Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 326(3): G264-G273, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38258487

RESUMEN

Exercise as a lifestyle modification is a frontline therapy for nonalcoholic fatty liver disease (NAFLD), but how components of exercise attenuate steatosis is unclear. To uncouple the effect of increased muscle mass from weight loss in obesity, myostatin knockout mice were bred on a lean and obese db/db background. Myostatin deletion increases gastrocnemius (Gastrocn.) mass and reduces hepatic steatosis and hepatic sterol regulatory element binding protein 1 (Srebp1) expression in obese mice, with no impact on adiposity or body weight. Interestingly, hypermuscularity reduces hepatic NADPH oxidase 1 (Nox1) expression but not NADPH oxidase 4 (Nox4) in db/db mice. To evaluate a deterministic function of Nox1 on steatosis, Nox1 knockout mice were bred on a lean and db/db background. NOX1 deletion significantly attenuates hepatic oxidant stress, steatosis, and Srebp1 programming in obese mice to parallel hypermuscularity, with no improvement in adiposity, glucose control, or hypertriglyceridemia to suggest off-target effects. Directly assessing the role of NOX1 on SREBP1, insulin (Ins)-mediated SREBP1 expression was significantly increased in either NOX1, NADPH oxidase organizer 1 (NOXO1), and NADPH oxidase activator 1 (NOXA1) or NOX5-transfected HepG2 cells versus ?-galactosidase control virus, indicating superoxide is the key mechanistic agent for the actions of NOX1 on SREBP1. Metabolic Nox1 regulators were evaluated using physiological, genetic, and diet-induced animal models that modulated upstream glucose and insulin signaling, identifying hyperinsulinemia as the key metabolic derangement explaining Nox1-induced steatosis in obesity. GEO data revealed that hepatic NOX1 predicts steatosis in obese humans with biopsy-proven NAFLD. Taken together, these data suggest that hypermuscularity attenuates Srebp1 expression in db/db mice through a NOX1-dependent mechanism.NEW & NOTEWORTHY This study documents a novel mechanism by which changes in body composition, notably increased muscle mass, protect against fatty liver disease. This mechanism involves NADPH oxidase 1 (NOX1), an enzyme that increases superoxide and increases insulin signaling, leading to increased fat accumulation in the liver. NOX1 may represent a new early target for preventing fatty liver to stave off later liver diseases such as cirrhosis or liver cancer.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Ratones , Insulina/metabolismo , Hígado/metabolismo , Ratones Noqueados , Ratones Obesos , Músculo Esquelético/metabolismo , Miostatina , NADPH Oxidasa 1/metabolismo , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/metabolismo , Superóxidos/metabolismo
2.
Gastroenterology ; 165(1): 71-87, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37030336

RESUMEN

BACKGROUND & AIMS: Visceral smooth muscle cells (SMCs) are an integral component of the gastrointestinal (GI) tract that regulate GI motility. SMC contraction is regulated by posttranslational signaling and the state of differentiation. Impaired SMC contraction is associated with significant morbidity and mortality, but the mechanisms regulating SMC-specific contractile gene expression, including the role of long noncoding RNAs (lncRNAs), remain largely unexplored. Herein, we reveal a critical role of Carmn (cardiac mesoderm enhancer-associated noncoding RNA), an SMC-specific lncRNA, in regulating visceral SMC phenotype and contractility of the GI tract. METHODS: Genotype-Tissue Expression and publicly available single-cell RNA sequencing (scRNA-seq) data sets from embryonic, adult human, and mouse GI tissues were interrogated to identify SMC-specific lncRNAs. The functional role of Carmn was investigated using novel green fluorescent protein (GFP) knock-in (KI) reporter/knock-out (KO) mice. Bulk RNA-seq and single nucleus RNA sequencing (snRNA-seq) of colonic muscularis were used to investigate underlying mechanisms. RESULTS: Unbiased in silico analyses and GFP expression patterns in Carmn GFP KI mice revealed that Carmn is highly expressed in GI SMCs in humans and mice. Premature lethality was observed in global Carmn KO and inducible SMC-specific KO mice due to GI pseudo-obstruction and severe distension of the GI tract, with dysmotility in cecum and colon segments. Histology, GI transit, and muscle myography analysis revealed severe dilation, significantly delayed GI transit, and impaired GI contractility in Carmn KO vs control mice. Bulk RNA-seq of GI muscularis revealed that loss of Carmn promotes SMC phenotypic switching, as evidenced by up-regulation of extracellular matrix genes and down-regulation of SMC contractile genes, including Mylk, a key regulator of SMC contraction. snRNA-seq further revealed SMC Carmn KO not only compromised myogenic motility by reducing contractile gene expression but also impaired neurogenic motility by disrupting cell-cell connectivity in the colonic muscularis. These findings may have translational significance, because silencing CARMN in human colonic SMCs significantly attenuated contractile gene expression, including MYLK, and decreased SMC contractility. Luciferase reporter assays showed that CARMN enhances the transactivation activity of the master regulator of SMC contractile phenotype, myocardin, thereby maintaining the GI SMC myogenic program. CONCLUSIONS: Our data suggest that Carmn is indispensable for maintaining GI SMC contractile function in mice and that loss of function of CARMN may contribute to human visceral myopathy. To our knowledge this is the first study showing an essential role of lncRNA in the regulation of visceral SMC phenotype.


Asunto(s)
Contracción Muscular , Músculo Liso , ARN Largo no Codificante , Animales , Humanos , Ratones , Diferenciación Celular , Células Cultivadas , Ratones Noqueados , Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 43(10): e381-e395, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37586054

RESUMEN

BACKGROUND: Obesity is associated with increased risk of cardiovascular disease, but underlying mechanisms remain elusive. Metabolic dysfunction, especially hyperglycemia, is thought to be a major contributor, but how glucose impacts vascular function is unclear. GAL3 (galectin-3) is a sugar-binding lectin upregulated by hyperglycemia, but its role as a causative mechanism of cardiovascular disease remains poorly understood. Therefore, the objective of this study was to determine the role of GAL3 in regulating microvascular endothelial vasodilation in obesity. METHODS: GAL3 was measured and found to be markedly increased in the plasma of overweight and obese patients, as well as in the microvascular endothelium of diabetic patients. To investigate causative mechanisms in cardiovascular disease, mice deficient in GAL3 were bred with obese db/db mice to generate lean, lean GAL3 knockout, obese, and obese GAL3 knockout genotypes. Endothelial cell-specific GAL3 knockout mice with novel AAV-induced obesity recapitulated whole-body knockout studies to confirm cell specificity. RESULTS: Deletion of GAL3 did not alter body mass, adiposity, or plasma indices of glycemia and lipidemia, but levels of plasma reactive oxygen species as assessed by plasma thiobarbituric acid reactive substances were normalized in obese GAL3 knockout mice. Obese mice exhibited profound endothelial dysfunction and hypertension, both of which were rescued by GAL3 deletion. Isolated microvascular endothelial cells from obese mice had increased expression of NOX1 (nicotinamide adenine dinucleotide phosphate oxidase 1), which we have previously shown to contribute to increased oxidative stress and endothelial dysfunction, which was normalized in microvascular endothelium from mice lacking GAL3. Cell-specific deletion confirmed that endothelial GAL3 regulates obesity-induced NOX1 overexpression and subsequent microvascular function. Furthermore, improvement of metabolic syndrome by increasing muscle mass, improving insulin signaling, or treating with metformin decreased microvascular GAL3, and thereby NOX1, expression levels. CONCLUSIONS: Deletion of GAL3 normalizes microvascular endothelial function in obese db/db mice, likely through a NOX1-mediated mechanism. Pathological levels of GAL3, and in turn NOX1, are amenable to improvements in metabolic status, presenting a potential therapeutic target to ameliorate pathological cardiovascular consequences of obesity.


Asunto(s)
Enfermedades Cardiovasculares , Hiperglucemia , Hipertensión , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Hiperglucemia/metabolismo , Ratones Noqueados , Ratones Obesos , NADPH Oxidasa 1/metabolismo , NADPH Oxidasas/metabolismo , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , Estrés Oxidativo
4.
Am J Physiol Gastrointest Liver Physiol ; 323(4): G387-G400, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35997288

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is associated with disruption of homeostatic lipid metabolism, but underlying processes are poorly understood. One possible mechanism is impairment in hepatic circadian rhythm, which regulates key lipogenic mediators in the liver and whose circadian oscillation is diminished in obesity. Nobiletin enhances biological rhythms by activating RAR-related orphan receptor nuclear receptor, protecting against metabolic syndrome in a clock-dependent manner. The effect of nobiletin in NAFLD is unclear. In this study, we investigate the clock-enhancing effects of nobiletin in genetically obese (db/db) PER2::LUCIFERASE reporter mice with fatty liver. We report microarray expression data suggesting hepatic circadian signaling is impaired in db/db mice with profound hepatic steatosis. Circadian PER2 activity, as assessed by mRNA and luciferase assay, was significantly diminished in liver of db/db PER2::LUCIFERASE reporter mice. Continuous animal monitoring systems and constant dark studies suggest the primary circadian defect in db/db mice lies within peripheral hepatic oscillators and not behavioral rhythms or the master clock. In vitro, nobiletin restored PER2 amplitude in lipid-laden PER2::LUCIFERASE reporter macrophages. In vivo, nobiletin dramatically upregulated core clock gene expression, hepatic PER2 activity, and ameliorated steatosis in db/db PER2::LUCIFERASE reporter mice. Mechanistically, nobiletin reduced serum insulin levels, decreased hepatic Srebp1c, Acaca1, Tnfα, and Fgf21 expression, but did not improve Plin2, Plin5, or Cpt1, suggesting nobiletin attenuates steatosis in db/db mice via downregulation of hepatic lipid accumulation. These data suggest restoring endogenous rhythm with nobiletin resolves steatosis in obesity, proposing that hypothesis that targeting the biological clock may be an attractive therapeutic strategy for NAFLD.NEW & NOTEWORTHY NAFLD is the most common chronic liver disease, but underlying mechanisms are unclear. We show here that genetically obese (db/db) mice with fatty liver have impaired hepatic circadian rhythm. Hepatic Per2 expression and PER2 reporter activity are diminished in db/db PER2::LUCIFERASE mice. The biological clock-enhancer nobiletin restores hepatic PER2 in db/db PER2::LUCIFERASE mice, resolving steatosis via downregulation of Srebp1c. These studies suggest targeting the circadian clock may be beneficial strategy in NAFLD.


Asunto(s)
Relojes Circadianos , Insulinas , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Ritmo Circadiano , Ratones Obesos , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Relojes Circadianos/genética , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Luciferasas/metabolismo , Luciferasas/farmacología , ARN Mensajero , Insulinas/metabolismo , Insulinas/farmacología , Lípidos/farmacología , Ratones Endogámicos C57BL
5.
Circ Res ; 121(5): 502-511, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28684629

RESUMEN

RATIONALE: Early vascular changes in metabolic disease that precipitate the development of cardiovascular complications are largely driven by reactive oxygen species accumulation, yet the extent to which excess reactive oxygen species derive from specific NADPH oxidase isoforms remains ill defined. OBJECTIVE: Identify the role of Nox1 in the development of microvascular dysfunction in metabolic disease. METHODS AND RESULTS: Four genotypes were generated by breeding Nox1 knockout mice with db/db mice: lean (HdbWnox1), lean Nox1 knockout (HdbKnox1), obese (KdbWnox1), and obese KK (KdbKnox1). The degree of adiposity, insulin resistance, and dyslipidemia in KW mice was not influenced by Nox1 deletion as determined by nuclear magnetic resonance spectroscopy, glucose tolerance tests, and plasma analyses. Endothelium-dependent responses to acetylcholine in pressurized mesenteric arteries were reduced in KW versus HW (P<0.01), whereas deletion of Nox1 in KW mice normalized dilation. Vasodilator responses after inhibition of NO synthase blunted acetylcholine responses in KK and lean controls, but had no impact in KW, attributing recovered dilatory capacity in KK to normalization of NO. Acetylcholine responses were improved (P<0.05) with Tempol, and histochemistry revealed oxidative stress in KW animals, whereas Tempol had no impact and reactive oxygen species staining was negligible in KK. Blunted dilatory responses to an NO donor and loss of myogenic tone in KW animals were also rescued with Nox1 deletion. CONCLUSIONS: Nox1 deletion reduces oxidant load and restores microvascular health in db/db mice without influencing the degree of metabolic dysfunction. Therefore, targeted Nox1 inhibition may be effective in the prevention of vascular complications.


Asunto(s)
Eliminación de Gen , Enfermedades Metabólicas/genética , Microvasos/fisiología , Músculo Liso Vascular/fisiología , NADH NADPH Oxidorreductasas/deficiencia , NADH NADPH Oxidorreductasas/genética , Animales , Glucemia/metabolismo , Masculino , Enfermedades Metabólicas/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , NADPH Oxidasa 1 , Estrés Oxidativo/fisiología
7.
Clin Sci (Lond) ; 130(11): 881-93, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26935109

RESUMEN

Protein tyrosine phosphatase 1b (Ptp1b) is a negative regulator of leptin and insulin-signalling pathways. Its targeted deletion in proopiomelanocortin (POMC) neurons protects mice from obesity and diabetes by increasing energy expenditure. Inflammation accompanies increased energy expenditure. Therefore, the present study aimed to determine whether POMC-Ptp1b deletion increases energy expenditure via an inflammatory process, which would impair endothelial function. We characterized the metabolic and cardiovascular phenotypes of Ptp1b+/+ and POMC-Ptp1b-/- mice. Clamp studies revealed that POMC-Ptp1b deletion reduced body fat and increased energy expenditure as evidenced by a decrease in feed efficiency and an increase in oxygen consumption and respiratory exchange ratio. POMC-Ptp1b deletion induced a 2.5-fold increase in plasma tumour necrosis factor α (TNF-α) levels and elevated body temperature. Vascular studies revealed an endothelial dysfunction in POMC-Ptp1b-/- mice. Nitric oxide synthase inhibition [N-nitro-L-arginine methyl ester (L-NAME)] reduced relaxation to a similar extent in Ptp1b+/+ and POMC-Ptp1b-/- mice. POMC-Ptp1b deletion decreased ROS-scavenging enzymes [superoxide dismutases (SODs)] whereas it increased ROS-generating enzymes [NADPH oxidases (NOXs)] and cyclooxygenase-2 (COX-1) expression, in aorta. ROS scavenging or NADPH oxidase inhibition only partially improved relaxation whereas COX-2 inhibition and thromboxane-A2 (TXA2) antagonism fully restored relaxation in POMC-Ptp1b-/- mice Chronic treatment with the soluble TNF-α receptor etanercept decreased body temperature, restored endothelial function and reestablished aortic COX-2, NOXs and SOD expression to their baseline levels in POMC-Ptp1b-/- mice. However, etanercept promoted body weight gain and decreased energy expenditure in POMC-Ptp1b-/- mice. POMC-Ptp1b deletion increases plasma TNF-α levels, which contribute to body weight regulation via increased energy expenditure and impair endothelial function via COX-2 and ROS-dependent mechanisms.


Asunto(s)
Células Endoteliales/metabolismo , Metabolismo Energético/genética , Neuronas/metabolismo , Proopiomelanocortina/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Factor de Necrosis Tumoral alfa/metabolismo , Tejido Adiposo/metabolismo , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Ciclooxigenasa 2/metabolismo , Metabolismo Energético/fisiología , Ratones Transgénicos , Obesidad/genética , Obesidad/metabolismo , Factor de Necrosis Tumoral alfa/genética , Aumento de Peso/genética
8.
Microcirculation ; 22(4): 257-66, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25660131

RESUMEN

OBJECTIVE: Perturbation of daily rhythm increases cardiovascular risk. The aim of this study was to determine whether obesity alters circadian gene expression and microvascular function in lean mice and obese (db/db) mice. METHODS: Mice were subjected to normal LD or DD to alter circadian rhythm. Metabolic parameters and microvascular vasoreactivity were evaluated. Array studies were conducted in the am and pm cycles to assess the rhythmicity of the entire genomics. Rhythmic expression of specific clock genes (Bmal1, Clock, Npas2, Per1, Per2, and Cry1), clock output genes (dbp), and vascular relaxation-related genes (eNOS, GTPCH1) were assessed. RESULTS: Obesity was associated with metabolic dysfunction and impaired endothelial dilation in the microvasculature. Circadian rhythm of gene expression was suppressed 80% in both macro- and microcirculations of obese mice. Circadian disruption with DD increased fasting serum glucose and HbA1c in obese but not lean mice. Endothelium-dependent dilation was attenuated in obese mice and in lean mice subjected to DD. Rhythmic expression of per1 and dbp was depressed in obesity. Expression of eNOS expression was suppressed and GTPCH1 lost rhythmic expression both in obesity and by constant darkness. CONCLUSION: These results suggest that obesity reduces circadian gene expression in concert with impaired endothelial function. The causal relationship remains to be determined.


Asunto(s)
Aorta , Relojes Circadianos , Regulación de la Expresión Génica , Microcirculación , Obesidad , Animales , Aorta/metabolismo , Aorta/fisiopatología , Masculino , Ratones , Obesidad/metabolismo , Obesidad/fisiopatología
9.
bioRxiv ; 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37131826

RESUMEN

Rationale: Obesity increases the risk of cardiovascular disease (CVD) through mechanisms that remain incompletely defined. Metabolic dysfunction, especially hyperglycemia, is thought to be a major contributor but how glucose impacts vascular function is unclear. Galectin-3 (GAL3) is a sugar binding lectin upregulated by hyperglycemia but its role as a causative mechanism of CVD remains poorly understood. Objective: To determine the role of GAL3 in regulating microvascular endothelial vasodilation in obesity. Methods and Results: GAL3 was markedly increased in the plasma of overweight and obese patients, as well as in the microvascular endothelium of diabetic patients. To investigate a role for GAL3 in CVD, mice deficient in GAL3 were bred with obese db/db mice to generate lean, lean GAL3 knockout (KO), obese, and obese GAL3 KO genotypes. GAL3 KO did not alter body mass, adiposity, glycemia or lipidemia, but normalized elevated markers of reactive oxygen species (TBARS) in plasma. Obese mice exhibited profound endothelial dysfunction and hypertension, both of which were rescued by GAL3 deletion. Isolated microvascular endothelial cells (EC) from obese mice had increased NOX1 expression, which we have previously shown to contribute to increased oxidative stress and endothelial dysfunction, and NOX1 levels were normalized in EC from obese mice lacking GAL3. EC-specific GAL3 knockout mice made obese using a novel AAV-approach recapitulated whole-body knockout studies, confirming that endothelial GAL3 drives obesity-induced NOX1 overexpression and endothelial dysfunction. Improved metabolism through increased muscle mass, enhanced insulin signaling, or metformin treatment, decreased microvascular GAL3 and NOX1. GAL3 increased NOX1 promoter activity and this was dependent on GAL3 oligomerization. Conclusions: Deletion of GAL3 normalizes microvascular endothelial function in obese db/db mice, likely through a NOX1-mediated mechanism. Pathological levels of GAL3 and in turn, NOX1, are amenable to improvements in metabolic status, presenting a potential therapeutic target to ameliorate pathological cardiovascular consequences of obesity.

10.
Arterioscler Thromb Vasc Biol ; 31(2): 392-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21051667

RESUMEN

OBJECTIVE: The Pin1 prolyl isomerase acts in concert with proline-directed protein kinases to regulate function of protein substrates through isomerization of peptide bonds that link phosphoserine or phosphothreonine to proline. We sought to determine whether Pin1 interacts with endothelial nitric oxide synthase (eNOS) in endothelial cells in a manner that depends on proline-directed phosphorylation of the eNOS enzyme and whether this interaction influences basal or agonist-stimulated eNOS activity. METHODS AND RESULTS: Inhibitors of the extracellular-regulated kinase (ERK) 1/2 MAP kinases inhibit proline-directed phosphorylation of eNOS at serine 116 (Ser116) in bovine aortic endothelial cells (BAECs). Moreover, eNOS and Pin1 can be coimmunoprecipitated from BAECs only when Ser116 is phosphorylated. In addition, phosphomimetic Ser116Asp eNOS, but not wild-type eNOS, can be coimmunoprecipitated with Pin1 coexpressed in COS-7 cells. Inhibition of Pin1 in BAECs by juglone or by dominant negative Pin1 increases basal and agonist-stimulated NO release from the cells, whereas overexpression of wild-type Pin1 in BAECs suppresses basal and agonist-stimulated NO production. Overexpression of wild-type Pin1 in intact aortae also reduces agonist-induced relaxation of aortic rings. CONCLUSIONS: Our results demonstrate a novel form of eNOS regulation in endothelial cells and blood vessels through Ser116 phosphorylation-dependent interaction of eNOS with Pin1.


Asunto(s)
Endotelio Vascular/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Células COS , Bovinos , Células Cultivadas , Chlorocebus aethiops , Endotelio Vascular/citología , Humanos , Modelos Animales , Peptidilprolil Isomerasa de Interacción con NIMA , Isomerasa de Peptidilprolil/genética , Fosforilación , Transducción de Señal/fisiología , Transfección
11.
Front Physiol ; 13: 887559, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35600313

RESUMEN

Obese individuals are at significantly elevated risk of developing cardiovascular disease (CVD). Additionally, obesity has been associated with disrupted circadian rhythm, manifesting in abnormal sleeping and feeding patterns. To date, the mechanisms linking obesity, circadian disruption, and CVD are incompletely understood, and insight into novel mechanistic pathways is desperately needed to improve therapeutic potential and decrease morbidity and mortality. The objective of this study was to investigate the roles of metabolic and circadian disruptions in obesity and assess their contributions in promoting vascular disease. Lean (db/+) and obese (db/db) mice were subjected to 12 weeks of constant darkness to differentiate diurnal and circadian rhythms, and were assessed for changes in metabolism, gene expression, and vascular function. Expression of endothelial nitric oxide synthase (eNOS), an essential enzyme for vascular health, was blunted in obesity and correlated with the oscillatory loss of the novel regulator cezanne (OTUD7B). Lean mice subjected to constant darkness displayed marked reduction in vasodilatory capacity, while endothelial dysfunction of obese mice was not further compounded by diurnal insult. Endothelial gene expression of essential circadian clock components was altered in obesity, but imperfectly phenocopied in lean mice housed in constant darkness, suggesting overlapping but separate mechanisms driving endothelial dysfunction in obesity and circadian disruption. Taken together, these data provide insight into the nature of endothelial circadian rhythm in obesity and suggest a distinct mechanism by which obesity causes a unique circadian defect in the vasculature.

12.
Circ Res ; 105(10): 1013-22, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19797171

RESUMEN

RATIONALE: Obesity is a risk factor for cardiovascular dysfunction, yet the underlying factors driving this impaired function remain poorly understood. Insulin resistance is a common pathology in obese patients and has been shown to impair vascular function. Whether insulin resistance or obesity, itself, is causal remains unclear. OBJECTIVE: The present study tested the hypothesis that insulin resistance is the underlying mediator for impaired NO-mediated dilation in obesity by genetic deletion of the insulin-desensitizing enzyme protein tyrosine phosphatase (PTP)1B in db/db mice. METHODS AND RESULTS: The db/db mouse is morbidly obese, insulin-resistant, and has tissue-specific elevation in PTP1B expression compared to lean controls. In db/db mice, PTP1B deletion improved glucose clearance, dyslipidemia, and insulin receptor signaling in muscle and fat. Hepatic insulin signaling in db/db mice was not improved by deletion of PTP1B, indicating specific amelioration of peripheral insulin resistance. Additionally, obese mice demonstrate an impaired endothelium dependent and independent vasodilation to acetylcholine and sodium nitroprusside, respectively. This impairment, which correlated with increased superoxide in the db/db mice, was corrected by superoxide scavenging. Increased superoxide production was associated with increased expression of NAD(P)H oxidase 1 and its molecular regulators, Noxo1 and Noxa1. CONCLUSIONS: Deletion of PTP1B improved both endothelium dependent and independent NO-mediated dilation and reduced superoxide generation in db/db mice. PTP1B deletion did not affect any vascular function in lean mice. Taken together, these data reveal a role for peripheral insulin resistance as the mediator of vascular dysfunction in obesity.


Asunto(s)
Endotelio Vascular/enzimología , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Resistencia a la Insulina , Leptina/metabolismo , Obesidad/enzimología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Acetilcolina/farmacología , Proteínas Adaptadoras Transductoras de Señales , Tejido Adiposo/enzimología , Animales , Dislipidemias/enzimología , Dislipidemias/genética , Glucosa/genética , Glucosa/metabolismo , Leptina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Obesos , Músculos/enzimología , NADH NADPH Oxidorreductasas/biosíntesis , NADH NADPH Oxidorreductasas/genética , NADPH Oxidasa 1 , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Nitroprusiato/farmacología , Obesidad/genética , Oxidación-Reducción/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteínas/genética , Proteínas/metabolismo , Superóxidos/metabolismo , Vasodilatación/efectos de los fármacos , Vasodilatación/genética , Vasodilatadores/farmacología
13.
Am J Physiol Heart Circ Physiol ; 299(1): H55-61, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20418477

RESUMEN

Obesity is a risk factor for stroke, but the determinants of increased stroke risk in obesity are unknown. We have previously reported that obese Zucker rats (OZRs) have a worse stroke outcome and display evidence of remodeling of the middle cerebral artery (MCA), in parallel with hypertension, compared with lean controls. This study tested the hypothesis that hypertension is an essential determinant of cerebral vascular remodeling and increased stroke damage in OZRs. Blood pressure was measured by telemetry in lean and obese rats with and without hydrochlorthiazide (HCT; 2 mg.kg(-1).day(-1)) from 8 to 15 wk of age. A separate group of rats was also chronically fed a low-sodium (LS) diet. Vessel structure was assessed in isolated, pressurized MCAs. Cerebral ischemia was induced for 60 min using an intralumenal suture technique, followed by 24 h of reperfusion. HCT treatment effectively prevented the increase in blood pressure in obese rats; however, the LS diet did not lower pressure. Importantly, infarct size was normalized by HCT after ischemia-reperfusion injury. Additionally, HCT improved the changes in MCA structure observed in untreated OZRs. There were no benefits of the LS diet on stroke injury or vessel structure. These results indicate that increased pressure is essential for driving the changes in infarct size in OZRs.


Asunto(s)
Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Hidroclorotiazida/farmacología , Hipertensión/prevención & control , Infarto de la Arteria Cerebral Media/prevención & control , Obesidad/tratamiento farmacológico , Animales , Monitoreo Ambulatorio de la Presión Arterial , Dieta Hiposódica , Modelos Animales de Enfermedad , Hipertensión/etiología , Hipertensión/patología , Hipertensión/fisiopatología , Infarto de la Arteria Cerebral Media/etiología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Obesidad/complicaciones , Obesidad/patología , Obesidad/fisiopatología , Ratas , Ratas Zucker , Factores de Riesgo , Índice de Severidad de la Enfermedad , Telemetría , Factores de Tiempo
14.
Microcirculation ; 16(5): 414-23, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19382001

RESUMEN

Obesity and diabetes are major risk factors for the development of vascular disease in the lower limbs. Previous studies have demonstrated reduced nitric oxide (NO)-mediated vasodilation, increased adrenergic constriction, and inward, atrophic remodeling in the limb circulation of obese Zucker rats, but the component of the "metabolic syndrome" driving these changes is unclear. Because insulin resistance precedes the state of frank diabetes, the current study hypothesized that insulin resistance independent of obesity induced by fructose feeding would impair microvascular function in the skeletal muscle circulation in lean Zucker rats (LZR). A 66% fructose diet impaired glucose tolerance and induced moderate insulin resistance with no changes in whole-body hemodynamics of anesthetized rats (FF-LZR), compared to control LZR. NO-mediated vasodilation of isolated gracilis arteries, assessed in vitro with acetylcholine and sodium nitroprusside, was reduced approximately 20% in FF-LZR vs. LZR. NO-independent cGMP-mediated vasodilation was unimpaired. Pretreatment of isolated vessels with the superoxide scavenger, tempol, improved responses to both vasodilators. Reactivity to adrenergic stimulation was unaltered in FF-LZR vs. LZR, although constriction to endothelin was increased. Structural and passive mechanical characteristics of isolated gracilis arteries were similar in both LZR and FF-LZR. Taken together, these findings indicate that moderate insulin resistance is sufficient to impair endothelial function in an oxidant-dependent manner in the rat hindlimb circulation. Other aspects of skeletal muscle vascular function documented in obese models, specifically adrenergic tone and inward remodeling, must reflect either severe insulin resistance or other aspects of obesity. The factors accounting for nonendothelial vasculopathies remain unknown.


Asunto(s)
Arterias/metabolismo , Endotelio Vascular/metabolismo , Fructosa/farmacología , Resistencia a la Insulina , Músculo Esquelético/irrigación sanguínea , Edulcorantes/farmacología , Acetilcolina/farmacología , Animales , GMP Cíclico , Dieta , Endotelio Vascular/patología , Miembro Posterior/irrigación sanguínea , Masculino , Músculo Esquelético/metabolismo , Óxido Nítrico/metabolismo , Nitroprusiato/farmacología , Obesidad/metabolismo , Oxidantes/metabolismo , Ratas , Ratas Zucker , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
15.
J Am Heart Assoc ; 7(16): e009358, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30369309

RESUMEN

Background Obesity compromises cardiometabolic function and is associated with hypertension and chronic kidney disease. Exercise ameliorates these conditions, even without weight loss. Although the mechanisms of exercise's benefits remain unclear, augmented lean body mass is a suspected mechanism. Myostatin is a potent negative regulator of skeletal muscle mass that is upregulated in obesity and downregulated with exercise. The current study tested the hypothesis that deletion of myostatin would increase muscle mass and reduce blood pressure and kidney injury in obesity. Methods and Results Myostatin knockout mice were crossed to db/db mice, and metabolic and cardiovascular functions were examined. Deletion of myostatin increased skeletal muscle mass by ≈50% to 60% without concomitant weight loss or reduction in fat mass. Increased blood pressure in obesity was prevented by the deletion of myostatin, but did not confer additional benefit against salt loading. Kidney injury was evident because of increased albuminuria, which was abolished in obese mice lacking myostatin. Glycosuria, total urine volume, and whole kidney NOX-4 levels were increased in obesity and prevented by myostatin deletion, arguing that increased muscle mass provides a multipronged defense against renal dysfunction in obese mice. Conclusions These experimental observations suggest that loss of muscle mass is a novel risk factor in obesity-derived cardiovascular dysfunction. Interventions that increase muscle mass, either through exercise or pharmacologically, may help limit cardiovascular disease in obese individuals.


Asunto(s)
Hipertensión/fisiopatología , Músculo Esquelético/fisiología , Obesidad/fisiopatología , Insuficiencia Renal Crónica/fisiopatología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Composición Corporal , Glucosuria Renal/fisiopatología , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Ratones Noqueados , Ratones Obesos , Miostatina/genética , NADPH Oxidasa 4/efectos de los fármacos , NADPH Oxidasa 4/metabolismo , Factores de Riesgo , Cloruro de Sodio Dietético/farmacología
16.
Physiol Rep ; 5(23)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29192067

RESUMEN

The objective of this study is to test the hypothesis that increased muscle mass has positive effects on cardiovascular function. Specifically, we tested the hypothesis that increases in lean body mass caused by deletion of myostatin improves cardiac performance and vascular function. Echocardiography was used to quantify left ventricular function at baseline and after acute administration of propranolol and isoproterenol to assess ß-adrenergic reactivity. Additionally, resistance vessels in several beds were removed, cannulated, pressurized to 60 mmHg and reactivity to vasoactive stimuli was assessed. Hemodynamics were measured using in vivo radiotelemetry. Myostatin deletion results in increased fractional shortening at baseline. Additionally, arterioles in the coronary and muscular microcirculations are more sensitive to endothelial-dependent dilation while nonmuscular beds or the aorta were unaffected. ß-adrenergic dilation was increased in both coronary and conduit arteries, suggesting a systemic effect of increased muscle mass on vascular function. Overall hemodynamics and physical characteristics (heart weight and size) remained unchanged. Myostatin deletion mimics in part the effects of exercise on cardiovascular function. It significantly increases lean muscle mass and results in muscle-specific increases in endothelium-dependent vasodilation. This suggests that increases in muscle mass may serve as a buffer against pathological states that specifically target cardiac function (heart failure), the ß-adrenergic system (age), and nitric oxide bio-availability (atherosclerosis). Taken together, pharmacological inhibition of the myostatin pathway could prove an excellent mechanism by which the benefits of exercise can be conferred in patients that are unable to exercise.


Asunto(s)
Vasos Coronarios/metabolismo , Corazón/fisiología , Microvasos/metabolismo , Miostatina/genética , Vasodilatación , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Presión Sanguínea , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiología , Eliminación de Gen , Frecuencia Cardíaca , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos ICR , Microvasos/efectos de los fármacos , Microvasos/fisiología , Músculo Esquelético/crecimiento & desarrollo , Propranolol/farmacología , Función Ventricular
17.
Physiol Rep ; 4(12)2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27335430

RESUMEN

Insulin resistance is a powerful predictor of cardiovascular disease; however, the mechanistic link remains unclear. This study aims to determine if early cardiovascular changes associated with short-term fructose feeding in the absence of obesity manifest as abnormal blood pressure control. Metabolic dysfunction was induced in Lean Zucker rats by short-term high-fructose feeding. Rats were implanted with telemetry devices for the measurement of mean arterial blood pressure (MAP) and subjected to air jet stress at 5 and 8 weeks after feeding. Additional animals were catheterized under anesthesia for the determination of MAP and blood flow responses in the hind limb and mesenteric vascular beds to intravenous injection of isoproterenol (0.001-0.5 µm), a ß-adrenergic agonist. Metabolic dysfunction in high-fructose rats was not accompanied by changes in 24-h MAP Yet, animals fed a high-fructose diet for 8 weeks exhibited a marked impairment in blood pressure recovery after air-jet stress. Dose-dependent decreases in MAP and peripheral blood flow in response to isoproterenol treatment were significantly attenuated in high-fructose rats. These data suggest that impaired blood pressure recovery to acute mental stress precedes the onset of hypertension in the early stages of insulin resistance. Further, blunted responses to isoproterenol implicate ß2-adrenergic sensitivity as a possible mechanism responsible for altered blood pressure control after short-term high-fructose feeding.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Carbohidratos de la Dieta/farmacología , Fructosa/farmacología , Estrés Psicológico/fisiopatología , Agonistas Adrenérgicos beta/farmacología , Animales , Carbohidratos de la Dieta/efectos adversos , Fructosa/administración & dosificación , Fructosa/efectos adversos , Miembro Posterior/irrigación sanguínea , Resistencia a la Insulina , Isoproterenol/farmacología , Masculino , Arterias Mesentéricas/fisiología , Ratas , Ratas Zucker , Flujo Sanguíneo Regional , Estrés Psicológico/metabolismo
18.
Placenta ; 36(10): 1204-6, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26282853

RESUMEN

Toll-like receptor (TLR)-regulated protein kinases and inflammatory cytokines were activated in fetal vascular smooth muscle cells (VSMC) treated with palmitate. Tumor necrosis factor (TNFα) and interleukin-6 (IL6) were increased and correlated with expression of TLRs in the labyrinth placentae of high fat (HF)-fed rats with increased plasma lipids and visceral adiposity. Thus, local induction of TLR signaling via saturated fatty acids (SFA) may in part contribute to placental inflammation in diet-induced maternal obesity.


Asunto(s)
Obesidad/metabolismo , Placenta/metabolismo , Complicaciones del Embarazo/metabolismo , Receptores Toll-Like/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Inflamación/etiología , Inflamación/metabolismo , Interleucina-6/metabolismo , Miocitos del Músculo Liso/metabolismo , Obesidad/etiología , Palmitatos/efectos adversos , Embarazo , Complicaciones del Embarazo/etiología , Distribución Aleatoria , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
19.
J Am Heart Assoc ; 3(3): e000854, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-24965025

RESUMEN

BACKGROUND: A sedentary lifestyle is an independent risk factor for cardiovascular disease and exercise has been shown to ameliorate this risk. Inactivity is associated with a loss of muscle mass, which is also reversed with isometric exercise training. The relationship between muscle mass and vascular function is poorly defined. The aims of the current study were to determine whether increasing muscle mass by genetic deletion of myostatin, a negative regulator of muscle growth, can influence vascular function in mesenteric arteries from obese db/db mice. METHODS AND RESULTS: Myostatin expression was elevated in skeletal muscle of obese mice and associated with reduced muscle mass (30% to 50%). Myostatin deletion increased muscle mass in lean (40% to 60%) and obese (80% to 115%) mice through increased muscle fiber size (P<0.05). Myostatin deletion decreased adipose tissue in lean mice, but not obese mice. Markers of insulin resistance and glucose tolerance were improved in obese myostatin knockout mice. Obese mice demonstrated an impaired endothelial vasodilation, compared to lean mice. This impairment was improved by superoxide dismutase mimic Tempol. Deletion of myostatin improved endothelial vasodilation in mesenteric arteries in obese, but not in lean, mice. This improvement was blunted by nitric oxide (NO) synthase inhibitor l-NG-nitroarginine methyl ester (l-NAME). Prostacyclin (PGI2)- and endothelium-derived hyperpolarizing factor (EDHF)-mediated vasodilation were preserved in obese mice and unaffected by myostatin deletion. Reactive oxygen species) was elevated in the mesenteric endothelium of obese mice and down-regulated by deletion of myostatin in obese mice. Impaired vasodilation in obese mice was improved by NADPH oxidase inhibitor (GKT136901). Treatment with sepiapterin, which increases levels of tetrahydrobiopterin, improved vasodilation in obese mice, an improvement blocked by l-NAME. CONCLUSIONS: Increasing muscle mass by genetic deletion of myostatin improves NO-, but not PGI2- or EDHF-mediated vasodilation in obese mice; this vasodilation improvement is mediated by down-regulation of superoxide.


Asunto(s)
Vasos Sanguíneos/fisiología , Ratones Obesos/fisiología , Aminoácidos , Animales , Cromo , Óxidos N-Cíclicos/farmacología , Resistencia a la Insulina/fisiología , Masculino , Ratones Noqueados , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/fisiología , Músculo Esquelético , Miostatina/genética , Miostatina/fisiología , NADPH Oxidasas/antagonistas & inhibidores , NG-Nitroarginina Metil Éster/farmacología , Ácidos Nicotínicos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Pterinas/farmacología , Pirazoles/farmacología , Piridonas/farmacología , Especies Reactivas de Oxígeno/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Marcadores de Spin , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
20.
Physiol Rep ; 1(6): e00146, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24400148

RESUMEN

Obesity is a major cause of hypertension, but links between the obese and hypertensive states remain incompletely understood. A major component of cardiovascular function in obese individuals is a state of sympathoactivation. A postulated mechanism of this sympathoactivation is the activation of specific classes of neurons commonly associated with metabolic control, which also affect sympathetic outflow to cardiovascular targets. One class of neurons is characterized by expression of melanocortin-4 receptors (MC4R) which are activated by metabolic signals such as leptin and insulin. In this study, we examined the effects of deletion of MC4R in a novel rat model. MC4R knockout (KO) rats are obese and profoundly insulin resistant without frank diabetes. Despite these conditions, MC4R KO rats are normotensive. Moderate bradycardia and significant increases in peripheral resistance were evident in MC4R KO rats. To determine if the dissociation between hypertension and obesity was associated with changes in vascular function, in vitro reactivity to vasoactive agents and in vivo reactivity to sympathetic blockade were examined. Vasodilator function was not affected by obesity in MC4R KO rats. Reactivity to phenylephrine was reduced, suggesting desensitization of adrenergic signaling. In response to ganglionic blockade with mecamylamine, blood pressure and hindlimb resistance fell more in MC4R KO rats, suggesting that sympathoactivation of the vascular was still evident, despite the absence of hypertension. These findings suggest that obesity causes sympathoactivation of the vasculature despite the absence of MC4R. Dissociation of obesity from hypertension in this model may reflect more renal mechanisms of blood pressure control.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA