Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Lab Invest ; 99(3): 387-398, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30237457

RESUMEN

Polymerase delta-interacting protein 2 (Poldip2) is a multi-functional protein with numerous roles in the vasculature, including the regulation of cell apoptosis and migration, as well as extracellular matrix deposition; however, its role in VSMC proliferation and neointimal formation is unknown. In this study, we investigated the role of Poldip2 in intraluminal wire-injury induced neointima formation and proliferation of vascular smooth muscle cells in vitro and in vivo. Poldip2 expression was observed in the intima and media of human atherosclerotic arteries, where it colocalized with proliferating cell nuclear antigen (PCNA). Wire injury of femoral arteries of Poldip2+/+ mice induced robust neointimal formation after 2 weeks, which was impaired in Poldip2+/‒ mice. PCNA expression was significantly reduced and expression of the cell cycle inhibitor p21 was significantly increased in wire-injured arteries of Poldip2+/‒ animals compared to wild-type controls. No difference was observed in apoptosis. Downregulation of Poldip2 in rat aortic smooth muscle cells significantly reduced serum-induced proliferation and PCNA expression, but upregulated p21 expression. Downregulation of p21 using siRNA reversed the inhibition of proliferation induced by knockdown of Poldip2. These results indicate that Poldip2 plays a critical role in the proliferation of VSMCs.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Proliferación Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Mitocondriales/deficiencia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Neointima/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/deficiencia , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proliferación Celular/genética , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Neointima/patología , Neointima/prevención & control , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , ARN Interferente Pequeño/genética , Ratas , Superóxidos/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 38(10): 2423-2434, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354218

RESUMEN

Objective- Actin cytoskeleton assembly and organization, as a result of focal adhesion (FA) formation during cell adhesion, are dependent on reactive oxygen species and the cellular redox environment. Poldip2 (polymerase δ-interacting protein 2), a novel regulator of NOX4 (NADPH oxidase 4), plays a significant role in reactive oxygen species production and cytoskeletal remodeling. Thus, we hypothesized that endogenous reactive oxygen species derived from Poldip2/NOX4 contribute to redox regulation of actin and cytoskeleton assembly during integrin-mediated cell adhesion. Approach and Results- Using vascular smooth muscle cells, we verified that hydrogen peroxide (H2O2) levels increase during integrin-mediated cell attachment as a result of activation of NOX4. Filamentous actin (F-actin) was oxidized by sulfenylation during cell attachment, with a peak at 3 hours (0.80±0.04 versus 0.08±0.13 arbitrary units at time zero), which was enhanced by overexpression of Poldip2. Depletion of Poldip2 or NOX4 using siRNA, or scavenging of endogenous H2O2 with catalase, inhibited F-actin oxidation by 78±26%, 99±1%, and 98±1%, respectively. To determine the consequence of F-actin oxidation, we examined the binding of F-actin to vinculin, a protein involved in FA complexes that regulates FA maturation. Vinculin binding during cell adhesion as well as migration capacity were inhibited after transfection with actin containing 2 oxidation-resistant point mutations (C272A and C374A). Silencing of Poldip2 or NOX4 also impaired actin-vinculin interaction, which disturbed maturation of FAs and inhibited cell migration. Conclusions- These results suggest that integrin engagement during cell attachment activates Poldip2/Nox4 to oxidize actin, which modulates FA assembly.


Asunto(s)
Citoesqueleto de Actina/enzimología , Proteínas Portadoras/metabolismo , Adhesión Celular , Integrinas/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , NADPH Oxidasa 4/metabolismo , Proteínas Nucleares/metabolismo , Vinculina/metabolismo , Citoesqueleto de Actina/genética , Animales , Proteínas Portadoras/genética , Movimiento Celular , Células Cultivadas , Humanos , Peróxido de Hidrógeno/metabolismo , Músculo Liso Vascular/ultraestructura , Miocitos del Músculo Liso/ultraestructura , NADPH Oxidasa 4/genética , Proteínas Nucleares/genética , Oxidación-Reducción , Ratas , Transducción de Señal
3.
Arterioscler Thromb Vasc Biol ; 35(5): 1198-206, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25814672

RESUMEN

OBJECTIVE: Focal adhesions (FAs) link the cytoskeleton to the extracellular matrix and as such play important roles in growth, migration, and contractile properties of vascular smooth muscle cells. Recently, it has been shown that downregulation of Nox4, a transforming growth factor (TGF) ß-inducible, hydrogen peroxide (H2O2)-producing enzyme, affects the number of FAs. However, the effectors downstream of Nox4 that mediate FA regulation are unknown. The FA resident protein H2O2-inducible clone (Hic)-5 is H2O2 and TGFß inducible, and a binding partner of the heat shock protein (Hsp) 27. The objective of this study was to elucidate the mechanism, by which Hic-5 and Hsp27 participate in TGFß-induced, Nox4-mediated vascular smooth muscle cell adhesion and migration. APPROACH AND RESULTS: Through a combination of molecular biology and biochemistry techniques, we found that TGFß, by a Nox4-dependent mechanism, induces the expression and interaction of Hic-5 and Hsp27, which is essential for Hic-5 localization to FAs. Importantly, we found that Hic-5 expression is required for the TGFß-mediated increase in FA number, adhesive forces and migration. Mechanistically, Nox4 downregulation impedes Smad (small body size and mothers against decapentaplegic) signaling by TGFß, and Hsp27 and Hic-5 upregulation by TGFß is blocked in small body size and mothers against decapentaplegic 4-deficient cells. CONCLUSIONS: Hic-5 and Hsp27 are effectors of Nox4 required for TGFß-stimulated FA formation, adhesion strength and migration in vascular smooth muscle cell.


Asunto(s)
Proteínas de Choque Térmico HSP27/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/metabolismo , NADPH Oxidasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adhesión Celular/genética , Adhesión Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Células Cultivadas , Adhesiones Focales/genética , Adhesiones Focales/fisiología , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Músculo Liso Vascular/citología , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Sensibilidad y Especificidad , Transducción de Señal
4.
Am J Physiol Heart Circ Physiol ; 307(7): H945-57, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25063792

RESUMEN

Polymerase-δ-interacting protein 2 (Poldip2) interacts with NADPH oxidase 4 (Nox4) and regulates migration; however, the precise underlying mechanisms are unclear. Here, we investigated the role of Poldip2 in focal adhesion turnover, as well as traction force generation and polarization. Poldip2 overexpression (AdPoldip2) in vascular smooth muscle cells (VSMCs) impairs PDGF-induced migration and induces a characteristic phenotype of long cytoplasmic extensions. AdPoldip2 also prevents the decrease in spreading and increased aspect ratio observed in response to PDGF and slightly impairs cell contraction. Moreover, AdPoldip2 blocks focal adhesion dissolution and sustains H2O2 levels in focal adhesions, whereas Poldip2 knockdown (siPoldip2) significantly decreases the number of focal adhesions. RhoA activity is unchanged when focal adhesion dissolution is stimulated in control cells but increases in AdPoldip2-treated cells. Inhibition of RhoA blocks Poldip2-mediated attenuation of focal adhesion dissolution, and overexpression of RhoA or focal adhesion kinase (FAK) reverses the loss of focal adhesions induced by siPoldip2, indicating that RhoA and FAK mediate the effect of Poldip2 on focal adhesions. Nox4 silencing prevents focal adhesion stabilization by AdPoldip2 and induces a phenotype similar to siPoldip2, suggesting a role for Nox4 in Poldip2-induced focal adhesion stability. As a consequence of impaired focal adhesion turnover, PDGF-treated AdPoldip2 cells are unable to reduce and polarize traction forces, a necessary first step in migration. These results implicate Poldip2 in VSMC migration via regulation of focal adhesion turnover and traction force generation in a Nox4/RhoA/FAK-dependent manner.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular , Adhesiones Focales/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Proteínas Portadoras/genética , Adhesión Celular , Polaridad Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Peróxido de Hidrógeno/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Proteína de Unión al GTP rhoA/metabolismo
5.
Circ Res ; 111(1): 56-65, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22619279

RESUMEN

RATIONALE: The type I subclass of coronins, a family of actin-binding proteins, regulates various actin-dependent cellular processes, including migration. However, the existence and role of coronins in vascular smooth muscle cell (VSMC) migration has yet to be determined. OBJECTIVE: The goal of the present study was to define the mechanism by which coronins regulate platelet-derived growth factor (PDGF)-induced VSMC migration. METHODS AND RESULTS: Coronin 1B (Coro1B) and 1C (Coro1C) were both found to be expressed in VSMCs at the mRNA and protein levels. Downregulation of Coro1B by siRNA increases PDGF-induced migration, while downregulation of Coro1C has no effect. We confirmed through kymograph analysis that the Coro1B-downregulation-mediated increase in migration is directly linked to increased lamellipodial protraction rate and protrusion distance in VSMC. In other cell types, coronins exert their effects on lamellipodia dynamics by an inhibitory interaction with the ARP2/3 complex, which is disrupted by the phosphorylation of Coro1B. We found that PDGF induces phosphorylation of Coro1B on serine-2 via PKCε, leading to a decrease in the interaction of Coro1B with the ARP2/3 complex. VSMCs transfected with a phosphodeficient S2A Coro1B mutant showed decreased migration in response to PDGF, suggesting that the phosphorylation of Coro1B is required for the promotion of migration by PDGF. In both the rat and mouse, Coro1B phosphorylation was increased in response to vessel injury in vivo. CONCLUSIONS: Our data suggest that phosphorylation of Coro1B and the subsequent reduced interaction with ARP2/3 complex participate in PDGF-induced VSMC migration, an important step in vascular lesion formation.


Asunto(s)
Movimiento Celular , Proteínas de Microfilamentos/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Animales , Becaplermina , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Quimografía , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Músculo Liso Vascular/patología , Mutación , Miocitos del Músculo Liso/patología , Neointima , Fosforilación , Proteína Quinasa C-epsilon/metabolismo , Seudópodos/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Serina , Transducción de Señal , Factores de Tiempo , Transfección
6.
Am J Physiol Heart Circ Physiol ; 305(5): H747-55, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23812391

RESUMEN

Vascular diseases frequently accompany diabetes mellitus. Based on the current understanding of atherosclerosis as an inflammatory disorder of the vascular wall, it has been speculated that diabetes may accelerate atherosclerosis by inducing a proinflammatory milieu in the vasculature. ANG II and bone morphogenic proteins (BMPs) have been implicated in vascular inflammation. We evaluated the effect of angiotensin receptor blockade by valsartan and BMP inhibition by noggin on markers of vascular inflammation in a mouse model of diabetes. Noggin had no effect on blood pressure but decreased serum glucose levels, whereas valsartan significantly decreased blood pressure, but not serum glucose. Both inhibitors reduced reactive oxygen species production in the aorta. Additionally, noggin and valsartan diminish gene transcription and protein expression of various inflammatory molecules in the vascular wall. These observations indicate that although both inhibitors block superoxide production and have similar effects on inflammatory gene expression, glycemia and blood pressure may represent a secondary target differentially affected by noggin and valsartan. Our data clearly identify the BMP pathway as a potentially potent therapeutic target in diabetic inflammatory vascular disease.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Portadoras/uso terapéutico , Diabetes Mellitus Tipo 2/complicaciones , Hiperglucemia/prevención & control , Vasculitis/prevención & control , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Glucemia/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Proteínas Portadoras/farmacología , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Hiperglucemia/fisiopatología , Masculino , Ratones , Ratones Mutantes , Especies Reactivas de Oxígeno/metabolismo , Tetrazoles/farmacología , Tetrazoles/uso terapéutico , Valina/análogos & derivados , Valina/farmacología , Valina/uso terapéutico , Valsartán , Vasculitis/fisiopatología
7.
Nat Genet ; 33(1): 23-4, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12496758

RESUMEN

Human mitochondrial transcription factor B1 (h-mtTFB1) has an unprecedented relationship to RNA methyltransferases. Here, we show that this protein methylates a conserved stem-loop in bacterial 16S rRNA and that the homologous sequence in the human mitochondrial 12S molecule is similarly modified. Thus, h-mtTFB1 appears to be dual-function protein, acting both as a transcription factor and an rRNA-modification enzyme.


Asunto(s)
Aminoglicósidos , Mitocondrias/genética , Conformación de Ácido Nucleico , ARN Ribosómico/química , ARN Ribosómico/metabolismo , Factores de Transcripción/metabolismo , Antibacterianos/farmacología , Secuencia de Bases , Secuencia Conservada , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Escherichia coli/genética , Eliminación de Gen , Humanos , Metilación , Metiltransferasas/genética , ARN Bacteriano/química , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , ARN Ribosómico/genética , ARN Ribosómico 16S/química , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Factores Estimuladores hacia 5'
8.
Am J Physiol Cell Physiol ; 301(6): C1415-22, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21900690

RESUMEN

Angiotensin II (ANG II) has been implicated in the pathogenesis of diabetic micro- and macrovascular disease. In vascular smooth muscle cells (VSMCs), ANG II phosphorylates and degrades insulin receptor substrate-1 (IRS-1). While the pathway responsible for IRS-1 degradation in this system is unknown, c-Jun NH(2)-terminal kinase (JNK) has been linked with serine phosphorylation of IRS-1 and insulin resistance. We investigated the role of JNK in ANG II-induced IRS-1 phosphorylation, degradation, Akt activation, glucose uptake, and hypertrophic signaling, focusing on three IRS-1 phosphorylation sites: Ser302, Ser307, and Ser632. Maximal IRS-1 phosphorylation on Ser632 occurred at 5 min, on Ser307 at 30 min, and on Ser302 at 60 min. The JNK inhibitor SP600125 reduced ANG II-induced IRS-1 Ser307 phosphorylation (by 80%), IRS-1 Ser302 phosphorylation (by 70%), and IRS-1 Ser632 phosphorylation (by 50%). However, JNK inhibition had no effect on ANG II-mediated IRS-1 degradation, nor did it reverse the ANG II-induced decrease in Akt phosphorylation or glucose uptake. Transfection of VSMCs with mutants S307A, S302A, or S632A of IRS-1 did not block ANG II-mediated IRS-1 degradation. In contrast, JNK inhibition attenuated insulin-induced upregulation of collagen and smooth muscle α-actin in ANG II-pretreated cells. We conclude that phosphorylation of Ser307, Ser302, and Ser632 of IRS-1 is not involved in ANG II-mediated IRS-1 degradation, and that JNK alone does not mediate ANG II-stimulated IRS-1 degradation, but rather is responsible for the hypertrophic effects of insulin on smooth muscle.


Asunto(s)
Angiotensina II/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Transducción de Señal/fisiología , Actinas , Animales , Western Blotting , Inhibidores Enzimáticos/farmacología , Hipertrofia/metabolismo , Inmunoprecipitación , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Fosforilación , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transfección
9.
Circ Res ; 105(3): 249-59, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19574552

RESUMEN

RATIONALE: NADPH oxidases (Noxes) regulate vascular physiology and contribute to the pathogenesis of vascular disease. In vascular smooth muscle cells (VSMCs), the interactions of individual Nox homologs with regulatory proteins are poorly defined. OBJECTIVE: The objective of this study was to identify novel NADPH oxidase regulatory proteins. METHODS AND RESULTS: Using a yeast 2-hybrid screen, we identified a novel p22phox binding partner, Poldip2, and demonstrated that it associates with p22phox, NADPH oxidase (Nox)1, and Nox4 and colocalizes with p22phox at sites of Nox4 localization. Poldip2 increases Nox4 enzymatic activity by 3-fold and positively regulates basal reactive oxygen species production in VSMCs (O2(.-): 86.3+/-15.6% increase; H2O2: 40.7+/-4.5% increase). Overexpression of Poldip2 activates Rho (180.2+/-24.8% increase), strengthens focal adhesions, and increases stress fiber formation. These phenotypic changes are blocked by dominant negative Rho. In contrast, depletion of either Poldip2 or Nox4 results in a loss of these structures, which is rescued by adding back active Rho. Cell migration, which requires dynamic cytoskeletal remodeling, is impaired by either excess (70.1+/-14.7% decrease) or insufficient Poldip2 (63.5+/-5.9% decrease). CONCLUSIONS: These results suggest that Poldip2 associates with p22phox to activate Nox4, leading to regulation of focal adhesion turnover and VSMC migration, thus linking reactive oxygen species production and cytoskeletal remodeling. Poldip2 may be a novel therapeutic target for vascular pathologies with a significant VSMC migratory component, such as restenosis and atherosclerosis.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Citoesqueleto/metabolismo , Músculo Liso Vascular/metabolismo , NADPH Oxidasas/metabolismo , Proteínas Nucleares/metabolismo , Animales , Movimiento Celular/fisiología , Células Cultivadas , Humanos , Músculo Liso Vascular/citología , NADPH Oxidasa 4 , Oxidación-Reducción , Ratas , Especies Reactivas de Oxígeno/metabolismo , Saccharomyces cerevisiae , Proteína de Unión al GTP rhoA/metabolismo
10.
Free Radic Biol Med ; 45(3): 329-35, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18466778

RESUMEN

The proliferation of vascular smooth muscle cells is important in the pathogenesis of many vascular diseases. Reactive oxygen species (ROS) produced by NADPH oxidases in smooth muscle cells have been shown to participate in signaling cascades regulating proliferation induced by platelet-derived growth factor (PDGF), a powerful smooth muscle mitogen. We sought to determine the role of Nox5 in the regulation of PDGF-stimulated human aortic smooth muscle cell (HASMC) proliferation. Cultured HASMC were found to express four isoforms of Nox5. When HASMC stimulated with PDGF were pretreated with N-acetyl cysteine (NAC), proliferation was significantly reduced. Proliferation induced by PDGF was also heavily dependent on JAK/STAT activation, as the JAK inhibitor, AG490, was able to completely abolish PDGF-stimulated HASMC growth. Specific knockdown of Nox5 with a siRNA strategy reduced PDGF-induced HASMC ROS production and proliferation. Additionally, siRNA to Nox5 inhibited PDGF-stimulated JAK2 and STAT3 phosphorylation. ROS produced by Nox5 play an important role in PDGF-induced JAK/STAT activation and HASMC proliferation.


Asunto(s)
Aorta/metabolismo , Proteínas de la Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Aorta/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Janus Quinasa 2/efectos de los fármacos , Janus Quinasa 2/metabolismo , Proteínas de la Membrana/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , NADPH Oxidasa 5 , NADPH Oxidasas/efectos de los fármacos , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Transfección
11.
Mol Cell Biol ; 22(4): 1116-25, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11809803

RESUMEN

A critical step toward understanding mitochondrial genetics and its impact on human disease is to identify and characterize the full complement of nucleus-encoded factors required for mitochondrial gene expression and mitochondrial DNA (mtDNA) replication. Two factors required for transcription initiation from a human mitochondrial promoter are h-mtRNA polymerase and the DNA binding transcription factor, h-mtTFA. However, based on studies in model systems, the existence of a second human mitochondrial transcription factor has been postulated. Here we report the isolation of a cDNA encoding h-mtTFB, the human homolog of Saccharomyces cerevisiae mitochondrial transcription factor B (sc-mtTFB) and the first metazoan member of this class of transcription factors to which a gene has been assigned. Recombinant h-mtTFB is capable of binding mtDNA in a non-sequence-specific fashion and activates transcription from the human mitochondrial light-strand promoter in the presence of h-mtTFA in vitro. Remarkably, h-mtTFB and its fungal homologs are related in primary sequence to a superfamily of N6 adenine RNA methyltransferases. This observation, coupled with the ability of recombinant h-mtTFB to bind S-adenosylmethionine in vitro, suggests that a structural, and perhaps functional, relationship exists between this class of transcription factors and this family of RNA modification enzymes and that h-mtTFB may perform dual functions during mitochondrial gene expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Metiltransferasas/metabolismo , Mitocondrias/fisiología , Proteínas Mitocondriales , Proteínas Nucleares , S-Adenosilmetionina/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Proteínas Luminiscentes/metabolismo , Metiltransferasas/genética , Microscopía Fluorescente , Mitocondrias/química , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/metabolismo , S-Adenosilmetionina/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Factores de Transcripción/química , Factores de Transcripción/genética
12.
Nucleic Acids Res ; 30(9): 1929-34, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11972329

RESUMEN

The steady-state amounts of mitochondrial transcripts and transcription proteins were analyzed during mtDNA depletion and subsequent repletion to gain insight into the regulation of human mitochondrial gene expression. As documented previously, HeLa cells depleted of mtDNA via treatment with ethidium bromide (EB) were found to contain reduced steady-state levels of the mitochondrial transcription factor h-mtTFA. When partially mtDNA-depleted cells were cultured in the absence of EB, h-mtTFA recovered to normal levels at a significantly slower rate than mtDNA. Human mtRNA polymerase exhibited a similar depletion-repletion profile, suggesting that the mitochondrial transcription machinery is coordinately regulated in response to changes in mtDNA copy number. Newly synthesized mitochondrial transcripts were detected early in the recovery phase, despite the fact that mtDNA, h-mtTFA and h-mtRNA polymerase were simultaneously depleted. Although delayed relative to mtDNA, the amounts of h-mtTFA and h-mtRNA polymerase sharply increased during the later stages of the recovery phase, which was accompanied by accelerated rates of transcription and mtDNA replication. Altogether, these data indicate that when mtDNA copy number is low, it is beneficial to prevent accumulation of mitochondrial transcription proteins. In addition, h-mtTFA and h-mtRNA polymerase are either normally present in excess of the amount required for transcription or their activity is up-regulated to ensure continued expression and transcription-dependent replication of the mitochondrial genome during mtDNA-depleted states.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Nucleares , ARN/biosíntesis , Transcripción Genética , Replicación del ADN , Proteínas de Unión al ADN/biosíntesis , ARN Polimerasas Dirigidas por ADN/metabolismo , Etidio/farmacología , Células HeLa , Humanos , Cinética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/biosíntesis , ARN Mitocondrial , Factores de Transcripción/biosíntesis , Regulación hacia Arriba , Proteínas de Unión al GTP rho/genética
13.
PLoS One ; 8(11): e79657, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24236150

RESUMEN

In adult tissue, vascular smooth muscle cells (VSMCs) exist in a differentiated phenotype, which is defined by the expression of contractile proteins and lack of proliferation. After vascular injury, VSMC adopt a synthetic phenotype associated with proliferation, migration and matrix secretion. The transition between phenotypes is a consequence of the extracellular environment, and in particular, is regulated by agonists such as the pro-differentiating cytokine transforming growth factor ß (TGFß) and the pro-proliferative cytokine platelet derived growth factor (PDGF). In this study, we investigated the interplay between TGFß and PDGF with respect to their ability to regulate VSMC proliferation. Stimulation of human aortic VSMC with TGFß completely blocked proliferation induced by all isoforms of PDGF, as measured by DNA synthesis and total cell number. Mechanistically, PDGF-induced Cyclin D1 mRNA and protein expression was inhibited by TGFß. TGFß had no effect on PDGF activation of its receptor and ERK1/2, but inhibited Akt activation. However, constitutively active Akt did not reverse the inhibitory effect of TGFß on Cyclin D1 expression even though inhibition of the proteasome blocked the effect of TGFß. siRNA against Smad4 completely reversed the inhibitory effect of TGFß on PDGF-induced Cyclin D1 expression and restored proliferation in response to PDGF. Moreover, siRNA against KLF5 prevented Cyclin D1 upregulation by PDGF and overexpression of KLF5 partially reversed TGFß-induced inhibition of Cyclin D1 expression. Taken together, our results demonstrate that KLF5 is required for PDGF-induced Cyclin D1 expression, which is inhibited by TGFß via a Smad dependent mechanism, resulting in arrest of VSMCs in the G1 phase of the cell cycle.


Asunto(s)
Ciclina D1/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Adolescente , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Proteolisis , Transducción de Señal/efectos de los fármacos , Proteína Smad4/metabolismo , Transcripción Genética
14.
PLoS One ; 7(3): e33211, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427991

RESUMEN

Senescence, a hallmark of mammalian aging, is associated with the onset and progression of cardiovascular disease. Angiotensin II (Ang II) signaling and zinc homeostasis dysfunction are increased with age and are linked to cardiovascular disease, but the relationship among these processes has not been investigated. We used a model of cellular senescence induced by Ang II in vascular smooth muscle cells (VSMCs) to explore the role of zinc in vascular dysfunction. We found that Ang II-induced senescence is a zinc-dependent pathway mediated by the downregulation of the zinc transporters ZnT3 and ZnT10, which work to reduce cytosolic zinc. Zinc mimics Ang II by increasing reactive oxygen species (ROS), activating NADPH oxidase activity and Akt, and by downregulating ZnT3 and ZnT10 and inducing senescence. Zinc increases Ang II-induced senescence, while the zinc chelator TPEN, as well as overexpression of ZnT3 or ZnT10, decreases ROS and prevents senescence. Using HEK293 cells, we found that ZnT10 localizes in recycling endosomes and transports zinc into vesicles to prevent zinc toxicity. Zinc and ZnT3/ZnT10 downregulation induces senescence by decreasing the expression of catalase. Consistently, ZnT3 and ZnT10 downregulation by siRNA increases ROS while downregulation of catalase by siRNA induces senescence. Zinc, siZnT3 and siZnT10 downregulate catalase by a post-transcriptional mechanism mediated by decreased phosphorylation of ERK1/2. These data demonstrate that zinc homeostasis dysfunction by decreased expression of ZnT3 or ZnT10 promotes senescence and that Ang II-induced senescence is a zinc and ROS-dependent process. Our studies suggest that zinc might also affect other ROS-dependent processes induced by Ang II, such as hypertrophy and migration of smooth muscle cells.


Asunto(s)
Angiotensina II/farmacología , Proteínas de Transporte de Catión/metabolismo , Senescencia Celular/fisiología , Regulación hacia Abajo/efectos de los fármacos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Zinc/metabolismo , Angiotensina II/metabolismo , Animales , Western Blotting , Senescencia Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Masculino , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasas/metabolismo , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Free Radic Biol Med ; 50(2): 354-62, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21074607

RESUMEN

In contrast to other cell types, vascular smooth muscle cells modify their phenotype in response to external signals. NADPH oxidase 4 (Nox4) is critical for maintenance of smooth muscle gene expression; however, the underlying mechanisms are incompletely characterized. Using smooth muscle α-actin (SMA) as a prototypical smooth muscle gene and transforming growth factor-ß (TGF-ß) as a differentiating agent, we examined Nox4-dependent signaling. TGF-ß increases Nox4 expression and activity in human aortic smooth muscle cells (HASMC). Transfection of HASMC with siRNA against Nox4 (siNox4) abolishes TGF-ß-induced SMA expression and stress fiber formation. siNox4 also significantly inhibits TGF-ß-stimulated p38MAPK phosphorylation, as well as that of its substrate, mitogen-activated protein kinase-activated protein kinase-2. Moreover, the p38MAPK inhibitor SB-203580 nearly completely blocks the SMA increase induced by TGF-ß. Inhibition of either p38MAPK or NADPH oxidase-derived reactive oxygen species impairs the TGF-ß-induced phosphorylation of Ser103 on serum response factor (SRF) and reduces its transcriptional activity. Binding of SRF to myocardin-related transcription factor (MRTF) is also necessary, because downregulation of MRTF by siRNA abolishes TGF-ß-induced SMA expression. Taken together, these data suggest that Nox4 regulates SMA expression via activation of a p38MAPK/SRF/MRTF pathway in response to TGF-ß.


Asunto(s)
Actinas/metabolismo , NADPH Oxidasas/metabolismo , Factor de Respuesta Sérica/metabolismo , Fibras de Estrés/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Actinas/genética , Aorta/citología , Aorta/metabolismo , Western Blotting , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Imidazoles/farmacología , Luciferasas/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Fosforilación/efectos de los fármacos , Piridinas/farmacología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Respuesta Sérica/genética , Transducción de Señal/efectos de los fármacos , Fibras de Estrés/patología , Factor de Crecimiento Transformador beta/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA