Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Cardiovasc Disord ; 17(1): 271, 2017 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-29047330

RESUMEN

BACKGROUND: The NR4A3 orphan nuclear hormone receptor, NOR1, functions as a constitutively active transcription factor to regulate inflammation, proliferation, and cell survival during pathological vascular remodeling. Inflammatory processes represent key mechanisms leading to abdominal aortic aneurysm (AAA) formation. However, a role of NOR1 in AAA formation has not been investigated previously. METHODS: Inflammatory gene expression was analyzed in bone marrow-derived macrophages isolated from NOR1-deficient mice. Low-density lipoprotein receptor-deficient (LDLr-/-) mice were irradiated and reconstituted with hematopoietic stem cells obtained from NOR1-/- or wild-type littermate mice. Animals were infused with angiotensin II and fed a diet enriched in saturated fat to induce AAA formation. Quantification of AAA formation was performed by ultrasound and ex vivo measurements. RESULTS: Among 184 inflammatory genes that were analyzed, 36 genes were differentially regulated in LPS-treated NOR1-deficient macrophages. Albeit this difference in gene regulation, NOR1-deficiency in hematopoietic stem cells did not affect development of AAA formation in bone marrow-derived stem cell transplanted LDLr-deficient mice. CONCLUSION: NOR1 deletion induced differential inflammatory gene transcription in macrophages but did not influence AAA formation in mice.


Asunto(s)
Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Receptores de Esteroides/deficiencia , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/deficiencia , Receptores de Hormona Tiroidea/genética , Animales , Inflamación/genética , Inflamación/metabolismo , Inflamación/prevención & control , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Arterioscler Thromb Vasc Biol ; 37(2): 301-311, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27932351

RESUMEN

OBJECTIVE: Aberrant proliferation of smooth muscle cells (SMC) in response to injury induces pathological vascular remodeling during atherosclerosis and neointima formation. Telomerase is rate limiting for tissue renewal and cell replication; however, the physiological role of telomerase in vascular diseases remains to be determined. The goal of the present study was to determine whether telomerase reverse transcriptase (TERT) affects proliferative vascular remodeling and to define the molecular mechanism by which TERT supports SMC proliferation. APPROACH AND RESULTS: We first demonstrate high levels of TERT expression in replicating SMC of atherosclerotic and neointimal lesions. Using a model of guidewire-induced arterial injury, we demonstrate decreased neointima formation in TERT-deficient mice. Studies in SMC isolated from TERT-deficient and TERT overexpressing mice with normal telomere length established that TERT is necessary and sufficient for cell proliferation. TERT deficiency did not induce a senescent phenotype but resulted in G1 arrest albeit hyperphosphorylation of the retinoblastoma protein. This proliferative arrest was associated with stable silencing of the E2F1-dependent S-phase gene expression program and not reversed by ectopic overexpression of E2F1. Finally, chromatin immunoprecipitation and accessibility assays revealed that TERT is recruited to E2F1 target sites and promotes chromatin accessibility for E2F1 by facilitating the acquisition of permissive histone modifications. CONCLUSIONS: These data indicate a previously unrecognized role for TERT in neointima formation through epigenetic regulation of proliferative gene expression in SMC.


Asunto(s)
Aterosclerosis/enzimología , Ensamble y Desensamble de Cromatina , Factor de Transcripción E2F1/metabolismo , Silenciador del Gen , Músculo Liso Vascular/enzimología , Neointima , Telomerasa/deficiencia , Telomerasa/metabolismo , Lesiones del Sistema Vascular/enzimología , Acetilación , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Sitios de Unión , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Factor de Transcripción E2F1/genética , Arteria Femoral/enzimología , Arteria Femoral/lesiones , Arteria Femoral/patología , Puntos de Control de la Fase G1 del Ciclo Celular , Predisposición Genética a la Enfermedad , Histonas/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Fenotipo , Fosforilación , Unión Proteica , Interferencia de ARN , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Telomerasa/genética , Factores de Tiempo , Transfección , Remodelación Vascular , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/patología
3.
JACC Basic Transl Sci ; 1(1-2): 49-60, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27127803

RESUMEN

OBJECTIVES: The present study sought to investigate the mechanisms underlying the mitogenic function of telomerase and to test the hypothesis that everolimus, commonly used on drug-eluting stents, suppresses smooth muscle cells (SMC) proliferation by targeting telomerase. BACKGROUND: Proliferation of SMC during neointima formation is prevented by drug-eluting stents. Although the replicative capacity of mammalian cells is enhanced by telomerase expression, the contribution of telomerase to the proliferative response underlying neointima formation and its potential role as a pharmacological target remain to be investigated. METHODS: We first employed constitutive expression of telomerase reverse transcriptase (TERT) in cell systems to study transcriptional mechanisms by which telomerase activates a mitogenic program. Second, overexpression of telomerase in mice provided a model to study the role of telomerase as a drug target for the antiproliferative efficacy of everolimus. RESULTS: Inhibition of neointima formation by everolimus is lost in mice overexpressing TERT, indicating that repression of telomerase confers the antiproliferative efficacy of everolimus. Everolimus reduces TERT expression in SMC through an Ets-1-dependent inhibition of promoter activation. The inhibition of TERT-dependent SMC proliferation by everolimus occurred in the absence of telomere shortening but rather as a result of a G1→S phase arrest. Although everolimus failed to inhibit phosphorylation of the retinoblastoma protein as the gatekeeper of S-phase entry, it potently repressed downstream target genes. Using chromatin immunoprecipitation assays, we finally demonstrate that TERT induces E2F binding to S-phase gene promoters and supports histone acetylation, effects that are inhibited by everolimus and mediate its antiproliferative activity. CONCLUSIONS: These results characterize telomerase as a previously unrecognized target for the antiproliferative activity of everolimus. Our studies further identify a novel mitogenic pathway in SMC, which depends on the epigenetic activation of S-phase gene promoters by TERT.

4.
J Cell Physiol ; 231(6): 1276-82, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26505494

RESUMEN

Telomerase reverse transcriptase (TERT) maintains telomeres and is rate limiting for replicative life span. While most somatic tissues silence TERT transcription resulting in telomere shortening, cells derived from cancer or cardiovascular diseases express TERT and activate telomerase. In the present study, we demonstrate that histone deacetylase (HDAC) inhibition induces TERT transcription and promoter activation. At the protein level in contrast, HDAC inhibition decreases TERT protein abundance through enhanced degradation, which decreases telomerase activity and induces senescence. Finally, we demonstrate that HDAC inhibition decreases TERT expression during vascular remodeling in vivo. These data illustrate a differential regulation of TERT transcription and protein stability by HDAC inhibition and suggest that TERT may constitute an important target for the anti-proliferative efficacy of HDAC inhibitors.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Hidroxilaminas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Regiones Promotoras Genéticas , Quinolinas/farmacología , Telomerasa/metabolismo , Activación Transcripcional/efectos de los fármacos , Animales , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/genética , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Neointima , Proteolisis , Interferencia de ARN , Ratas , Telomerasa/genética , Transfección , Remodelación Vascular/efectos de los fármacos , Lesiones del Sistema Vascular/tratamiento farmacológico , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
5.
FEBS Lett ; 588(24): 4825-30, 2014 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-25451221

RESUMEN

The nuclear receptor NOR1 is an immediate-early response gene implicated in the transcriptional control of proliferation. Since the expression level of NOR1 is rapidly induced through cAMP response element binding (CREB) protein-dependent promoter activation, we investigated the contribution of histone acetylation to this transient induction. We demonstrate that NOR1 transcription is induced by histone deacetylase (HDAC) inhibition and by depletion of HDAC1 and HDAC3. HDAC inhibition activated the NOR1 promoter, increased histone acetylation and augmented the recruitment of phosphorylated CREB to the promoter. Furthermore, HDAC inhibition increased Ser133 phosphorylation of CREB and augmented NOR1 protein stability. These data outline previously unrecognized mechanisms of NOR1 regulation and illustrate a key role for histone acetylation in the rapid induction of NOR1.


Asunto(s)
Proteínas de Unión al ADN/genética , Epigénesis Genética , Histonas/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores Nucleares Huérfanos/genética , Acetilación/efectos de los fármacos , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Estabilidad Proteica/efectos de los fármacos , Ratas
6.
Stem Cells ; 32(9): 2419-29, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24806827

RESUMEN

The NR4A orphan nuclear receptor NOR1 functions as a constitutively active transcription factor regulating cellular inflammation and proliferation. In this study, we used bone marrow transplantation to determine the selective contribution of NOR1 expression in hematopoietic stem cells to the development of atherosclerosis. Reconstitution of lethally irradiated apoE(-/-) mice with NOR1-deficient hematopoietic stem cells accelerated atherosclerosis formation and macrophage recruitment following feeding a diet enriched in saturated fat. NOR1 deficiency in hematopoietic stem cells induced splenomegaly and monocytosis, specifically the abundance of inflammatory Ly6C(+) monocytes. Bone marrow transplantation studies further confirmed that NOR1 suppresses the proliferation of macrophage and dendritic progenitor (MDP) cells. Expression analysis identified RUNX1, a critical regulator of hematopoietic stem cell expansion, as a target gene suppressed by NOR1 in MDP cells. Finally, in addition to inducing Ly6C(+) monocytosis, NOR1 deletion increased the replicative rate of lesional macrophages and induced local foam cell formation within the atherosclerotic plaque. Collectively, our studies demonstrate that NOR1 deletion in hematopoietic stem cells accelerates atherosclerosis formation by promoting myelopoiesis in the stem cell compartment and by inducing local proatherogenic activities in the macrophage, including lesional macrophage proliferation and foam cell formation.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Células Madre Hematopoyéticas/metabolismo , Monocitos/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Receptores de Esteroides/deficiencia , Receptores de Hormona Tiroidea/deficiencia , Animales , Proliferación Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Células HEK293 , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Placa Aterosclerótica/genética , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo , Transfección
7.
Obesity (Silver Spring) ; 19(12): 2429-32, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21959341

RESUMEN

Excessive accumulation of reactive oxygen species (ROS) in adipose tissue has been implicated in the development of insulin resistance and type 2 diabetes. However, emerging evidence suggests a physiologic role of ROS in cellular signaling and insulin sensitivity. In this study, we demonstrate that pharmacologic depletion of the antioxidant glutathione in mice prevents diet-induced obesity, increases energy expenditure and locomotor activity, and enhances insulin sensitivity. These observations support a beneficial role of ROS in glucose homeostasis and warrant further research to define the regulation of metabolism and energy balance by ROS.


Asunto(s)
Dieta/efectos adversos , Metabolismo Energético , Glutatión/metabolismo , Resistencia a la Insulina , Obesidad/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/metabolismo , Conducta Animal , Glucemia/metabolismo , Homeostasis , Locomoción , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Transducción de Señal
8.
J Biol Chem ; 286(41): 35485-35493, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21868379

RESUMEN

Members of the NR4A subgroup of the nuclear hormone receptor superfamily have emerged as key transcriptional regulators of proliferation and inflammation. NOR1 constitutes a ligand-independent transcription factor of this subgroup and induces cell proliferation; however, the transcriptional mechanisms underlying this mitogenic role remain to be defined. Here, we demonstrate that the F-box protein SKP2 (S phase kinase-associated protein 2), the substrate-specific receptor of the ubiquitin ligase responsible for the degradation of p27(KIP1) through the proteasome pathway, constitutes a direct transcriptional target for NOR1. Mitogen-induced Skp2 expression is silenced in vascular smooth muscle cells (VSMC) isolated from Nor1-deficient mice or transfected with Nor1 siRNA. Conversely, adenovirus-mediated overexpression of NOR1 induces Skp2 expression in VSMC and decreases protein abundance of its target p27. Transient transfection experiments establish that NOR1 transactivates the Skp2 promoter through a nerve growth factor-induced clone B response element (NBRE). Electrophoretic mobility shift and chromatin immunoprecipitation assays further revealed that NOR1 is recruited to this NBRE site in the Skp2 promoter in response to mitogenic stimulation. In vivo Skp2 expression is increased during the proliferative response underlying neointima formation, and this transcriptional induction depends on the expression of NOR1. Finally, we demonstrate that overexpression of Skp2 rescues the proliferative arrest of Nor1-deficient VSMC. Collectively, these results characterize Skp2 as a novel NOR1-regulated target gene and detail a previously unrecognized transcriptional cascade regulating mitogen-induced VSMC proliferation.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Regulación Enzimológica de la Expresión Génica , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Elementos de Respuesta , Proteínas Quinasas Asociadas a Fase-S/biosíntesis , Animales , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Ratones , Ratones Noqueados , Neointima/genética , Neointima/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Proteínas Quinasas Asociadas a Fase-S/genética , Activación Transcripcional/genética
9.
PLoS One ; 6(4): e18532, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21533223

RESUMEN

Aging constitutes a major independent risk factor for the development of type 2 diabetes and is accompanied by insulin resistance and adipose tissue dysfunction. One of the most important factors implicitly linked to aging and age-related chronic diseases is the accumulation of oxidative stress. However, the effect of increased oxidative stress on adipose tissue biology remains elusive. In this study, we demonstrate that aging in mice results in a loss of fat mass and the accumulation of oxidative stress in adipose tissue. In vitro, increased oxidative stress through glutathione depletion inhibits preadipocyte differentiation. This inhibition of adipogenesis is at least in part the result of reduced cell proliferation and an inhibition of G(1)→S-phase transition during the initial mitotic clonal expansion of the adipocyte differentiation process. While phosphorylation of the retinoblastoma protein (Rb) by cyclin/cdk complexes remains unaffected, oxidative stress decreases the expression of S-phase genes downstream of Rb. This silencing of S phase gene expression by increased oxidative stress is mediated through a transcriptional mechanism involving the inhibition of E2F recruitment and transactivation of its target promoters. Collectively, these data demonstrate a previously unrecognized role of oxidative stress in the regulation of adipogenesis which may contribute to age-associated adipose tissue dysfunction.


Asunto(s)
Tejido Adiposo/metabolismo , Envejecimiento/metabolismo , Estrés Oxidativo , Células 3T3-L1 , Tejido Adiposo/crecimiento & desarrollo , Animales , Secuencia de Bases , Composición Corporal , Inmunoprecipitación de Cromatina , Cartilla de ADN , Glutatión/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Arterioscler Thromb Vasc Biol ; 31(4): 851-60, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21233448

RESUMEN

OBJECTIVE: Proliferation of smooth muscle cells (SMC) in response to vascular injury is central to neointimal vascular remodeling. There is accumulating evidence that histone acetylation constitutes a major epigenetic modification for the transcriptional control of proliferative gene expression; however, the physiological role of histone acetylation for proliferative vascular disease remains elusive. METHODS AND RESULTS: In the present study, we investigated the role of histone deacetylase (HDAC) inhibition in SMC proliferation and neointimal remodeling. We demonstrate that mitogens induce transcription of HDAC 1, 2, and 3 in SMC. Short interfering RNA-mediated knockdown of either HDAC 1, 2, or 3 and pharmacological inhibition of HDAC prevented mitogen-induced SMC proliferation. The mechanisms underlying this reduction of SMC proliferation by HDAC inhibition involve a growth arrest in the G(1) phase of the cell cycle that is due to an inhibition of retinoblastoma protein phosphorylation. HDAC inhibition resulted in a transcriptional and posttranscriptional regulation of the cyclin-dependent kinase inhibitors p21(Cip1) and p27(Kip). Furthermore, HDAC inhibition repressed mitogen-induced cyclin D1 mRNA expression and cyclin D1 promoter activity. As a result of this differential cell cycle-regulatory gene expression by HDAC inhibition, the retinoblastoma protein retains a transcriptional repression of its downstream target genes required for S phase entry. Finally, we provide evidence that these observations are applicable in vivo by demonstrating that HDAC inhibition decreased neointima formation and expression of cyclin D1 in a murine model of vascular injury. CONCLUSIONS: These findings identify HDAC as a critical component of a transcriptional cascade regulating SMC proliferation and suggest that HDAC might play a pivotal role in the development of proliferative vascular diseases, including atherosclerosis and in-stent restenosis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Hidroxilaminas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Quinolinas/farmacología , Túnica Media/efectos de los fármacos , Lesiones del Sistema Vascular/tratamiento farmacológico , Acetilación , Animales , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Factores de Transcripción E2F/metabolismo , Histona Desacetilasas/genética , Histonas/metabolismo , Hiperplasia , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Fosforilación , Interferencia de ARN , Ratas , Proteína de Retinoblastoma/metabolismo , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Túnica Media/enzimología , Túnica Media/lesiones , Túnica Media/patología , Lesiones del Sistema Vascular/enzimología , Lesiones del Sistema Vascular/patología
11.
Arterioscler Thromb Vasc Biol ; 31(2): 253-60, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21088250

RESUMEN

OBJECTIVE: Abdominal aortic aneurysms (AAA) are an age-related vascular disease and an important cause of morbidity and mortality. In this study, we sought to determine whether the catalytic component of telomerase, telomerase reverse transcriptase (TERT), modulates angiotensin (Ang) II-induced AAA formation. METHODS AND RESULTS: Low-density lipoprotein receptor-deficient (LDLr-/-) mice were lethally irradiated and reconstituted with bone marrow-derived cells from TERT-deficient (TERT-/-) mice or littermate wild-type mice. Mice were placed on a diet enriched in cholesterol, and AAA formation was quantified after 4 weeks of Ang II infusion. Repopulation of LDLr-/- mice with TERT-/- bone marrow-derived cells attenuated Ang II-induced AAA formation. TERT-deficient recipient mice revealed modest telomere attrition in circulating leukocytes at the study end point without any overt effect of the donor genotype on white blood cell counts. In mice repopulated with TERT-/- bone marrow, aortic matrix metalloproteinase-2 (MMP-2) activity was reduced, and TERT-/- macrophages exhibited decreased expression and activity of MMP-2 in response to stimulation with Ang II. Finally, we demonstrated in transient transfection studies that TERT overexpression activates the MMP-2 promoter in macrophages. CONCLUSIONS: TERT deficiency in bone marrow-derived macrophages attenuates Ang II-induced AAA formation in LDLr-/- mice and decreases MMP-2 expression. These results point to a previously unrecognized role of TERT in the pathogenesis of AAA.


Asunto(s)
Angiotensina II/efectos adversos , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/prevención & control , Macrófagos/enzimología , Telomerasa/deficiencia , Animales , Trasplante de Médula Ósea , Movimiento Celular , Células Cultivadas , Elastina/metabolismo , Genotipo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Receptores de LDL/genética , Receptores de LDL/metabolismo , Telomerasa/genética , Telómero
12.
Arterioscler Thromb Vasc Biol ; 31(2): 245-52, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21106948

RESUMEN

OBJECTIVE: Telomerase serves as a critical regulator of tissue renewal. Although telomerase activity is inducible in response to various environmental cues, it remains unknown whether telomerase is activated during the inflammatory remodeling underlying atherosclerosis formation. To address this question, we investigated in the present study the regulation of telomerase in macrophages and during atherosclerosis development in low-density lipoprotein receptor-deficient mice. METHODS AND RESULTS: We demonstrate that inflammatory stimuli activate telomerase in macrophages by inducing the expression of the catalytic subunit telomerase reverse transcriptase (TERT). Reporter and chromatin immunoprecipitation assays identified a previously unrecognized nuclear factor-κB (NF-κB) response element in the TERT promoter, to which NF-κB is recruited during inflammation. Inhibition of NF-κB signaling completely abolished the induction of TERT expression, characterizing TERT as a bona fide NF-κB target gene. Furthermore, functional experiments revealed that TERT deficiency results in a senescent cell phenotype. Finally, we demonstrate high levels of TERT expression in macrophages of human atherosclerotic lesions and establish that telomerase is activated during atherosclerosis development in low-density lipoprotein receptor-deficient mice. CONCLUSIONS: These results characterize TERT as a previously unrecognized NF-κB target gene in macrophages and demonstrate that telomerase is activated during atherosclerosis. This induction of TERT expression prevents macrophage senescence and may have important implications for the development of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Inflamación/metabolismo , Macrófagos/enzimología , Telomerasa/metabolismo , Animales , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Células Cultivadas , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Humanos , Inflamación/patología , Inflamación/fisiopatología , Lipopolisacáridos/farmacología , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Telomerasa/genética , Factor de Necrosis Tumoral alfa/farmacología
13.
Circ Res ; 107(4): 501-11, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20558821

RESUMEN

RATIONALE: The orphan nuclear receptor NOR1 is a member of the evolutionary highly conserved and ligand-independent NR4A subfamily of the nuclear hormone receptor superfamily. Members of this subfamily have been characterized as early response genes regulating essential biological processes including inflammation and proliferation; however, the role of NOR1 in atherosclerosis remains unknown. OBJECTIVE: The goal of the present study was to determine the causal contribution of NOR1 to atherosclerosis development and to identify the mechanism by which this nuclear receptor participates in the disease process. METHODS AND RESULTS: In the present study, we demonstrate expression of NOR1 in endothelial cells of human atherosclerotic lesions. In response to inflammatory stimuli, NOR1 expression is rapidly induced in endothelial cells through a nuclear factor kappaB-dependent transactivation of the NOR1 promoter. Overexpression of NOR1 in human endothelial cells increased the expression of vascular cell adhesion molecule (VCAM)-1 and intercellular adhesion molecule-1, whereas NOR1 deficiency altered adhesion molecule expression in response to inflammatory stimuli. Transient transfection experiments and chromatin immunoprecipitation assays revealed that NOR1 induces VCAM-1 promoter activity by binding to a canonical response element for NR4A receptors in the VCAM-1 promoter. Further functional studies confirmed that NOR1 mediates monocyte adhesion by inducing VCAM-1 and intercellular adhesion molecule-1 expression in endothelial cells. Finally, we demonstrate that NOR1 deficiency reduces hypercholesterolemia-induced atherosclerosis formation in apoE(-/-) mice by decreasing the macrophage content of the lesion. CONCLUSIONS: In concert, these studies identify a novel pathway underlying monocyte adhesion and establish that NOR1 serves a previously unrecognized atherogenic role in mice by positively regulating monocyte recruitment to the vascular wall.


Asunto(s)
Aterosclerosis/metabolismo , Proteínas de Unión al ADN/deficiencia , Monocitos/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Receptores de Esteroides/deficiencia , Receptores de Hormona Tiroidea/deficiencia , Animales , Aterosclerosis/patología , Aterosclerosis/prevención & control , Adhesión Celular/fisiología , Células Cultivadas , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Proteínas de Unión al ADN/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Elementos de Respuesta/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Circulation ; 119(4): 577-86, 2009 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19153266

RESUMEN

BACKGROUND: The neuron-derived orphan receptor-1 (NOR1) belongs to the evolutionary highly conserved and most ancient NR4A subfamily of the nuclear hormone receptor superfamily. Members of this subfamily function as early-response genes regulating key cellular processes, including proliferation, differentiation, and survival. Although NOR1 has previously been demonstrated to be required for smooth muscle cell proliferation in vitro, the role of this nuclear receptor for the proliferative response underlying neointima formation and target genes trans-activated by NOR1 remain to be defined. METHODS AND RESULTS: Using a model of guidewire-induced arterial injury, we demonstrate decreased neointima formation in NOR1(-/-) mice compared with wild-type mice. In vitro, NOR1-deficient smooth muscle cells exhibit decreased proliferation as a result of a G(1)-->S phase arrest of the cell cycle and increased apoptosis in response to serum deprivation. NOR1 deficiency alters phosphorylation of the retinoblastoma protein by preventing mitogen-induced cyclin D1 and D2 expression. Conversely, overexpression of NOR1 induces cyclin D1 expression and the transcriptional activity of the cyclin D1 promoter in transient reporter assays. Gel shift and chromatin immunoprecipitation assays identified a putative response element for NR4A receptors in the cyclin D1 promoter, to which NOR1 is recruited in response to mitogenic stimulation. Finally, we provide evidence that these observations are applicable in vivo by demonstrating decreased cyclin D1 expression during neointima formation in NOR1-deficient mice. CONCLUSIONS: These experiments characterize cyclin D1 as an NOR1-regulated target gene in smooth muscle cells and demonstrate that NOR1 deficiency decreases neointima formation in response to vascular injury.


Asunto(s)
Proteínas de Unión al ADN/genética , Músculo Liso Vascular/lesiones , Músculo Liso Vascular/fisiología , Receptores de Esteroides/genética , Cicatrización de Heridas/fisiología , Animales , Aorta/citología , Apoptosis/fisiología , División Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Vasos Coronarios/citología , Ciclina D1/genética , Ciclina D2 , Ciclinas/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F/metabolismo , Expresión Génica/fisiología , Humanos , Ratones , Ratones Mutantes , Músculo Liso Vascular/citología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Fosforilación/fisiología , Regiones Promotoras Genéticas/fisiología , Ratas , Receptores de Esteroides/metabolismo , Proteína de Retinoblastoma/metabolismo , Túnica Íntima/citología , Túnica Íntima/lesiones , Túnica Íntima/fisiología
15.
Circ Res ; 103(10): 1155-63, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18818403

RESUMEN

Peroxisome proliferator-activated receptor (PPAR)alpha, the molecular target for fibrates used to treat dyslipidemia, exerts pleiotropic effects on vascular cells. In vascular smooth muscle cells (VSMCs), we have previously demonstrated that PPARalpha activation suppresses G(1)-->S cell cycle progression by targeting the cyclin-dependent kinase inhibitor p16(INK4a) (p16). In the present study, we demonstrate that this inhibition of VSMC proliferation by PPARalpha is mediated through a p16-dependent suppression of telomerase activity, which has been implicated in key cellular functions including proliferation. PPARalpha activation inhibited mitogen-induced telomerase activity by repressing the catalytic subunit telomerase reverse transcriptase (TERT) through negative cross-talk with an E2F-1-dependent trans-activation of the TERT promoter. This trans-repression involved the recruitment of the retinoblastoma (RB) family proteins p107 and p130 to the TERT promoter resulting in impaired E2F-1 binding, an effect that was dependent on p16. The inhibition of cell proliferation by PPARalpha activation was lost in VSMCs following TERT overexpression or knockdown, pointing to a key role of telomerase as a target for the antiproliferative effects of PPARalpha. Finally, we demonstrate that PPARalpha agonists suppress telomerase activation during the proliferative response following vascular injury, indicating that these findings are applicable in vivo. In concert, these results demonstrate that the antiproliferative effects of PPARalpha in VSMCs depend on the suppression of telomerase activity by targeting the p16/RB/E2F transcriptional cascade.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fase G1/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Miocitos del Músculo Liso/enzimología , PPAR alfa/metabolismo , Fase S/fisiología , Telomerasa/biosíntesis , Animales , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Dislipidemias/enzimología , Dislipidemias/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Ratones , Mitógenos/metabolismo , Mitógenos/farmacología , PPAR alfa/agonistas , Regiones Promotoras Genéticas/fisiología , Ratas , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/metabolismo , Proteína p130 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Telomerasa/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología
16.
J Clin Invest ; 117(10): 2877-88, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17823662

RESUMEN

Obesity is associated with a state of chronic, low-grade inflammation characterized by abnormal cytokine production and macrophage infiltration into adipose tissue, which may contribute to the development of insulin resistance. During immune responses, tissue infiltration by macrophages is dependent on the expression of osteopontin, an extracellular matrix protein and proinflammatory cytokine that promotes monocyte chemotaxis and cell motility. In the present study, we used a murine model of diet-induced obesity to examine the role of osteopontin in the accumulation of adipose tissue macrophages and the development of insulin resistance during obesity. Mice exposed to a high-fat diet exhibited increased plasma osteopontin levels, with elevated expression in macrophages recruited into adipose tissue. Obese mice lacking osteopontin displayed improved insulin sensitivity in the absence of an effect on diet-induced obesity, body composition, or energy expenditure. These mice further demonstrated decreased macrophage infiltration into adipose tissue, which may reflect both impaired macrophage motility and attenuated monocyte recruitment by stromal vascular cells. Finally, obese osteopontin-deficient mice exhibited decreased markers of inflammation, both in adipose tissue and systemically. Taken together, these results suggest that osteopontin may play a key role in linking obesity to the development of insulin resistance by promoting inflammation and the accumulation of macrophages in adipose tissue.


Asunto(s)
Tejido Adiposo/inmunología , Resistencia a la Insulina/inmunología , Macrófagos/inmunología , Obesidad/inmunología , Osteopontina/fisiología , Animales , Quimiocina CCL2/metabolismo , Quimiotaxis/genética , Grasas de la Dieta/administración & dosificación , Inflamación/genética , Inflamación/inmunología , Ratones , Ratones Mutantes , Obesidad/complicaciones , Osteopontina/genética
17.
Diabetes ; 56(6): 1662-70, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17360982

RESUMEN

Osteopontin (OPN) is a proinflammatory cytokine implicated in the chemoattraction of monocytes and the development of atherosclerosis. Peroxisome proliferator-activated receptor (PPAR)alpha, a ligand-activated transcription factor with pleiotropic anti-inflammatory effects in macrophages, is the molecular target for fibrates, which are frequently used to treat dyslipidemia in patients with type 2 diabetes at high risk for cardiovascular disease. In the present study, we examined the regulation of OPN by PPARalpha agonists in macrophages and determined the effect of fibrate treatment on OPN plasma levels in patients with type 2 diabetes. Treatment of human macrophages with the PPARalpha ligands bezafibrate or WY14643 inhibited OPN expression. PPARalpha ligands suppressed OPN promoter activity, and an activator protein (AP)-1 consensus site conferred this repression. Overexpression of c-Fos and c-Jun reversed the inhibitory effect of PPARalpha ligands on OPN transcription, and, in chromatin immunoprecipitation assays, PPARalpha ligands inhibited c-Fos and phospho-c-Jun binding to the OPN promoter. Moreover, c-Fos and phospho-c-Jun protein expression was inhibited by PPARalpha agonists, indicating that PPARalpha ligands suppress OPN expression through negative cross talk with AP-1-dependent transactivation of the OPN promoter. This inhibitory effect of PPARalpha ligands on OPN expression was absent in PPARalpha-deficient macrophages, suggesting a receptor-mediated mechanism of OPN suppression. Finally, treatment of type 2 diabetic patients with bezafibrate significantly decreased OPN plasma levels. These results demonstrate a novel mechanism whereby PPARalpha ligands may impact macrophage inflammatory responses and decrease early proinflammatory markers for cardiovascular disease.


Asunto(s)
Bezafibrato/farmacología , Diabetes Mellitus Tipo 2/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/fisiología , Osteopontina/genética , PPAR alfa/agonistas , Animales , Línea Celular , Humanos , Hipolipemiantes/farmacología , Ligandos , Ratones , Monocitos/fisiología , Osteopontina/sangre , Plásmidos , Reacción en Cadena de la Polimerasa , Pirimidinas/farmacología , Transfección
18.
J Biol Chem ; 281(44): 33467-76, 2006 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16945922

RESUMEN

Members of the nuclear hormone receptor superfamily function as key transcriptional regulators of inflammation and proliferation in cardiovascular diseases. In addition to the ligand-dependent peroxisome proliferator-activated receptors and liver X receptors, this family of transcription factors includes a large number of orphan receptors, and their role in vascular diseases remains to be investigated. The neuron-derived orphan receptor-1 (NOR1) belongs to the ligand-independent NR4A subfamily, which has been implicated in cell proliferation, differentiation, and apoptosis. In this study, we demonstrate NOR1 expression in vascular smooth muscle cells (SMC) of human atherosclerotic lesions. In response to mitogenic stimulation with platelet-derived growth factor (PDGF), SMC rapidly express NOR1 through an ERK-MAPK-dependent signaling pathway. 5'-deletion analysis, site-directed mutagenesis, and transactivation experiments demonstrate that PDGF-induced NOR1 expression is mediated through a cAMP-response element-binding protein (CREB)-dependent transactivation of the NOR1 promoter. Consequently, short interfering RNA-mediated depletion of CREB abolished PDGF-induced NOR1 expression in SMC. Furthermore, PDGF induced Ser-133 phosphorylation of CREB and subsequent binding to the CRE sites of the endogenous NOR1 promoter. Functional analysis demonstrated that PDGF induces NOR1 transactivation of its consensus NGFI-B-response elements (NBRE) in SMC. We finally demonstrate that SMC isolated from NOR1-deficient mice exhibit decreased cell proliferation and characterize cyclin D1 and D2 as NOR1 target genes in SMC. These experiments indicate that PDGF-induced NOR1 transcription in SMC is mediated through CREB-dependent transactivation of the NOR1 promoter and further demonstrate that NOR1 functions as a key transcriptional regulator of SMC proliferation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Animales , Aterosclerosis/metabolismo , Proliferación Celular , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Proteínas del Tejido Nervioso/genética , Fosfoserina/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Mensajero/genética , Ratas , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Elementos de Respuesta , Transcripción Genética/genética , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...