Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(11)2022 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-35682840

RESUMEN

Reprogramming of metabolic pathways in monocytes and macrophages can induce a proatherosclerotic inflammatory memory called trained innate immunity. Here, we have analyzed the role of the Liver X receptor (LXR), a crucial regulator of metabolism and inflammation, in oxidized low-density lipoprotein (oxLDL)-induced trained innate immunity. Human monocytes were incubated with LXR agonists, antagonists, and oxLDL for 24 h. After five days of resting time, cells were restimulated with the TLR-2 agonist Pam3cys. OxLDL priming induced the expression of LXRα but not LXRß. Pharmacologic LXR activation was enhanced, while LXR inhibition prevented the oxLDL-induced inflammatory response. Furthermore, LXR inhibition blocked the metabolic changes necessary for epigenetic reprogramming associated with trained immunity. In fact, enrichment of activating histone marks at the IL-6 and TNFα promotor was reduced following LXR inhibition. Based on the differential expression of the LXR isoforms, we inhibited LXRα and LXRß genes using siRNA in THP1 cells. As expected, siRNA-mediated knock-down of LXRα blocked the oxLDL-induced inflammatory response, while knock-down of LXRß had no effect. We demonstrate a specific and novel role of the LXRα isoform in the regulation of oxLDL-induced trained immunity. Our data reveal important aspects of LXR signaling in innate immunity with relevance to atherosclerosis formation.


Asunto(s)
Lipoproteínas LDL , Receptores Nucleares Huérfanos , Humanos , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacología , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Macrófagos/metabolismo , Receptores Nucleares Huérfanos/genética , ARN Interferente Pequeño/metabolismo
2.
Herz ; 46(3): 242-249, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31965195

RESUMEN

BACKGROUND: Patients at high risk of bleeding requiring percutaneous coronary intervention (PCI) need careful evaluation of both their thrombotic and their bleeding risks. In these patients, a polymer-free metallic stent coated with biolimus-A9 (BA9-DCS) followed by 1­month dual antiplatelet therapy (DAPT) could be a safe option; however, real-world data are still lacking. We analyzed the performance of the device in a real-world scenario. METHODS: Patients assessed as being at high risk of bleeding with an indication for PCI were treated with BA9-DCS and DAPT consisting of aspirin (100 mg/day) and clopidogrel (75 mg/day) for at least 1 month, followed by either oral anticoagulation or single antiplatelet therapy. No exclusion criteria were used. The primary endpoint was the occurrence of an adverse event after PCI, i.e. severe bleeding requiring hospitalization, ischemic or hemorrhagic stroke, clinically driven stent thrombosis, myocardial infarction or cardiac death. RESULTS: Overall, 89 patients were enrolled in this study [median age 75 (66-81) years; 27 females (30%)] and 171 interventions were performed. During a median follow-up of 203 (145-273) days the primary endpoint occurred in 20 patients (23%): 12 (13%) had clinically significant bleeding, four (5%) ischemic stroke and four (5%) died from cardiac causes related neither to stent thrombosis nor to acute myocardial infarction. Female gender emerged as the only statistically significant predictor of an adverse event (adjusted hazard ratio (HR) 3.3; 95% confidence interval (CI): 1.2-8.7, p = 0.017). CONCLUSION: In real-world patients at high risk of bleeding, implantation of the polymer-free metallic stent coated with Biolimus-A9 (Biofreedom®; Biosensors Europe, Morges, Switzerland) followed by 1 -month DAPT showed encouraging results without any stent thrombosis.


Asunto(s)
Enfermedad de la Arteria Coronaria , Stents Liberadores de Fármacos , Intervención Coronaria Percutánea , Anciano , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Quimioterapia Combinada , Europa (Continente) , Femenino , Humanos , Inhibidores de Agregación Plaquetaria/efectos adversos , Polímeros , Sirolimus/análogos & derivados , Suiza , Resultado del Tratamiento
3.
J Mol Cell Cardiol ; 146: 121-132, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32726647

RESUMEN

Trained innate immunity describes the metabolic reprogramming and long-term proinflammatory activation of innate immune cells in response to different pathogen or damage associated molecular patterns, such as oxidized low-density lipoprotein (oxLDL). Here, we have investigated whether the regulatory networks of trained innate immunity also control endothelial cell activation following oxLDL treatment. Human aortic endothelial cells (HAECs) were primed with oxLDL for 24 h. After a resting time of 4 days, cells were restimulated with the TLR2-agonist PAM3cys4. OxLDL priming induced a proinflammatory memory with increased production of inflammatory cytokines such as IL-6, IL-8 and MCP-1 in response to PAM3cys4 restimulation. This memory formation was dependent on TLR2 activation. Furthermore, oxLDL priming of HAECs caused characteristic metabolic and epigenetic reprogramming, including activation of mTOR-HIF1α-signaling with increases in glucose consumption and lactate production, as well as epigenetic modifications in inflammatory gene promoters. Inhibition of mTOR-HIF1α-signaling or histone methyltransferases blocked the observed phenotype. Furthermore, primed HAECs showed epigenetic activation of ICAM-1 and increased ICAM-1 expression in a HIF1α-dependent manner. Accordingly, live cell imaging revealed increased monocyte adhesion and transmigration following oxLDL priming. In summary, we demonstrate that oxLDL-mediated endothelial cell activation represents an immunologic event, which triggers metabolic and epigenetic reprogramming. Molecular mechanisms regulating trained innate immunity in innate immune cells also regulate this sustained proinflammatory phenotype in HAECs with enhanced atheroprone cell functions. Further research is necessary to elucidate the detailed metabolic regulation and the functional relevance for atherosclerosis formation in vivo.


Asunto(s)
Células Endoteliales/metabolismo , Memoria Inmunológica/efectos de los fármacos , Lipoproteínas LDL/farmacología , Aorta/metabolismo , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamación/patología , Monocitos/efectos de los fármacos , Fenotipo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/metabolismo
4.
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
5.
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
6.
Eur Radiol ; 26(5): 1420-30, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26304802

RESUMEN

OBJECTIVES: Whether whole-body MRI can predict occurrence of recurrent events in patients with diabetes mellitus. METHODS: Whole-body MRI was prospectively applied to 61 diabetics and assessed for arteriosclerosis and ischemic cerebral/myocardial changes. Occurrence of cardiocerebral events and diabetic comorbidites was determined. Patients were stratified whether no, a single or recurrent events arose. As a secondary endpoint, events were stratified into organ system-specific groups. RESULTS: During a median follow-up of 70 months, 26 diabetics developed a total of 39 events; 18 (30%) developed one, 8 (13%) recurrent events. Between diabetics with no, a single and recurrent events, a stepwise higher burden was observed for presence of left ventricular (LV) hypo-/akinesia (3/28/75%, p < 0.0001), myocardial delayed-contrast-enhancement (17/33/63%, p = 0.001), carotid artery stenosis (11/17/63%, p = 0.005), peripheral artery stenosis (26/56/88%, p = 0.0006) and vessel score (1.00/1.30/1.76, p < 0.0001). After adjusting for clinical characteristics, LV hypo-/akinesia (hazard rate ratio = 6.57, p < 0.0001) and vessel score (hazard rate ratio = 12.29, p < 0.0001) remained independently associated. Assessing organ system risk, cardiac and cerebral MR findings predicted more strongly events in their respective organ system. Vessel-score predicted both cardiac and cerebral, but not non-cardiocerebral, events. CONCLUSION: Whole-body MR findings predict occurrence of recurrent events in diabetics independent of clinical characteristics, and may concurrently provide organ system-specific risk. KEY POINTS: • Patients with long-standing diabetes mellitus are at high risk for recurrent events. • Whole-body MRI predicts occurrence of recurrent events independently of clinical characteristics. • The vessel score derived from whole-body angiography is a good general risk-marker. • Whole-body MRI may also provide organ-specific risk assessment. • Current findings may indicate benefits of whole-body MRI for risk stratification.


Asunto(s)
Isquemia Encefálica/patología , Angiopatías Diabéticas/patología , Isquemia Miocárdica/patología , Anciano , Estenosis Carotídea/patología , Enfermedad de la Arteria Coronaria/patología , Diagnóstico Precoz , Femenino , Humanos , Arteriosclerosis Intracraneal/patología , Angiografía por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Recurrencia , Medición de Riesgo , Imagen de Cuerpo Entero/métodos
7.
J Mol Cell Cardiol ; 81: 23-33, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25640159

RESUMEN

Phosphodiesterase 4 (PDE4) activity mediates cAMP-dependent smooth muscle cell (SMC) activation following vascular injury. In this study we have investigated the effects of specific PDE4 inhibition with roflumilast on SMC proliferation and inflammatory activation in vitro and neointima formation following guide wire-induced injury of the femoral artery in mice in vivo. In vitro, roflumilast did not affect SMC proliferation, but diminished TNF-α induced expression of the vascular cell adhesion molecule 1 (VCAM-1). Specific activation of the cAMP effector Epac, but not PKA activation mimicked the effects of roflumilast on VCAM-1 expression. Consistently, the reduction of VCAM-1 expression was rescued following inhibition of Epac. TNF-α induced NFκB p65 translocation and VCAM-1 promoter activity were not altered by roflumilast in SMCs. However, roflumilast treatment and Epac activation repressed the induction of the activating epigenetic histone mark H3K4me2 at the VCAM-1 promoter, while PKA activation showed no effect. Furthermore, HDAC inhibition blocked the inhibitory effect of roflumilast on VCAM-1 expression. Both, roflumilast and Epac activation reduced monocyte adhesion to SMCs in vitro. Finally, roflumilast treatment attenuated femoral artery intima-media ratio by more than 50% after 4weeks. In summary, PDE4 inhibition regulates VCAM-1 through a novel Epac-dependent mechanism, which involves regulatory epigenetic components and reduces neointima formation following vascular injury. PDE4 inhibition and Epac activation might represent novel approaches for the treatment of vascular diseases, including atherosclerosis and in-stent restenosis.


Asunto(s)
Aminopiridinas/farmacología , Benzamidas/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Factores de Intercambio de Guanina Nucleótido/genética , Neointima/prevención & control , Inhibidores de Fosfodiesterasa 4/farmacología , Molécula 1 de Adhesión Celular Vascular/genética , Lesiones del Sistema Vascular/tratamiento farmacológico , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Ciclopropanos/farmacología , Arteria Femoral/efectos de los fármacos , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Monocitos/citología , Monocitos/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Neointima/genética , Neointima/metabolismo , Neointima/patología , Ratas , Transducción de Señal , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Lesiones del Sistema Vascular/genética , Lesiones del Sistema Vascular/metabolismo , Lesiones del Sistema Vascular/patología
8.
STAR Protoc ; 5(1): 102776, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38117658

RESUMEN

Non-immune cells, like innate immune cells, can develop a memory-like phenotype in response to priming with microbial compounds or certain metabolites, which enables an enhanced response to a secondary unspecific stimulus. This paper describes a step-by-step protocol for the induction and analysis of trained immunity in human endothelial and smooth muscle cells. We then describe steps for cell culture with cryopreserved vascular cells, subcultivation, and induction of trained immunity. We then provide detailed procedures for downstream analysis using ELISA and qPCR. For complete details on the use and execution of this protocol, please refer to Sohrabi et al. (2020)1 and Shcnack et al.2.


Asunto(s)
Células Endoteliales , Inmunidad Entrenada , Humanos , Técnicas de Cultivo de Célula , Ensayo de Inmunoadsorción Enzimática , Miocitos del Músculo Liso
9.
Radiology ; 269(3): 730-7, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24023074

RESUMEN

PURPOSE: To study the predictive value of whole-body magnetic resonance (MR) imaging for the occurrence of cardiac and cerebrovascular events in a cohort of patients with diabetes mellitus (DM). MATERIALS AND METHODS: This HIPAA-compliant study was approved by the institutional review board. Informed consent was obtained from all patients before enrollment into the study. The authors followed up 65 patients with DM (types 1 and 2) who underwent a comprehensive, contrast material-enhanced whole-body MR imaging protocol, including brain, cardiac, and vascular sequences at baseline. Follow-up was performed by phone interview. The primary endpoint was a major adverse cardiac and cerebrovascular event (MACCE), which was defined as composite cardiac-cerebrovascular death, myocardial infarction, cerebrovascular event, or revascularization. MR images were assessed for the presence of systemic atherosclerotic vessel changes, white matter lesions, and myocardial changes. Kaplan-Meier survival and Cox regression analyses were performed to determine associations. RESULTS: Follow-up was completed in 61 patients (94%; median age, 67.5 years; 30 women [49%]; median follow-up, 70 months); 14 of the 61 patients (23%) experienced MACCE. Although normal whole-body MR imaging excluded MACCE during the follow-up period (0%; 95% confidence interval [CI]: 0%, 17%), any detectable ischemic and/or atherosclerotic changes at whole-body MR imaging (prevalence, 66%) conferred a cumulative event rate of 20% at 3 years and 35% at 6 years. Whole-body MR imaging summary estimate of disease was strongly predictive for MACCE (one increment of vessel score and each territory with atherosclerotic changes: hazard ratio, 13.2 [95% CI: 4.5, 40.1] and 3.9 [95% CI: 2.2, 7.5], respectively), also beyond clinical characteristics as well as individual cardiac or cerebrovascular MR findings. CONCLUSION: These initial data indicate that disease burden as assessed with whole-body MR imaging confers strong prognostic information in patients with DM. Online supplemental material is available for this article.


Asunto(s)
Enfermedades Cardiovasculares/diagnóstico , Trastornos Cerebrovasculares/diagnóstico , Complicaciones de la Diabetes/diagnóstico , Imagen por Resonancia Magnética/métodos , Imagen de Cuerpo Entero , Anciano , Medios de Contraste , Determinación de Punto Final , Femenino , Estudios de Seguimiento , Humanos , Entrevistas como Asunto , Masculino , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Riesgo
10.
Curr Atheroscler Rep ; 15(4): 319, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23423526

RESUMEN

Epigenetics involve heritable and acquired changes in gene transcription that occur independently of the DNA sequence. Epigenetic mechanisms constitute a hierarchic upper-level of transcriptional control through complex modifications of chromosomal components and nuclear structures. These modifications include, for example, DNA methylation or post-translational modifications of core histones; they are mediated by various chromatin-modifying enzymes; and ultimately they define the accessibility of a transcriptional complex to its target DNA. Integrating epigenetic mechanisms into the pathophysiologic concept of complex and multifactorial diseases such as atherosclerosis may significantly enhance our understanding of related mechanisms and provide promising therapeutic approaches. Although still in its infancy, intriguing scientific progress has begun to elucidate the role of epigenetic mechanisms in vascular biology, particularly in the control of smooth muscle cell phenotypes. In this review, we will summarize epigenetic pathways in smooth muscle cells, focusing on mechanisms involved in the regulation of vascular remodeling.


Asunto(s)
Aterosclerosis/genética , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Aterosclerosis/fisiopatología , Proliferación Celular , Metilación de ADN , Epigénesis Genética , Histonas/metabolismo , Humanos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo
11.
Curr Atheroscler Rep ; 15(5): 319, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23630979

RESUMEN

Epigenetics involve heritable and acquired changes in gene transcription that occur independently of the DNA sequence. Epigenetic mechanisms constitute a hierarchic upper-level of transcriptional control through complex modifications of chromosomal components and nuclear structures. These modifications include, for example, DNA methylation or post-translational modifications of core histones; they are mediated by various chromatin-modifying enzymes; and ultimately they define the accessibility of a transcriptional complex to its target DNA. Integrating epigenetic mechanisms into the pathophysiologic concept of complex and multifactorial diseases such as atherosclerosis may significantly enhance our understanding of related mechanisms and provide promising therapeutic approaches. Although still in its infancy, intriguing scientific progress has begun to elucidate the role of epigenetic mechanisms in vascular biology, particularly in the control of smooth muscle cell phenotypes. In this review, we will summarize epigenetic pathways in smooth muscle cells, focusing on mechanisms involved in the regulation of vascular remodeling.


Asunto(s)
Aterosclerosis/genética , Epigénesis Genética/genética , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Ensamble y Desensamble de Cromatina , Metilación de ADN , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Procesamiento Proteico-Postraduccional
12.
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
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.
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
15.
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
16.
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
17.
Clin Cardiol ; 45(6): 650-656, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35373849

RESUMEN

BACKGROUND: Atrial fibrillation is the most important risk factor for left atrial appendage (LAA) thrombi, a potentially life-threatening condition. Thrombus resolution may prevent embolic events and allow rhythm-control strategies, which have been shown to reduce cardiovascular complications. HYPOTHESIS: There is no significant difference between phenprocoumon and non-Vitamin K-dependent oral anticoagulants (NOACs) in the resolution of LAA-thrombi in a real-world setting. METHODS: Consecutive patients with LAA-thrombi from June 2013 to June 2017 were included in an observational single-center analysis. The primary endpoint was defined as the resolution of the thrombus. The observational period was 1 year. Resolutions rates in patients on phenprocoumon or NOACs were compared and the time to resolution was analyzed. RESULTS: We identified 114 patients with LAA-thrombi. There was no significant difference in the efficacy of resolution between phenprocoumon and NOACs (p = .499) at the time of first control which took place after a mean of 58 ± 42.2 (median 48) days. At first control most thrombi were dissolved (74.6%). The analysis after set-time intervals revealed a resolution rate of 2/3 of LAA-thrombi after 8-10 weeks in the phenprocoumon and NOAC groups. After 12 weeks a higher number of thrombi had resolved in the presence of NOAC (89.3%) whereas in the presence of phenprocoumon 68.3% had resolved (p = .046). CONCLUSION: In this large observational study NOACs were found to be potent drugs for the resolution of LAA-thrombi. In addition, the resolution of LAA-thrombi was found to be faster in the presence of NOAC as compared to phenprocoumon.


Asunto(s)
Apéndice Atrial , Fibrilación Atrial , Trombosis , Administración Oral , Anticoagulantes/uso terapéutico , Apéndice Atrial/diagnóstico por imagen , Fibrilación Atrial/complicaciones , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/tratamiento farmacológico , Ecocardiografía Transesofágica , Humanos , Fenprocumón/uso terapéutico , Trombosis/etiología , Trombosis/prevención & control
18.
Front Cardiovasc Med ; 9: 943214, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36046186

RESUMEN

Despite scientific and clinical advances during the last 50 years cardiovascular disease continues to be the main cause of death worldwide. Especially patients with diabetes display a massive increased cardiovascular risk compared to patients without diabetes. Over the last two decades we have learned that cardiometabolic and cardiovascular diseases are driven by inflammation. Despite the fact that the gastrointestinal tract is one of the largest leukocyte reservoirs of our bodies, the relevance of gut immune cells for cardiovascular disease is largely unknown. First experimental evidence suggests an important relevance of immune cells in the intestinal tract for the development of metabolic and cardiovascular disease in mice. Mice specifically lacking gut immune cells are protected against obesity, diabetes, hypertension and atherosclerosis. Importantly antibody mediated inhibition of leukocyte homing into the gut showed similar protective metabolic and cardiovascular effects. Targeting gut immune cells might open novel therapeutic approaches for the treatment of cardiometabolic and cardiovascular diseases.

19.
Nat Chem Biol ; 5(10): 749-57, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19597507

RESUMEN

We report the efficacy of a new peptide with agonism at the glucagon and GLP-1 receptors that has potent, sustained satiation-inducing and lipolytic effects. Selective chemical modification to glucagon resulted in a loss of specificity, with minimal change to inherent activity. The structural basis for the co-agonism appears to be a combination of local positional interactions and a change in secondary structure. Two co-agonist peptides differing from each other only in their level of glucagon receptor agonism were studied in rodent obesity models. Administration of PEGylated peptides once per week normalized adiposity and glucose tolerance in diet-induced obese mice. Reduction of body weight was achieved by a loss of body fat resulting from decreased food intake and increased energy expenditure. These preclinical studies indicate that when full GLP-1 agonism is augmented with an appropriate degree of glucagon receptor activation, body fat reduction can be substantially enhanced without any overt adverse effects.


Asunto(s)
Péptido 1 Similar al Glucagón/agonistas , Obesidad/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , Polietilenglicoles/química , Receptores de Glucagón/agonistas , Tejido Adiposo/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Peso Corporal/efectos de los fármacos , AMP Cíclico/biosíntesis , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Obesos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Conformación Proteica
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA