator sirtuin 1 (Sirt1) selectively augments HIF-2 signaling during hypoxia. However, the mechanism by which Sirt1 maintains activity during hypoxia is unknown. In this report, we demonstrate that Sirt1 gene expression increases in a HIF-dependent manner during hypoxia in Hep3B and in HT1080 cells. Impairment of HIF signaling affects Sirt1 deacetylase activity as decreased HIF-1 signaling results in the appearance of acetylated HIF-2α, which is detected without pharmacological inhibition of Sirt1. We also find that Sirt1 augments HIF-2 mediated, but not HIF-1 mediated, transcriptional activation of the isolated Sirt1 promoter. These data in summary reveal a bidirectional link of HIF and Sirt1 signaling during hypoxia.


Asunto(s)
Regulación de la Expresión Génica , Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia , Sirtuina 1/biosíntesis , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Humanos , Ratones , Ratones Endogámicos C57BL , Procesamiento Proteico-Postraduccional , Transducción de Señal , Activación Transcripcional
14.
J Biol Chem ; 286(43): 37676-91, 2011 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-21873652

RESUMEN

Loss-of-function mutations in 1-acylglycerol-3-phosphate O-acyltransferase (AGPAT) 2 in humans and mice result in loss of both the white and brown adipose tissues from birth. AGPAT2 generates precursors for the synthesis of glycerophospholipids and triacylglycerols. Loss of adipose tissue, or lipodystrophy, results in hyperinsulinemia, diabetes mellitus, and severe hepatic steatosis. Here, we analyzed biochemical properties of human AGPAT2 and its close homolog, AGPAT1, and we studied their role in liver by transducing their expression via recombinant adenoviruses in Agpat2(-/-) mice. The in vitro substrate specificities of AGPAT1 and AGPAT2 are quite similar for lysophosphatidic acid and acyl-CoA. Protein homology modeling of both the AGPATs with glycerol-3-phosphate acyltransferase 1 (GPAT1) revealed that they have similar tertiary protein structure, which is consistent with their similar substrate specificities. When co-expressed, both isoforms co-localize to the endoplasmic reticulum. Despite such similarities, restoring AGPAT activity in liver by overexpression of either AGPAT1 or AGPAT2 in Agpat2(-/-) mice failed to ameliorate the hepatic steatosis. From these studies, we suggest that the role of AGPAT1 or AGPAT2 in liver lipogenesis is minimal and that accumulation of liver fat is primarily a consequence of insulin resistance and loss of adipose tissue in Agpat2(-/-) mice.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Tejido Adiposo/enzimología , Retículo Endoplásmico/enzimología , Hígado Graso/enzimología , Lipodistrofia/enzimología , Hígado/enzimología , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética , Tejido Adiposo/patología , Animales , Retículo Endoplásmico/genética , Retículo Endoplásmico/patología , Hígado Graso/genética , Hígado Graso/patología , Glicerofosfolípidos/biosíntesis , Glicerofosfolípidos/genética , Células HEK293 , Humanos , Resistencia a la Insulina/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Lipodistrofia/genética , Lipodistrofia/patología , Hígado/patología , Ratones , Ratones Noqueados , Transducción Genética , Triglicéridos/biosíntesis , Triglicéridos/genética
15.
J Biol Chem ; 286(17): 15116-25, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21357625

RESUMEN

G protein-coupled receptor (GPCR) pathways control glucose and fatty acid metabolism and the onset of obesity and diabetes. Regulators of G protein signaling (RGS) are GTPase-activating proteins (GAPs) for G(i) and G(q) α-subunits that control the intensity and duration of GPCR signaling. Herein we determined the role of Rgs16 in GPCR regulation of liver metabolism. Rgs16 is expressed during the last few hours of the daily fast in periportal hepatocytes, the oxygen-rich zone of the liver where lipolysis and gluconeogenesis predominate. Rgs16 knock-out mice had elevated expression of fatty acid oxidation genes in liver, higher rates of fatty acid oxidation in liver extracts, and higher plasma ß-ketone levels compared with wild type mice. By contrast, transgenic mice that overexpressed RGS16 protein specifically in liver exhibited reciprocal phenotypes as well as low blood glucose levels compared with wild type littermates and fatty liver after overnight fasting. The transcription factor carbohydrate response element-binding protein (ChREBP), which induces fatty acid synthesis genes in response to high carbohydrate feeding, was unexpectedly required during fasting for maximal Rgs16 transcription in liver and in cultured primary hepatocytes during gluconeogenesis. Thus, RGS16 provides a signaling mechanism for glucose production to inhibit GPCR-stimulated fatty acid oxidation in hepatocytes.


Asunto(s)
Ácidos Grasos/metabolismo , Proteínas Nucleares/fisiología , Proteínas RGS/fisiología , Factores de Transcripción/fisiología , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Ácidos Grasos/biosíntesis , Ácidos Grasos/genética , Gluconeogénesis , Glucosa/biosíntesis , Glucosa/fisiología , Hepatocitos/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Oxidación-Reducción , Receptores Acoplados a Proteínas G/metabolismo , Transcripción Genética
16.
Circ Res ; 107(2): 294-304, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20558820

RESUMEN

RATIONALE: Myocardial infarction (MI) results in loss of cardiac myocytes in the ischemic zone of the heart, followed by fibrosis and scar formation, which diminish cardiac contractility and impede angiogenesis and repair. Myofibroblasts, a specialized cell type that switches from a fibroblast-like state to a contractile, smooth muscle-like state, are believed to be primarily responsible for fibrosis of the injured heart and other tissues, although the transcriptional mediators of fibrosis and myofibroblast activation remain poorly defined. Myocardin-related transcription factors (MRTFs) are serum response factor (SRF) cofactors that promote a smooth muscle phenotype and are emerging as components of stress-responsive signaling. OBJECTIVE: We aimed to examine the effect of MRTF-A on cardiac remodeling and fibrosis. METHODS AND RESULTS: Here, we show that MRTF-A controls the expression of a fibrotic gene program that includes genes involved in extracellular matrix production and smooth muscle cell differentiation in the heart. In MRTF-A-null mice, fibrosis and scar formation following MI or angiotensin II treatment are dramatically diminished compared with wild-type littermates. This protective effect of MRTF-A deletion is associated with a reduction in expression of fibrosis-associated genes, including collagen 1a2, a direct transcriptional target of SRF/MRTF-A. CONCLUSIONS: We conclude that MRTF-A regulates myofibroblast activation and fibrosis in response to the renin-angiotensin system and post-MI remodeling.


Asunto(s)
Transdiferenciación Celular , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos del Músculo Liso/metabolismo , Transactivadores/metabolismo , Remodelación Ventricular , Amidas/farmacología , Angiotensina II/administración & dosificación , Animales , Secuencia de Bases , Células COS , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Chlorocebus aethiops , Colágeno/genética , Colágeno Tipo I , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibrosis , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fenotipo , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Factores de Tiempo , Transactivadores/deficiencia , Transactivadores/genética , Transcripción Genética , Transfección , Factor de Crecimiento Transformador beta1/metabolismo , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/genética , Quinasas Asociadas a rho/metabolismo
17.
J Biol Chem ; 285(29): 22103-13, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20466729

RESUMEN

Rare heterozygous mutations in the gene encoding surfactant protein A2 (SP-A2, SFTPA2) are associated with adult-onset pulmonary fibrosis and adenocarcinoma of the lung. We have previously shown that two recombinant SP-A2 mutant proteins (G231V and F198S) remain within the endoplasmic reticulum (ER) of A549 cells and are not secreted into the culture medium. The pathogenic mechanism of the mutant proteins is unknown. Here we analyze all common and rare variants of the surfactant protein A2, SP-A2, in both A549 cells and in primary type II alveolar epithelial cells. We show that, in contrast with all other SP-A2 variants, the mutant proteins are not secreted into the medium with wild-type SP-A isoforms, form fewer intracellular dimer and trimer oligomers, are partially insoluble in 0.5% Nonidet P-40 lysates of transfected A549 cells, and demonstrate greater protein instability in chymotrypsin proteolytic digestions. Both the G231V and F198S mutant SP-A2 proteins are destroyed via the ER-association degradation pathway. Expression of the mutant proteins increases the transcription of a BiP-reporter construct, expression of BiP protein, and production of an ER stress-induced XBP-1 spliced product. Human bronchoalveolar wash samples from individuals who are heterozygous for the G231V mutation have similar levels of total SP-A as normal family members, which suggests that the mechanism of disease does not involve an overt lack of secreted SP-A but instead involves an increase in ER stress of resident type II alveolar epithelial cells.


Asunto(s)
Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Mutación/genética , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Proteína A Asociada a Surfactante Pulmonar/genética , Estrés Fisiológico , Sustitución de Aminoácidos/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Líquido del Lavado Bronquioalveolar/química , Extractos Celulares , Línea Celular Tumoral , Detergentes/farmacología , Perros , Retículo Endoplásmico/efectos de los fármacos , Femenino , Humanos , Leupeptinas/farmacología , Masculino , Proteínas Mutantes/metabolismo , Linaje , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Estructura Cuaternaria de Proteína , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Fibrosis Pulmonar/fisiopatología , Proteína A Asociada a Surfactante Pulmonar/química , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Solubilidad/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos
18.
J Biol Chem ; 285(22): 16623-31, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20371871

RESUMEN

Regulator of calcineurin 1 (RCAN1) inhibits the protein phosphatase calcineurin and is required for appropriate immune responses, synaptic plasticity, vascular tone, angiogenesis, and cardiac remodeling. Expression of the RCAN1-4 isoform is under the control of the calcineurin-responsive transcription factor NFAT. Typically, NFATs act in cooperation with other transcription factors to achieve maximal activation of gene expression. In this study, we identify the CCAAT/enhancer binding protein beta (C/EBPbeta) as an NFAT binding partner that cooperates with NFAT to regulate RCAN1-4 expression. Numerous C/EBPbeta binding sites are conserved in the RCAN1-4 proximal promoter. Overexpression of C/EBPbeta increased activity of both the endogenous mouse Rcan1-4 gene and a human RCAN1-4 luciferase reporter. Binding of C/EBPbeta to multiple sites in the promoter was verified using electrophoretic mobility shift assays and chromatin immunoprecipitation. A direct interaction between C/EBPbeta and NFAT was demonstrated by co-immunoprecipitation of proteins and complex formation at NFAT-C/EBPbeta composite sites. Depletion of endogenous C/EBPbeta decreased maximal activation of RCAN1-4 expression by calcineurin, whereas inhibition of calcineurin did not alter the ability of C/EBPbeta to activate RCAN1-4 expression. Together, these findings suggest that calcineurin/NFAT activation of RCAN1-4 expression is in part dependent upon C/EBPbeta, whereas activation by C/EBPbeta is not dependent on calcineurin and may provide a calcineurin-independent pathway for regulating RCAN1-4 expression. Importantly, nuclear localization, C/EBPbeta DNA binding activity and occupancy of the Rcan1-4 promoter increased in mouse models of heart failure demonstrating in vivo activation of this pathway to regulate Rcan1-4 expression and ultimately shape the dynamics of calcineurin-dependent signaling.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Musculares/metabolismo , Factores de Transcripción NFATC/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Calcineurina/metabolismo , Proteínas de Unión al Calcio , Línea Celular , Núcleo Celular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Transducción de Señal , Fracciones Subcelulares/metabolismo
19.
J Biol Chem ; 285(8): 5122-6, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20038585

RESUMEN

Zipper-interacting protein kinase (ZIPK) is a member of the death-associated protein kinase family associated with apoptosis in nonmuscle cells where it phosphorylates myosin regulatory light chain (RLC) to promote membrane blebbing. ZIPK mRNA and protein are abundant in heart tissue and isolated ventricular neonatal rat cardiac myocytes. An unbiased substrate search performed with purified ZIPK on heart homogenates led to the discovery of a prominent 20-kDa protein substrate identified as RLC of ventricular myosin. Biochemical analyses showed ZIPK phosphorylated cardiac RLC at Ser-15 with a V(max) value 2-fold greater than the value for smooth/nonmuscle RLC; cardiac RLC is a favorable biochemical substrate. Knockdown of ZIPK in cardiac myocytes by small interfering RNA significantly decreased the extent of RLC Ser-15 phosphorylation. Thus, ZIPK may act as a cardiac RLC kinase and thereby affect contractility.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Ventrículos Cardíacos/metabolismo , Mioblastos Cardíacos/metabolismo , Contracción Miocárdica/fisiología , Cadenas Ligeras de Miosina/metabolismo , Animales , Proteínas Quinasas Asociadas a Muerte Celular , Ratones , Especificidad de Órganos/fisiología , Fosforilación/fisiología , Ratas
20.
Proc Natl Acad Sci U S A ; 105(28): 9745-50, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18621691

RESUMEN

A missense mutation in the alphaB-crystallin (CryAB) gene triggers a severe form of desmin-related cardiomyopathy (DRCM) characterized by accumulation of misfolded proteins. We hypothesized that autophagy increases in response to protein aggregates and that this autophagic activity is adaptive. Mutant CryAB (CryAB(R120G)) triggered a >2-fold increase in cardiomyocyte autophagic activity, and blunting autophagy increased the rate of aggregate accumulation and the abundance of insoluble CryAB(R120G)-associated aggregates. Cardiomyocyte-restricted overexpression of CryAB(R120G) in mice induced intracellular aggregate accumulation and systolic heart failure by 12 months. As early as 2 months (well before the earliest declines in cardiac function), we detected robust autophagic activity. To test the functional significance of autophagic activation, we crossed CryAB(R120G) mice with animals harboring heterozygous inactivation of beclin 1, a gene required for autophagy. Blunting autophagy in vivo dramatically hastened heart failure progression with a 3-fold increase in interstitial fibrosis, greater accumulation of polyubiquitinated proteins, larger and more extensive intracellular aggregates, accelerated ventricular dysfunction, and early mortality. This study reports activation of autophagy in DRCM. Further, our findings point to autophagy as an adaptive response in this proteotoxic form of heart disease.


Asunto(s)
Autofagia/fisiología , Cardiomiopatías/etiología , Desmina , Cadena B de alfa-Cristalina/genética , Animales , Fibrosis , Ratones , Ratones Mutantes , Mutación Missense , Miocitos Cardíacos , Poliubiquitina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA