Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell ; 175(3): 695-708.e13, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30293865

RESUMEN

We have uncovered the existence of extracellular vesicle (EV)-mediated signaling between cell types within the adipose tissue (AT) proper. This phenomenon became evident in our attempts at generating an adipocyte-specific knockout of caveolin 1 (cav1) protein. Although we effectively ablated the CAV1 gene in adipocytes, cav1 protein remained abundant. With the use of newly generated mouse models, we show that neighboring endothelial cells (ECs) transfer cav1-containing EVs to adipocytes in vivo, which reciprocate by releasing EVs to ECs. AT-derived EVs contain proteins and lipids capable of modulating cellular signaling pathways. Furthermore, this mechanism facilitates transfer of plasma constituents from ECs to the adipocyte. The transfer event is physiologically regulated by fasting/refeeding and obesity, suggesting EVs participate in the tissue response to changes in the systemic nutrient state. This work offers new insights into the complex signaling mechanisms that exist among adipocytes, stromal vascular cells, and, potentially, distal organs.


Asunto(s)
Adipocitos/metabolismo , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Ayuno/metabolismo , Transducción de Señal , Animales , Caveolina 1/genética , Caveolina 1/metabolismo , Línea Celular , Células Cultivadas , Endotelio Vascular/citología , Masculino , Ratones , Ratones Endogámicos C57BL
2.
Arterioscler Thromb Vasc Biol ; 43(1): 15-29, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36412195

RESUMEN

Cardiovascular disease is the most common cause of death worldwide, especially beyond the age of 65 years, with the vast majority of morbidity and mortality due to myocardial infarction and stroke. Vascular pathology stems from a combination of genetic risk, environmental factors, and the biologic changes associated with aging. The pathogenesis underlying the development of vascular aging, and vascular calcification with aging, in particular, is still not fully understood. Accumulating data suggests that genetic risk, likely compounded by epigenetic modifications, environmental factors, including diabetes and chronic kidney disease, and the plasticity of vascular smooth muscle cells to acquire an osteogenic phenotype are major determinants of age-associated vascular calcification. Understanding the molecular mechanisms underlying genetic and modifiable risk factors in regulating age-associated vascular pathology may inspire strategies to promote healthy vascular aging. This article summarizes current knowledge of concepts and mechanisms of age-associated vascular disease, with an emphasis on vascular calcification.


Asunto(s)
Enfermedades Cardiovasculares , Calcificación Vascular , Enfermedades Vasculares , Humanos , Calcificación Vascular/patología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología , Músculo Liso Vascular/patología , Enfermedades Cardiovasculares/patología , Miocitos del Músculo Liso/patología
3.
Circ Res ; 126(10): 1363-1378, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32160132

RESUMEN

RATIONALE: The PTH1R (PTH [parathyroid hormone]/PTHrP [PTH-related protein] receptor) is expressed in vascular smooth muscle (VSM) and increased VSM PTH1R signaling mitigates diet-induced arteriosclerosis in LDLR-/- mice. OBJECTIVE: To study the impact of VSM PTH1R deficiency, we generated mice SM22-Cre:PTH1R(fl/fl);LDLR-/- mice (PTH1R-VKO) and Cre-negative controls. METHODS AND RESULTS: Immunofluorescence and Western blot confirmed PTH1R expression in arterial VSM that was reduced by Cre-mediated knockout. PTH1R-VKO cohorts exhibited increased aortic collagen accumulation in vivo, and VSM cultures from PTH1R-VKO mice elaborated more collagen (2.5-fold; P=0.01) with elevated Col3a1 and Col1a1 expression. To better understand these profibrotic responses, we performed mass spectrometry on nuclear proteins extracted from Cre-negative controls and PTH1R-VKO VSM. PTH1R deficiency reduced Gata6 but upregulated the MADS (MCM1, Agamous, Deficiens, and Srf DNA-binding domain)-box transcriptional co-regulator, Mkl-1 (megakaryoblastic leukemia [translocation] 1). Co-transfection assays (Col3a1 promoter-luciferase reporter) confirmed PTH1R-mediated inhibition and Mkl-1-mediated activation of Col3a1 transcription. Regulation mapped to a conserved hybrid CT(A/T)6GG MADS-box cognate in the Col3a1 promoter. Mutations of C/G in this motif markedly reduced Col3a1 transcriptional regulation by PTH1R and Mkl-1. Upregulation of Col3a1 and Col1a1 in PTH1R-VKO VSM was inhibited by small interfering RNA targeting Mkl1 and by treatment with the Mkl-1 antagonist CCG1423 or the Rock (Rho-associated coiled-coil containing protein kinase)-2 inhibitor KD025. Chromatin precipitation demonstrated that VSM PTH1R deficiency increased Mkl-1 binding to Col3a1 and Col1a1, but not TNF, promoters. Proteomic studies of plasma extracellular vesicles and VSM from PTH1R-VKO mice identified C1r (complement component 1, r) and C1s (complement component 1, s), complement proteins involved in vascular collagen metabolism, as potential biomarkers. VSM C1r protein and C1r message were increased with PTH1R deficiency, mediated by Mkl-1-dependent transcription and inhibited by CCG1423 or KD025. CONCLUSIONS: PTH1R signaling restricts collagen production in the VSM lineage, in part, via Mkl-1 regulatory circuits that control collagen gene transcription. Strategies that maintain homeostatic VSM PTH1R signaling, as reflected in extracellular vesicle biomarkers of VSM PTH1R/Mkl-1 action, may help mitigate arteriosclerosis and vascular fibrosis.


Asunto(s)
Aterosclerosis/metabolismo , Colágeno Tipo I/metabolismo , Diabetes Mellitus/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Transactivadores/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrosis , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Receptor de Hormona Paratiroídea Tipo 1/deficiencia , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal , Transactivadores/genética , Transcripción Genética , Remodelación Vascular
4.
J Biol Chem ; 293(21): 7942-7968, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29626090

RESUMEN

In aortic vascular smooth muscle (VSM), the canonical Wnt receptor LRP6 inhibits protein arginine (Arg) methylation, a new component of noncanonical Wnt signaling that stimulates nuclear factor of activated T cells (viz NFATc4). To better understand how methylation mediates these actions, MS was performed on VSM cell extracts from control and LRP6-deficient mice. LRP6-dependent Arg methylation was regulated on >500 proteins; only 21 exhibited increased monomethylation (MMA) with concomitant reductions in dimethylation. G3BP1, a known regulator of arteriosclerosis, exhibited a >30-fold increase in MMA in its C-terminal domain. Co-transfection studies confirm that G3BP1 (G3BP is Ras-GAP SH3 domain-binding protein) methylation is inhibited by LRP6 and that G3BP1 stimulates NFATc4 transcription. NFATc4 association with VSM osteopontin (OPN) and alkaline phosphatase (TNAP) chromatin was increased with LRP6 deficiency and reduced with G3BP1 deficiency. G3BP1 activation of NFATc4 mapped to G3BP1 domains supporting interactions with RIG-I (retinoic acid inducible gene I), a stimulus for mitochondrial antiviral signaling (MAVS) that drives cardiovascular calcification in humans when mutated in Singleton-Merten syndrome (SGMRT2). Gain-of-function SGMRT2/RIG-I mutants increased G3BP1 methylation and synergized with osteogenic transcription factors (Runx2 and NFATc4). A chemical antagonist of G3BP, C108 (C108 is 2-hydroxybenzoic acid, 2-[1-(2-hydroxyphenyl)ethylidene]hydrazide CAS 15533-09-2), down-regulated RIG-I-stimulated G3BP1 methylation, Wnt/NFAT signaling, VSM TNAP activity, and calcification. G3BP1 deficiency reduced RIG-I protein levels and VSM osteogenic programs. Like G3BP1 and RIG-I deficiency, MAVS deficiency reduced VSM osteogenic signals, including TNAP activity and Wnt5-dependent nuclear NFATc4 levels. Aortic calcium accumulation is decreased in MAVS-deficient LDLR-/- mice fed arteriosclerotic diets. The G3BP1/RIG-I/MAVS relay is a component of Wnt signaling. Targeting this relay may help mitigate arteriosclerosis.


Asunto(s)
Antivirales/metabolismo , Aorta/patología , Arteriosclerosis/patología , Calcinosis/patología , ADN Helicasas/metabolismo , Miocitos del Músculo Liso/patología , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas Wnt/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Aorta/metabolismo , Arteriosclerosis/genética , Arteriosclerosis/metabolismo , Calcinosis/genética , Calcinosis/metabolismo , Calcio/metabolismo , Células Cultivadas , ADN Helicasas/genética , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , ARN Helicasas/genética , Proteínas con Motivos de Reconocimiento de ARN/genética , Receptores de LDL/fisiología , Transducción de Señal , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 37(2): 205-217, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28062508

RESUMEN

Diabetes mellitus increasingly afflicts our aging and dysmetabolic population. Type 2 diabetes mellitus and the antecedent metabolic syndrome represent the vast majority of the disease burden-increasingly prevalent in children and older adults. However, type 1 diabetes mellitus is also advancing in preadolescent children. As such, a crushing wave of cardiometabolic disease burden now faces our society. Arteriosclerotic calcification is increased in metabolic syndrome, type 2 diabetes mellitus, and type 1 diabetes mellitus-impairing conduit vessel compliance and function, thereby increasing the risk for dementia, stroke, heart attack, limb ischemia, renal insufficiency, and lower extremity amputation. Preclinical models of these dysmetabolic settings have provided insights into the pathobiology of arterial calcification. Osteochondrogenic morphogens in the BMP-Wnt signaling relay and transcriptional regulatory programs driven by Msx and Runx gene families are entrained to innate immune responses-responses activated by the dysmetabolic state-to direct arterial matrix deposition and mineralization. Recent studies implicate the endothelial-mesenchymal transition in contributing to the phenotypic drift of mineralizing vascular progenitors. In this brief overview, we discuss preclinical disease models that provide mechanistic insights-and point to challenges and opportunities to translate these insights into new therapeutic strategies for our patients afflicted with diabetes mellitus and its arteriosclerotic complications.


Asunto(s)
Arterias/metabolismo , Aterosclerosis/etiología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Angiopatías Diabéticas/etiología , Investigación Biomédica Traslacional , Calcificación Vascular/etiología , Animales , Animales Modificados Genéticamente , Arterias/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Angiopatías Diabéticas/metabolismo , Angiopatías Diabéticas/patología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hiperlipidemias/complicaciones , Hiperlipidemias/genética , Masculino , Fenotipo , Placa Aterosclerótica , Ratas , Transducción de Señal , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
6.
Curr Opin Lipidol ; 28(5): 387-396, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28723729

RESUMEN

PURPOSE OF REVIEW: Cardiometabolic diseases increasingly afflict our aging, dysmetabolic population. Complex signals regulating low-density lipoprotein receptor-related protein (LRP) and frizzled protein family members - the plasma membrane receptors for the cadre of Wnt polypeptide morphogens - contribute to the control of cardiovascular homeostasis. RECENT FINDINGS: Both canonical (ß-catenin-dependent) and noncanonical (ß-catenin-independent) Wnt signaling programs control vascular smooth muscle (VSM) cell phenotypic modulation in cardiometabolic disease. LRP6 limits VSM proliferation, reduces arteriosclerotic transcriptional reprogramming, and preserves insulin sensitivity while LRP5 restrains foam cell formation. Adipose, skeletal muscle, macrophages, and VSM have emerged as important sources of circulating Wnt ligands that are dynamically regulated during the prediabetes-diabetes transition with cardiometabolic consequences. Platelets release Dkk1, a LRP5/LRP6 inhibitor that induces endothelial inflammation and the prosclerotic endothelial-mesenchymal transition. By contrast, inhibitory secreted frizzled-related proteins shape the Wnt signaling milieu to limit myocardial inflammation with ischemia-reperfusion injury. VSM sclerostin, an inhibitor of canonical Wnt signaling in bone, restrains remodeling that predisposes to aneurysm formation, and is downregulated in aneurysmal vessels by epigenetic methylation. SUMMARY: Components of the Wnt signaling cascade represent novel targets for pharmacological intervention in cardiometabolic disease. Conversely, strategies targeting the Wnt signaling cascade for other therapeutic purposes will have cardiovascular consequences that must be delineated to establish clinically useful pharmacokinetic-pharmacodynamic relationships.


Asunto(s)
Enfermedades Cardiovasculares/patología , Vía de Señalización Wnt , Animales , Enfermedades Cardiovasculares/metabolismo , Humanos , Receptores de Lipoproteína/metabolismo
7.
Circ Res ; 117(2): 142-56, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-26034040

RESUMEN

RATIONALE: Wnt signaling regulates key aspects of diabetic vascular disease. OBJECTIVE: We generated SM22-Cre;LRP6(fl/fl);LDLR(-/-) mice to determine contributions of Wnt coreceptor low-density lipoprotein receptor-related protein 6 (LRP6) in the vascular smooth muscle lineage of male low-density lipoprotein receptor-null mice, a background susceptible to diet (high-fat diet)-induced diabetic arteriosclerosis. METHODS AND RESULTS: As compared with LRP6(fl/fl);LDLR(-/-) controls, SM22-Cre;LRP6(fl/fl);LDLR(-/-) (LRP6-VKO) siblings exhibited increased aortic calcification on high-fat diet without changes in fasting glucose, lipids, or body composition. Pulse wave velocity (index of arterial stiffness) was also increased. Vascular calcification paralleled enhanced aortic osteochondrogenic programs and circulating osteopontin (OPN), a matricellular regulator of arteriosclerosis. Survey of ligands and Frizzled (Fzd) receptor profiles in LRP6-VKO revealed upregulation of canonical and noncanonical Wnts alongside Fzd10. Fzd10 stimulated noncanonical signaling and OPN promoter activity via an upstream stimulatory factor (USF)-activated cognate inhibited by LRP6. RNA interference revealed that USF1 but not USF2 supports OPN expression in LRP6-VKO vascular smooth muscle lineage, and immunoprecipitation confirmed increased USF1 association with OPN chromatin. ML141, an antagonist of cdc42/Rac1 noncanonical signaling, inhibited USF1 activation, osteochondrogenic programs, alkaline phosphatase, and vascular smooth muscle lineage calcification. Mass spectrometry identified LRP6 binding to protein arginine methyltransferase (PRMT)-1, and nuclear asymmetrical dimethylarginine modification was increased with LRP6-VKO. RNA interference demonstrated that PRMT1 inhibits OPN and TNAP, whereas PRMT4 supports expression. USF1 complexes containing the histone H3 asymmetrically dimethylated on Arg-17 signature of PRMT4 are increased with LRP6-VKO. Jmjd6, a demethylase downregulated with LRP6 deficiency, inhibits OPN and TNAP expression, USF1: histone H3 asymmetrically dimethylated on Arg-17 complex formation, and transactivation. CONCLUSIONS: LRP6 restrains vascular smooth muscle lineage noncanonical signals that promote osteochondrogenic differentiation, mediated in part via USF1- and arginine methylation-dependent relays.


Asunto(s)
Arteriosclerosis/prevención & control , Calcinosis/prevención & control , Diabetes Mellitus Experimental/complicaciones , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/fisiología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo , Receptores de LDL/deficiencia , Vía de Señalización Wnt , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Arteriosclerosis/etiología , Arteriosclerosis/metabolismo , Calcinosis/etiología , Calcinosis/metabolismo , Diabetes Mellitus Experimental/patología , Grasas de la Dieta/efectos adversos , Receptores Frizzled/fisiología , Regulación de la Expresión Génica/fisiología , Histonas/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/deficiencia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/patología , Osteopontina/biosíntesis , Osteopontina/genética , Comunicación Paracrina , Mapeo de Interacción de Proteínas , Proteína-Arginina N-Metiltransferasas/metabolismo , Receptores de Superficie Celular , Receptores de LDL/genética , Factores Estimuladores hacia 5'/fisiología , Rigidez Vascular/fisiología
8.
Circ Res ; 113(2): 198-208, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23833294

RESUMEN

Calcific aortic valve disease (CAVD) increasingly afflicts our aging population. One third of our elderly have echocardiographic or radiological evidence of calcific aortic valve sclerosis, an early and subclinical form of CAVD. Age, sex, tobacco use, hypercholesterolemia, hypertension, and type II diabetes mellitus all contribute to the risk of disease that has worldwide distribution. On progression to its most severe form, calcific aortic stenosis, CAVD becomes debilitating and devastating, and 2% of individuals >60 years are affected by calcific aortic stenosis to the extent that surgical intervention is required. No effective pharmacotherapies exist for treating those at risk for clinical progression. It is becoming increasingly apparent that a diverse spectrum of cellular and molecular mechanisms converge to regulate valvular calcium load; this is evidenced not only in histopathologic heterogeneity of CAVD, but also from the multiplicity of cell types that can participate in valve biomineralization. In this review, we highlight our current understanding of CAVD disease biology, emphasizing molecular and cellular aspects of its regulation. We end by pointing to important biological and clinical questions that must be answered to enable sophisticated disease staging and the development of new strategies to treat CAVD medically.


Asunto(s)
Calcinosis/patología , Cardiomiopatías/patología , Cardiopatías Congénitas/patología , Enfermedades de las Válvulas Cardíacas/patología , Miocitos Cardíacos/patología , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Enfermedad de la Válvula Aórtica Bicúspide , Calcinosis/genética , Calcinosis/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/metabolismo , Humanos , Miocitos Cardíacos/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 34(11): 2387-93, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25189570

RESUMEN

Calcific aortic valve disease (CAVD) is increasingly prevalent worldwide with significant morbidity and mortality. Therapeutic options beyond surgical valve replacement are currently limited. In 2011, the National Heart Lung and Blood Institute assembled a working group on aortic stenosis. This group identified CAVD as an actively regulated disease process in need of further study. As a result, the Alliance of Investigators on CAVD was formed to coordinate and promote CAVD research, with the goals of identifying individuals at risk, developing new therapeutic approaches, and improving diagnostic methods. The group is composed of cardiologists, geneticists, imaging specialists, and basic science researchers. This report reviews the current status of CAVD research and treatment strategies with identification of areas in need of additional investigation for optimal management of this patient population.


Asunto(s)
Estenosis de la Válvula Aórtica/terapia , Válvula Aórtica/patología , Investigación Biomédica/tendencias , Calcinosis/terapia , Cardiopatías Congénitas/terapia , Enfermedades de las Válvulas Cardíacas/terapia , Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/diagnóstico , Estenosis de la Válvula Aórtica/fisiopatología , Enfermedad de la Válvula Aórtica Bicúspide , Calcinosis/diagnóstico , Calcinosis/fisiopatología , Procedimientos Quirúrgicos Cardíacos , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/fisiopatología , Enfermedades de las Válvulas Cardíacas/diagnóstico , Enfermedades de las Válvulas Cardíacas/fisiopatología , Implantación de Prótesis de Válvulas Cardíacas , Hemodinámica/fisiología , Humanos , Transducción de Señal/fisiología
10.
Arterioscler Thromb Vasc Biol ; 33(7): 1679-89, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23685555

RESUMEN

OBJECTIVE: Endothelial cells (ECs) can undergo an endothelial-mesenchymal transition with tissue fibrosis. Wnt- and Msx2-regulated signals participate in arteriosclerotic fibrosis and calcification. We studied the impact of Wnt7, Msx2, and Dkk1, a Wnt7 antagonist, on endothelial-mesenchymal transition in primary aortic ECs. APPROACH AND RESULTS: Transduction of aortic ECs with vectors expressing Dkk1 suppressed EC differentiation and induced a mineralizing myofibroblast phenotype. Dkk1 suppressed claudin 5, PECAM, cadherin 5 (Cdh5), Tie1, and Tie2. Dkk1 converted the cuboidal cell monolayer into a spindle-shaped multilayer and inhibited EC cord formation. Myofibroblast and osteogenic markers, SM22, type I collagen, Osx, Runx2, and alkaline phosphatase, were upregulated by Dkk1 via activin-like kinase/Smad pathways. Dkk1 increased fibrotic mineralization of aortic ECs cultured under osteogenic conditions--the opposite of mesenchymal cell responses. Msx2 and Wnt7b maintained morphology and upregulated markers of differentiated ECs. Deleting EC Wnt7b with the Cdh5-Cre transgene in Wnt7b(fl/fl);LDLR(-/-) mice upregulated aortic osteogenic genes (Osx, Sox9, Runx2, and Msx2) and nuclear phospho-Smad1/5, and increased collagen and calcium accumulation. CONCLUSIONS: Dkk1 enhances endothelial-mesenchymal transition in aortic ECs, whereas Wnt7b and Msx2 signals preserve EC phenotype. EC responses to Dkk1, Wnt7b, and Msx2 are the opposite of mesenchymal responses, coupling EC phenotypic stability with osteofibrogenic predilection during arteriosclerosis.


Asunto(s)
Aorta/metabolismo , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Miofibroblastos/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Arteriosclerosis/genética , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Biomarcadores/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Bovinos , Diferenciación Celular , Forma de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Fibrosis , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Noqueados , Miofibroblastos/patología , Neovascularización Fisiológica , Osificación Heterotópica/metabolismo , Fenotipo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción Genética , Transfección , Proteínas Wnt/deficiencia , Proteínas Wnt/genética
11.
Trends Endocrinol Metab ; 35(7): 648-660, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38429163

RESUMEN

Primary hyperparathyroidism (pHPT) afflicts our aging population with an incidence approaching 50 per 100 000 patient-years at a female:male ratio of ~3:1. Decisions surrounding surgical management are currently driven by age, hypercalcemia severity, presence of osteoporosis, renal insufficiency, or hypercalciuria with or without nephrolithiasis. Cardiovascular (CV) disease (CVD) is not systematically considered. This is notable since the parathyroid hormone (PTH) 1 receptor (PTH1R) is biologically active in the vasculature, and adjusted CV mortality risk is increased almost threefold in individuals with pHPT who do not meet contemporary recommendations for surgical cure. We provide an overview of epidemiology, pharmacology, and physiology that highlights the need to: (i) identify biomarkers that establish a healthy 'set point' for CV PTH1R signaling tone; (ii) better understand the pharmacokinetic-pharmacodynamic (PK-PD) relationships of PTH1R ligands in CV homeostasis; and (iii) incorporate CVD risk assessment into the management of hyperparathyroidism.


Asunto(s)
Enfermedades Cardiovasculares , Homeostasis , Hormona Paratiroidea , Receptor de Hormona Paratiroídea Tipo 1 , Transducción de Señal , Humanos , Enfermedades Cardiovasculares/metabolismo , Homeostasis/fisiología , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Hormona Paratiroidea/metabolismo , Transducción de Señal/fisiología , Hiperparatiroidismo Primario/metabolismo , Animales
14.
Circ Res ; 108(11): 1378-80, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21617134

RESUMEN

Vascular calcification increasingly afflicts our aging, dysmetabolic population. Once considered only a passive process of dead and dying cells, data from multiple laboratories worldwide have converged to demonstrate that vascular calcification is a highly regulated form of biomineralization. The goal of this thematic review series is to highlight what is known concerning the biological "players" and "game rules" with respect to vascular mineral metabolism. Armed with this understanding, it is hoped that novel therapeutic strategies can be crafted to prevent and treat vascular calcium accrual, to the benefit of our patients afflicted with arteriosclerotic valvular and vascular diseases.


Asunto(s)
Calcinosis , Enfermedades Vasculares , Calcinosis/metabolismo , Calcinosis/patología , Calcinosis/fisiopatología , Humanos , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología , Enfermedades Vasculares/fisiopatología
15.
Circ Res ; 109(5): 564-77, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21852555

RESUMEN

Vascular calcification increasingly afflicts our aging, dysmetabolic population. Once considered only a passive process of dead and dying cells, vascular calcification has now emerged as a highly regulated form of biomineralization organized by collagenous and elastin extracellular matrices. During skeletal bone formation, paracrine epithelial-mesenchymal and endothelial-mesenchymal interactions control osteochondrocytic differentiation of multipotent mesenchymal progenitor cells. These paracrine osteogenic signals, mediated by potent morphogens of the bone morphogenetic protein and wingless-type MMTV integration site family member (Wnt) superfamilies, are also active in the programming of arterial osteoprogenitor cells during vascular and valve calcification. Inflammatory cytokines, reactive oxygen species, and oxylipids-increased in the clinical settings of atherosclerosis, diabetes, and uremia that promote arteriosclerotic calcification-elicit the ectopic vascular activation of osteogenic morphogens. Specific extracellular and intracellular inhibitors of bone morphogenetic protein-Wnt signaling have been identified as contributing to the regulation of osteogenic mineralization during development and disease. These inhibitory pathways and their regulators afford the development of novel therapeutic strategies to prevent and treat valve and vascular sclerosis.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Enfermedades de las Válvulas Cardíacas/patología , Osteocitos/patología , Osteogénesis/fisiología , Enfermedades Vasculares/patología , Animales , Enfermedades de las Válvulas Cardíacas/metabolismo , Humanos , Comunicación Paracrina/fisiología , Esclerosis , Enfermedades Vasculares/metabolismo
17.
Endocrinology ; 165(2)2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38123514

RESUMEN

Wnt16 is expressed in bone and arteries, and maintains bone mass in mice and humans, but its role in cardiovascular physiology is unknown. We show that Wnt16 protein accumulates in murine and human vascular smooth muscle (VSM). WNT16 genotypes that convey risk for bone frailty also convey risk for cardiovascular events in the Dallas Heart Study. Murine Wnt16 deficiency, which causes postnatal bone loss, also reduced systolic blood pressure. Electron microscopy demonstrated abnormal VSM mitochondrial morphology in Wnt16-null mice, with reductions in mitochondrial respiration. Following angiotensin-II (AngII) infusion, thoracic ascending aorta (TAA) dilatation was greater in Wnt16-/- vs Wnt16+/+ mice (LDLR-/- background). Acta2 (vascular smooth muscle alpha actin) deficiency has been shown to impair contractile phenotype and worsen TAA aneurysm with concomitant reductions in blood pressure. Wnt16 deficiency reduced expression of Acta2, SM22 (transgelin), and other contractile genes, and reduced VSM contraction induced by TGFß. Acta2 and SM22 proteins were reduced in Wnt16-/- VSM as was Ankrd1, a prototypic contractile target of Yap1 and Taz activation via TEA domain (TEAD)-directed transcription. Wnt16-/- VSM exhibited reduced nuclear Taz and Yap1 protein accumulation. SiRNA targeting Wnt16 or Taz, but not Yap1, phenocopied Wnt16 deficiency, and Taz siRNA inhibited contractile gene upregulation by Wnt16. Wnt16 incubation stimulated mitochondrial respiration and contraction (reversed by verteporfin, a Yap/Taz inhibitor). SiRNA targeting Taz inhibitors Ccm2 and Lats1/2 mimicked Wnt16 treatment. Wnt16 stimulated Taz binding to Acta2 chromatin and H3K4me3 methylation. TEAD cognates in the Acta2 promoter conveyed transcriptional responses to Wnt16 and Taz. Wnt16 regulates cardiovascular physiology and VSM contractile phenotype, mediated via Taz signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Músculo Liso Vascular , Proteínas Wnt , Animales , Humanos , Masculino , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Fenotipo , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/metabolismo , Proteínas Wnt/genética
18.
J Biol Chem ; 286(4): 2933-45, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21098489

RESUMEN

Endothelial dysfunction leads to lethal vascular complications in diabetes and related metabolic disorders. Here, we demonstrate that de novo lipogenesis, an insulin-dependent process driven by the multifunctional enzyme fatty-acid synthase (FAS), maintains endothelial function by targeting endothelial nitric-oxide synthase (eNOS) to the plasma membrane. In mice with endothelial inactivation of FAS (FASTie mice), eNOS membrane content and activity were decreased. eNOS and FAS were physically associated; eNOS palmitoylation was decreased in FAS-deficient cells, and incorporation of labeled carbon into eNOS-associated palmitate was FAS-dependent. FASTie mice manifested a proinflammatory state reflected as increases in vascular permeability, endothelial inflammatory markers, leukocyte migration, and susceptibility to LPS-induced death that was reversed with an NO donor. FAS-deficient endothelial cells showed deficient migratory capacity, and angiogenesis was decreased in FASTie mice subjected to hindlimb ischemia. Insulin induced FAS in endothelial cells freshly isolated from humans, and eNOS palmitoylation was decreased in mice with insulin-deficient or insulin-resistant diabetes. Thus, disrupting eNOS bioavailability through impaired lipogenesis identifies a novel mechanism coordinating nutritional status and tissue repair that may contribute to diabetic vascular disease.


Asunto(s)
Células Endoteliales/metabolismo , Ácido Graso Sintasas/metabolismo , Lipogénesis/fisiología , Lipoilación/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/metabolismo , Ácido Graso Sintasas/genética , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Leucocitos/metabolismo , Lipogénesis/efectos de los fármacos , Lipopolisacáridos/farmacología , Lipoilación/efectos de los fármacos , Ratones , Ratones Mutantes , Donantes de Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética
20.
Circ Res ; 107(2): 271-82, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20489161

RESUMEN

RATIONALE: Vascular fibrosis and calcification contribute to diabetic arteriosclerosis, impairing Windkessel physiology necessary for distal tissue perfusion. Wnt family members, upregulated in arteries by the low-grade inflammation of "diabesity," stimulate type I collagen expression and osteogenic mineralization of mesenchymal progenitors via beta-catenin. Conversely, parathyroid hormone (PTH) inhibits aortic calcification in low-density lipoprotein receptor (LDLR)-deficient mice fed high fat diabetogenic diets (HFD). OBJECTIVE: We sought to determine the impact of vascular PTH receptor (PTH1R) activity on arteriosclerotic Wnt/beta-catenin signaling in vitro and in vivo. We generated SM-caPTH1R transgenic mice, a model in which the constitutively active PTH1R variant H223R (caPTH1R) is expressed only in the vasculature. METHODS AND RESULTS: The caPTH1R inhibited Wnt/beta-catenin signaling, collagen production, and vascular smooth muscle cell proliferation and calcification in vitro. Transgenic SM-caPTH1R;LDLR(+/-) mice fed HFD develop diabesity, with no improvements in fasting serum glucose, cholesterol, weight, body composition, or bone mass versus LDLR(+/-) siblings. SM-caPTH1R downregulated aortic Col1A1, Runx2, and Nox1 expression without altering TNF, Msx2, Wnt7a/b, or Nox4. The SM-caPTH1R transgene decreased aortic beta-catenin protein accumulation and signaling in diabetic LDLR(+/-) mice. Levels of aortic superoxide (a precursor of peroxide that activates pro-matrix metalloproteinase 9 and osteogenic signaling in vascular smooth muscle cells) were suppressed by the SM-caPTH1R transgene. Aortic calcification, collagen accumulation, and wall thickness were concomitantly reduced, enhancing vessel distensibility. CONCLUSIONS: Cell-autonomous vascular smooth muscle cell PTH1R activity inhibits arteriosclerotic Wnt/beta-catenin signaling and reduces vascular oxidative stress, thus limiting aortic type I collagen and calcium accrual in diabetic LDLR-deficient mice.


Asunto(s)
Arteriosclerosis/metabolismo , Diabetes Mellitus/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Arteriosclerosis/genética , Arteriosclerosis/patología , Calcinosis/metabolismo , Calcinosis/patología , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Modelos Animales de Enfermedad , Fibrosis , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/patología , Músculo Liso Vascular/patología , Mutación , Miocitos del Músculo Liso/patología , Estrés Oxidativo , Ratas , Receptor de Hormona Paratiroídea Tipo 1/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética , Superóxidos/metabolismo , Transcripción Genética , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA