Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 165(10)2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-39236000

RESUMO

BACKGROUND: While intermittent fasting leads to weight loss and improved glucose metabolism, food insecurity, the insufficient access to food for a healthy life, is associated with obesity and adverse cardiometabolic health, especially in women. We aimed to characterize the effects of intermittently restricted feeding on energy balance and glucose tolerance in female mice. METHODS: Female C57BL/6J mice were fed a high-fat, high-sucrose diet and intermittently food restricted to 60% of control littermates' ad libitum intake, starting at weaning and until week 19. Restricted mice were subsequently allowed ad libitum access to the same diet. Body composition and energy balance were measured at weeks 18.5, 19, 30, and 40. At week 42, mice underwent an intraperitoneal glucose tolerance test and plasma appetitive hormones measurements after nutrient gavage. RESULTS: During the food restriction phase, restricted mice accrued lower weight and fat mass than controls despite periodic ad libitum food access. Reintroduction of continuous ad libitum food caused increased food intake during the light phase and increased body mass in restricted mice. Minor differences in body composition-adjusted energy expenditure between groups were observed at week 40. At week 42, glucose tolerance was impaired in restricted mice compared to controls, and trends toward lower levels of postprandial anorexigenic hormones glucagon-like peptide-1 and pancreatic polypeptide were observed. CONCLUSION: Our findings suggest that repeated intermittent food restriction leads to changes in eating behavior that predispose to glucose intolerance when food is freely available. Future studies are needed to elucidate the specific mechanisms underlying these changes.


Assuntos
Composição Corporal , Dieta Hiperlipídica , Metabolismo Energético , Camundongos Endogâmicos C57BL , Animais , Feminino , Dieta Hiperlipídica/efeitos adversos , Camundongos , Teste de Tolerância a Glucose , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Sacarose Alimentar/administração & dosagem , Restrição Calórica , Obesidade/metabolismo , Obesidade/etiologia
2.
Hypertension ; 81(6): 1332-1344, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38629290

RESUMO

BACKGROUND: ANG (angiotensin II) elicits dipsogenic and pressor responses via activation of the canonical Gαq (G-protein component of the AT1R [angiotensin type 1 receptor])-mediated AT1R in the subfornical organ. Recently, we demonstrated that ARRB2 (ß-arrestin 2) global knockout mice exhibit a higher preference for salt and exacerbated pressor response to deoxycorticosterone acetate salt. However, whether ARRB2 within selective neuroanatomical nuclei alters physiological responses to ANG is unknown. Therefore, we hypothesized that ARRB2, specifically in the subfornical organ, counterbalances maladaptive dipsogenic and pressor responses to the canonical AT1R signaling. METHODS: Male and female Arrb2FLOX mice received intracerebroventricular injection of either adeno-associated virus (AAV)-Cre-GFP (green fluorescent protein) to induce brain-specific deletion of ARRB2 (Arrb2ICV-Cre). Arrb2FLOX mice receiving ICV-AAV-GFP were used as control (Arrb2ICV-Control). Infection with ICV-AAV-Cre primarily targeted the subfornical organ with few off targets. Fluid intake was evaluated using the 2-bottle choice paradigm with 1 bottle containing water and 1 containing 0.15 mol/L NaCl. RESULTS: Arrb2ICV-Cre mice exhibited a greater pressor response to acute ICV-ANG infusion. At baseline conditions, Arrb2ICV-Cre mice exhibited a significant increase in saline intake compared with controls, resulting in a saline preference. Furthermore, when mice were subjected to water-deprived or sodium-depleted conditions, which would naturally increase endogenous ANG levels, Arrb2ICV-Cre mice exhibited elevated saline intake. CONCLUSIONS: Overall, these data indicate that ARRB2 in selective cardiovascular nuclei in the brain, including the subfornical organ, counterbalances canonical AT1R responses to both exogenous and endogenous ANG. Stimulation of the AT1R/ARRB axis in the brain may represent a novel strategy to treat hypertension.


Assuntos
Pressão Sanguínea , Homeostase , Órgão Subfornical , beta-Arrestina 2 , Animais , Feminino , Masculino , Camundongos , Angiotensina II/farmacologia , beta-Arrestina 2/metabolismo , beta-Arrestina 2/genética , Pressão Sanguínea/fisiologia , Pressão Sanguínea/genética , Encéfalo/metabolismo , Homeostase/fisiologia , Camundongos Knockout , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Órgão Subfornical/metabolismo
3.
Cell Rep ; 33(4): 108270, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-33113371

RESUMO

The exact mechanisms underlying the metabolic effects of bariatric surgery remain unclear. Here, we demonstrate, using a combination of direct and indirect calorimetry, an increase in total resting metabolic rate (RMR) and specifically anaerobic RMR after Roux-en-Y gastric bypass (RYGB), but not sleeve gastrectomy (SG). We also show an RYGB-specific increase in splanchnic sympathetic nerve activity and "browning" of visceral mesenteric fat. Consequently, selective splanchnic denervation abolishes all beneficial metabolic outcomes of gastric bypass that involve changes in the endocannabinoid signaling within the small intestine. Furthermore, we demonstrate that administration of rimonabant, an endocannabinoid receptor-1 (CB1) inverse agonist, to obese mice mimics RYGB-specific effects on energy balance and splanchnic nerve activity. On the other hand, arachidonoylethanolamide (AEA), a CB1 agonist, attenuates the weight loss and metabolic signature of this procedure. These findings identify CB1 as a key player in energy regulation post-RYGB via a pathway involving the sympathetic nervous system.


Assuntos
Endocanabinoides/uso terapêutico , Derivação Gástrica/métodos , Sistema Nervoso Simpático/fisiologia , Animais , Endocanabinoides/farmacologia , Feminino , Humanos , Masculino , Camundongos
4.
Diabetes ; 68(8): 1591-1603, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31127052

RESUMO

The BBSome, a complex of eight Bardet-Biedl syndrome (BBS) proteins involved in cilia function, has emerged as an important regulator of energy balance, but the underlying cellular and molecular mechanisms are not fully understood. Here, we show that the control of energy homeostasis by the anorexigenic proopiomelanocortin (POMC) neurons and orexigenic agouti-related peptide (AgRP) neurons require intact BBSome. Targeted disruption of the BBSome by Bbs1 gene deletion in POMC or AgRP neurons increases body weight and adiposity. We demonstrate that obesity in mice lacking the Bbs1 gene in POMC neurons is associated with hyperphagia. Mechanistically, we present evidence implicating the BBSome in the trafficking of G protein-coupled neuropeptide Y Y2 receptor (NPY2R) and serotonin 5-hydroxytryptamine (HT)2C receptor (5-HT2CR) to cilia and plasma membrane, respectively. Consistent with this, loss of the BBSome reduced cell surface expression of the 5-HT2CR, interfered with serotonin-evoked increase in intracellular calcium and membrane potential, and blunted the anorectic and weight-reducing responses evoked by the 5-HT2cR agonist, lorcaserin. Finally, we show that disruption of the BBSome causes the 5-HT2CR to be stalled in the late endosome. Our results demonstrate the significance of the hypothalamic BBSome for the control of energy balance through regulation of trafficking of important metabolic receptors.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Peso Corporal/fisiologia , Hiperfagia/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Adiposidade/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Hiperfagia/genética , Hipotálamo/metabolismo , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Obesidade/genética , Transporte Proteico/fisiologia , Receptores de Neuropeptídeo Y/metabolismo , Receptores 5-HT2 de Serotonina/metabolismo
5.
Obesity (Silver Spring) ; 24(10): 2140-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27515998

RESUMO

OBJECTIVE: Diet-induced obesity has been shown to alter immune function in mice, but distinguishing the effects of obesity from changes in diet composition is complicated. It was hypothesized that immunological differences would exist between diet-induced obese (DIO) and obese-resistant (OB-Res) mice fed the same high-fat diet (HFD). METHODS: BALB/c mice were fed either standard chow or HFD to generate lean or DIO and OB-Res mice, respectively. Resulting mice were analyzed for serum immunologic and metabolic profiles and cellular immune parameters. RESULTS: BALB/c mice on HFD were categorized as DIO or OB-Res, based on body weight versus lean controls. DIO mice were physiologically distinct from OB-Res mice, whose serum insulin, leptin, gastric inhibitory polypeptide, and eotaxin concentrations remained similar to lean controls. DIO mice had increased macrophage(+) crown-like structures in white adipose tissue, although macrophage percentages were unchanged from OB-Res and lean mice. DIO mice also had decreased splenic CD4(+) T cells, elevated serum GM-CSF, and increased splenic CD11c(+) dendritic cells, but impaired dendritic cell stimulatory capacity (P < 0.05 vs. lean controls). These parameters were unaltered in OB-Res mice versus lean controls. CONCLUSIONS: Diet-induced obesity results in alterations in immune and metabolic profiles that are distinct from effects caused by HFD alone.


Assuntos
Dieta Hiperlipídica , Obesidade/metabolismo , Animais , Peso Corporal/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Quimiocina CCL11/sangue , Feminino , Insulina/sangue , Leptina/sangue , Masculino , Metaboloma , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/imunologia , Baço/metabolismo
6.
PLoS Genet ; 12(2): e1005890, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26926121

RESUMO

Bardet-Biedl syndrome (BBS) is a highly pleiotropic autosomal recessive disorder associated with a wide range of phenotypes including obesity. However, the underlying mechanism remains unclear. Here, we show that neuronal BBSome is a critical determinant of energy balance through its role in the regulation of the trafficking of the long signaling form of the leptin receptor (LRb). Targeted disruption of the BBSome by deleting the Bbs1 gene from the nervous system causes obesity in mice, and this phenotype is reproduced by ablation of the Bbs1 gene selectively in the LRb-expressing cells, but not from adipocytes. Obesity developed as a consequence of both increased food intake and decreased energy expenditure in mice lacking the Bbs1 gene in LRb-expressing cells. Strikingly, the well-known role of BBS proteins in the regulation of ciliary formation and function is unlikely to account for the obesogenic effect of BBS1 loss as disruption of the intraflagellar transport (IFT) machinery required for ciliogenesis by deleting the Ift88 gene in LRb-expressing cells caused a marginal increase in body weight and adiposity. Instead, we demonstrate that silencing BBS proteins, but not IFT88, impair the trafficking of the LRb to the plasma membrane leading to central leptin resistance in a manner independent of obesity. Our data also demonstrate that postnatal deletion of the Bbs1 gene in the mediobasal hypothalamus can cause obesity in mice, arguing against an early neurodevelopmental origin of obesity in BBS. Our results depict a novel mechanism underlying energy imbalance and obesity in BBS with potential implications in common forms of human obesity.


Assuntos
Síndrome de Bardet-Biedl/metabolismo , Membrana Celular/metabolismo , Receptores para Leptina/metabolismo , Animais , Síndrome de Bardet-Biedl/genética , Membrana Celular/genética , Metabolismo Energético/fisiologia , Feminino , Hipotálamo/fisiologia , Camundongos Mutantes , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Complexos Multiproteicos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Transporte Proteico , Receptores para Leptina/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
7.
Hypertension ; 65(6): 1341-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25895586

RESUMO

Endoplasmic reticulum stress has become an important mechanism in hypertension. We examined the role of endoplasmic reticulum stress in mediating the increased saline-intake and hypertensive effects in response to deoxycorticosterone acetate (DOCA)-salt. Intracerebroventricular delivery of the endoplasmic reticulum stress-reducing chemical chaperone tauroursodeoxycholic acid did not affect the magnitude of hypertension, but markedly decreased saline-intake in response to DOCA-salt. Increased saline-intake returned after tauroursodeoxycholic acid was terminated. Decreased saline-intake was also observed after intracerebroventricular infusion of 4-phenylbutyrate, another chemical chaperone. Immunoreactivity to CCAAT homologous binding protein, a marker of irremediable endoplasmic reticulum stress, was increased in the subfornical organ and supraoptic nucleus of DOCA-salt mice, but the signal was absent in control and CCAAT homologous binding protein-deficient mice. Electron microscopy revealed abnormalities in endoplasmic reticulum structure (decrease in membrane length, swollen membranes, and decreased ribosome numbers) in the subfornical organ consistent with endoplasmic reticulum stress. Subfornical organ-targeted adenoviral delivery of GRP78, a resident endoplasmic reticulum chaperone, decreased DOCA-salt-induced saline-intake. The increase in saline-intake in response to DOCA-salt was blunted in CCAAT homologous binding protein-deficient mice, but these mice exhibited a normal hypertensive response. We conclude that (1) brain endoplasmic reticulum stress mediates the saline-intake, but not blood pressure response to DOCA-salt, (2) DOCA-salt causes endoplasmic reticulum stress in the subfornical organ, which when attenuated by GRP78 blunts saline-intake, and (3) CCAAT homologous binding protein may play a functional role in DOCA-salt-induced saline-intake. The results suggest a mechanistic distinction between the importance of endoplasmic reticulum stress in mediating effects of DOCA-salt on saline-intake and blood pressure.


Assuntos
Encéfalo/metabolismo , Acetato de Desoxicorticosterona/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hipertensão/fisiopatologia , Cloreto de Sódio/farmacologia , Análise de Variância , Animais , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/fisiologia , Infusões Intraventriculares , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade , Cloreto de Sódio/metabolismo , Estatísticas não Paramétricas , Órgão Subfornical/efeitos dos fármacos , Órgão Subfornical/fisiopatologia
8.
Hypertension ; 64(4): 852-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25001273

RESUMO

Preeclampsia, a cardiovascular disorder of late pregnancy, is characterized as a low-renin hypertensive state relative to normotensive pregnancy. Because other nonpregnant low-renin hypertensive disorders often exhibit and are occasionally dependent on elevated arginine vasopressin (AVP) secretion, we hypothesized a possible use for plasma AVP measurements in the prediction of preeclampsia. Copeptin is an inert prosegment of AVP that is secreted in a 1:1 molar ratio and exhibits a substantially longer biological half-life compared with AVP, rendering it a clinically useful biomarker of AVP secretion. Copeptin was measured throughout pregnancy in maternal plasma from preeclamptic and control women. Maternal plasma copeptin was significantly higher throughout preeclamptic pregnancies versus control pregnancies. While controlling for clinically significant confounders (age, body mass index, chronic essential hypertension, twin gestation, diabetes mellitus, and history of preeclampsia) using multivariate regression, the association of higher copeptin concentration and the development of preeclampsia remained significant. Receiver operating characteristic analyses reveal that as early as the sixth week of gestation, elevated maternal plasma copeptin concentration is a highly significant predictor of preeclampsia throughout pregnancy. Finally, chronic infusion of AVP during pregnancy (24 ng per hour) is sufficient to phenocopy preeclampsia in C57BL/6J mice, causing pregnancy-specific hypertension, renal glomerular endotheliosis, proteinuria, and intrauterine growth restriction. These data implicate AVP release as a novel predictive biomarker for preeclampsia very early in pregnancy, identify chronic AVP infusion as a novel and clinically relevant model of preeclampsia in mice, and are consistent with a potential causative role for AVP in preeclampsia in humans.


Assuntos
Biomarcadores/sangue , Diagnóstico Precoce , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/diagnóstico , Vasopressinas/sangue , Adulto , Animais , Arginina Vasopressina/administração & dosagem , Arginina Vasopressina/sangue , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Feminino , Glicopeptídeos/sangue , Humanos , Hipertensão/sangue , Hipertensão/fisiopatologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pré-Eclâmpsia/fisiopatologia , Gravidez , Proteinúria/sangue , Curva ROC , Fatores de Tempo
9.
Am J Physiol Regul Integr Comp Physiol ; 307(4): R376-86, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24965793

RESUMO

Increased activity of the renin-angiotensin system within the brain elevates fluid intake, blood pressure, and resting metabolic rate. Renin and angiotensinogen are coexpressed within the same cells of the subfornical organ, and the production and action of ANG II through the ANG II type 1 receptor in the subfornical organ (SFO) are necessary for fluid intake due to increased activity of the brain renin-angiotensin system. We generated an inducible model of ANG II production by breeding transgenic mice expressing human renin in neurons controlled by the synapsin promoter with transgenic mice containing a Cre-recombinase-inducible human angiotensinogen construct. Adenoviral delivery of Cre-recombinase causes SFO-selective induction of human angiotensinogen expression. Selective production of ANG II in the SFO results in increased water intake but did not change blood pressure or resting metabolic rate. The increase in water intake was ANG II type 1 receptor-dependent. When given a choice between water and 0.15 M NaCl, these mice increased total fluid and sodium, but not water, because of an increased preference for NaCl. When provided a choice between water and 0.3 M NaCl, the mice exhibited increased fluid, water, and sodium intake, but no change in preference for NaCl. The increase in fluid intake was blocked by an inhibitor of PKC, but not ERK, and was correlated with increased phosphorylated cyclic AMP response element binding protein in the subfornical organ. Thus, increased production and action of ANG II specifically in the subfornical organ are sufficient on their own to mediate an increase in drinking through PKC.


Assuntos
Angiotensinogênio/metabolismo , Ingestão de Líquidos , Sistema Renina-Angiotensina , Renina/metabolismo , Órgão Subfornical/enzimologia , Angiotensinogênio/genética , Animais , Comportamento Animal , Pressão Sanguínea , Proteína de Ligação a CREB/metabolismo , Ingestão de Líquidos/efeitos dos fármacos , Comportamento de Ingestão de Líquido , Metabolismo Energético , Feminino , Humanos , Integrases/genética , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Regiões Promotoras Genéticas , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Renina/genética , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/genética , Transdução de Sinais , Cloreto de Sódio/administração & dosagem , Órgão Subfornical/efeitos dos fármacos , Sinapsinas/genética , Fatores de Tempo
10.
Hypertension ; 64(1): 141-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24777977

RESUMO

Angiotensin-II production in the subfornical organ acting through angiotensin-II type-1 receptors is necessary for polydipsia, resulting from elevated renin-angiotensin system activity. Protein kinase C and mitogen-activated protein kinase pathways have been shown to mediate effects of angiotensin-II in the brain. We investigated mechanisms that mediate brain angiotensin-II-induced polydipsia. We used double-transgenic sRA mice, consisting of human renin controlled by the neuron-specific synapsin promoter crossed with human angiotensinogen controlled by its endogenous promoter, which results in brain-specific overexpression of angiotensin-II, particularly in the subfornical organ. We also used the deoxycorticosterone acetate-salt model of hypertension, which exhibits polydipsia. Inhibition of protein kinase C, but not extracellular signal-regulated kinases, protein kinase A, or vasopressin V1A and V2 receptors, corrected the elevated water intake of sRA mice. Using an isoform selective inhibitor and an adenovirus expressing dominant negative protein kinase C-α revealed that protein kinase C-α in the subfornical organ was necessary to mediate elevated fluid and sodium intake in sRA mice. Inhibition of protein kinase C activity also attenuated polydipsia in the deoxycorticosterone acetate-salt model. We provide evidence that inducing protein kinase C activity centrally is sufficient to induce water intake in water-replete wild-type mice, and that cell surface localization of protein kinase C-α can be induced in cultured cells from the subfornical organ. These experimental findings demonstrate a role for central protein kinase C activity in fluid balance, and further mechanistically demonstrate the importance of protein kinase C-α signaling in the subfornical organ in fluid intake stimulated by angiotensin-II in the brain.


Assuntos
Encéfalo/metabolismo , Ingestão de Líquidos/fisiologia , Proteína Quinase C-alfa/metabolismo , Sistema Renina-Angiotensina/fisiologia , Órgão Subfornical/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Feminino , Hipertensão/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteína Quinase C-alfa/antagonistas & inibidores , Sistema Renina-Angiotensina/efeitos dos fármacos , Órgão Subfornical/efeitos dos fármacos
11.
Hypertension ; 63(2): 316-23, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24246383

RESUMO

The (pro)renin receptor (PRR), which binds both renin and prorenin, is a newly discovered component of the renin-angiotensin system that is highly expressed in the central nervous system. The significance of brain PRRs in mediating local angiotensin II formation and regulating blood pressure remains unclear. The current study was performed to test the hypothesis that PRR-mediated, nonproteolytic activation of prorenin is the main source of angiotensin II in the brain. Thus, PRR knockout in the brain is expected to prevent angiotensin II formation and development of deoxycorticosterone acetate-salt-induced hypertension. A neuron-specific PRR (ATP6AP2) knockout mouse model was generated using the Cre-LoxP system. Physiological parameters were recorded by telemetry. PRR expression, detected by immunostaining and reverse transcription-polymerase chain reaction, was significantly decreased in the brains of knockout mice compared with wild-type mice. Intracerebroventricular infusion of mouse prorenin increased blood pressure and angiotensin II formation in wild-type mice. This hypertensive response was abolished in PRR-knockout mice in association with a reduction in angiotensin II levels. Deoxycorticosterone acetate-salt increased PRR expression and angiotensin II formation in the brains of wild-type mice, an effect that was attenuated in PRR-knockout mice. PRR knockout in neurons prevented the development of deoxycorticosterone acetate-salt-induced hypertension as well as activation of cardiac and vasomotor sympathetic tone. In conclusion, nonproteolytic activation of prorenin through binding to the PRR mediates angiotensin II formation in the brain. Neuron-specific PRR knockout prevents the development of deoxycorticosterone acetate-salt-induced hypertension, possibly through diminished angiotensin II formation.


Assuntos
Hipertensão Renal/genética , Neurônios/fisiologia , ATPases Translocadoras de Prótons/genética , Receptores de Superfície Celular/genética , Cloreto de Sódio na Dieta/farmacologia , Angiotensina II/metabolismo , Animais , Pressão Sanguínea/fisiologia , Acetato de Desoxicorticosterona/farmacologia , Feminino , Hipertensão Renal/induzido quimicamente , Hipertensão Renal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Mineralocorticoides/farmacologia , Neurônios/citologia , ATPases Translocadoras de Prótons/metabolismo , Receptores de Superfície Celular/metabolismo , Receptor de Pró-Renina
12.
Hypertension ; 61(3): 716-22, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23266541

RESUMO

Although elevated renin-angiotensin system activity and angiotensinergic signaling within the brain are required for hypertension, polydipsia, and increased metabolic rate induced by deoxycorticosterone acetate (DOCA)-salt, the contribution of specific receptor subtypes and brain nuclei mediating these responses remains poorly defined. We hypothesized that angiotensin type 1a receptors (AT(1a)R) within the subfornical organ (SFO) mediate these responses. Transgenic mice carrying a conditional allele of the endogenous AT(1a)R (AT(1a)R(flox)) were administered an adenovirus encoding Cre-recombinase and enhanced green fluorescent protein (eGFP) or adenovirus encoding eGFP alone into the lateral cerebral ventricle. Adenovirus encoding Cre-recombinase reduced AT(1a)R mRNA and induced recombination in AT(1a)R(flox) genomic DNA specifically in the SFO, without significant effect in the paraventricular or arcuate nuclei, and also induced SFO-specific recombination in ROSA(TdTomato) reporter mice. The effect of SFO-targeted ablation of endogenous AT(1a)R was evaluated in AT(1a)R(flox) mice at 3 time points: (1) baseline, (2) 1 week after virus injection but before DOCA-salt, and (3) after 3 weeks of DOCA-salt. DOCA-salt-treated mice with deletion of AT(1a)R in SFO exhibited a blunted increase in arterial pressure. Increased sympathetic cardiac modulation and urine copeptin, a marker of vasopressin release, were both significantly reduced in DOCA-salt mice when AT(1a)R was deleted in the SFO. Additionally, deletion of AT(1a)R in the SFO significantly attenuated the polydipsia, polyuria, and sodium intake in response to DOCA-salt. Together, these data highlight the contribution of AT(1a)R in the SFO to arterial pressure regulation potentially through changes on sympathetic cardiac modulation, vasopressin release, and hydromineral balance in the DOCA-salt model of hypertension.


Assuntos
Desoxicorticosterona/efeitos adversos , Hipertensão/induzido quimicamente , Mineralocorticoides/efeitos adversos , Receptor Tipo 1 de Angiotensina/fisiologia , Órgão Subfornical/efeitos dos fármacos , Órgão Subfornical/fisiopatologia , Animais , Pressão Arterial/efeitos dos fármacos , Biomarcadores/urina , Glicopeptídeos/urina , Coração/efeitos dos fármacos , Coração/inervação , Masculino , Camundongos , Camundongos Transgênicos , Polidipsia/induzido quimicamente , Poliúria/induzido quimicamente , Receptor Tipo 1 de Angiotensina/genética , Recombinação Genética , Sódio/metabolismo , Sistema Nervoso Simpático/efeitos dos fármacos
13.
Hypertension ; 51(3): 712-8, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18250366

RESUMO

Angiotensin converting enzyme 2 (ACE2) has been linked to cardiac dysfunction and hypertension-induced cardiac pathophysiology. In this study, we used a gene overexpression approach to investigate the role of ACE2 in cardiac function and remodeling after myocardial infarction. Rats received an intracardiac injection of 4.5x10(8) lentivirus containing ACE2 cDNA, followed by permanent coronary artery ligation (CAL) of the left anterior descending artery. At 24 hours and 6 weeks after surgery, cardiac functions, viability, and pathophysiology were assessed by MRI) and by histological analysis. At 24 hours post-CAL, left ventricular (LV) anterior wall motion was stunted to the same extent in control CAL and lenti-ACE2-treated CAL rats. However lenti-ACE2-treated CAL rats showed a 60% reduction in delayed contrast-enhanced LV volume after gadodiamide injection, indicating early ischemic protection of myocardium by ACE2. At 6 weeks after CAL, lenti-ACE2 rats demonstrated a complete rescue of cardiac output, a 41% rescue of ejection fraction, a 44% rescue in contractility, a 37% rescue in motion, and a 53% rescue in LV anterior (infracted) wall thinning compared with control CAL rats. No changes were observed in the LV posterior (noninfarcted) wall other than an 81% rescue in motion produced by ACE2 in CAL rats. Finally, infarct size measured by 2,3,5-triphenyl-tetrazolium chloride staining was not significantly different between the ligated groups. These observations demonstrate that cardiac overexpression of ACE2 exerts protective influence on the heart during myocardial infarction by preserving cardiac functions, LV wall motion and contractility, and by attenuating LV wall thinning.


Assuntos
Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/prevenção & controle , Miocárdio/enzimologia , Peptidil Dipeptidase A/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , DNA Complementar/genética , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica/fisiologia , Terapia Genética/métodos , Coração/fisiopatologia , Lentivirus/genética , Imageamento por Ressonância Magnética , Masculino , Contração Miocárdica/efeitos dos fármacos , Contração Miocárdica/fisiologia , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/patologia , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Volume Sistólico/efeitos dos fármacos , Volume Sistólico/fisiologia , Transdução Genética , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/fisiologia
14.
Am J Physiol Heart Circ Physiol ; 292(2): H736-42, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17098828

RESUMO

Cardiac remodeling, which typically results from chronic hypertension or following an acute myocardial infarction, is a major risk factor for the development of heart failure and, ultimately, death. The renin-angiotensin system (RAS) has previously been established to play an important role in the progression of cardiac remodeling, and inhibition of a hyperactive RAS provides protection from cardiac remodeling and subsequent heart failure. Our previous studies have demonstrated that overexpression of angiotensin-converting enzyme 2 (ACE2) prevents cardiac remodeling and hypertrophy during chronic infusion of angiotensin II (ANG II). This, coupled with the knowledge that ACE2 is a key enzyme in the formation of ANG-(1-7), led us to hypothesize that chronic infusion of ANG-(1-7) would prevent cardiac remodeling induced by chronic infusion of ANG II. Infusion of ANG II into adult Sprague-Dawley rats resulted in significantly increased blood pressure, myocyte hypertrophy, and midmyocardial interstitial fibrosis. Coinfusion of ANG-(1-7) resulted in significant attenuations of myocyte hypertrophy and interstitial fibrosis, without significant effects on blood pressure. In a subgroup of animals also administered [d-Ala(7)]-ANG-(1-7) (A779), an antagonist to the reported receptor for ANG-(1-7), there was a tendency to attenuate the antiremodeling effects of ANG-(1-7). Chronic infusion of ANG II, with or without coinfusion of ANG-(1-7), had no effect on ANG II type 1 or type 2 receptor binding in cardiac tissue. Together, these findings indicate an antiremodeling role for ANG-(1-7) in cardiac tissue, which is not mediated through modulation of blood pressure or altered cardiac angiotensin receptor populations and may be at least partially mediated through an ANG-(1-7) receptor.


Assuntos
Angiotensina I/metabolismo , Cardiomegalia/metabolismo , Miocárdio/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Remodelação Ventricular , Análise de Variância , Angiotensina I/farmacologia , Angiotensina II/análogos & derivados , Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/etiologia , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Cardiomegalia/prevenção & controle , Modelos Animais de Doenças , Fibrose , Coração/efeitos dos fármacos , Hipertensão/induzido quimicamente , Hipertensão/complicações , Hipertensão/fisiopatologia , Masculino , Miocárdio/patologia , Fragmentos de Peptídeos/farmacologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Fatores de Tempo , Fator de Crescimento Transformador beta/sangue , Remodelação Ventricular/efeitos dos fármacos
15.
Am J Physiol Heart Circ Physiol ; 292(2): H727-35, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17085538

RESUMO

Our previous studies demonstrated that peripheral overexpression of angiotensin II (ANG II) type 2 receptors (AT(2)R) prevents hypertension-induced cardiac hypertrophy and remodeling without altering high blood pressure. This, coupled with the observations that AT(2)R play a role in the antihypertensive actions of ANG II type 1 receptor (AT(1)R) blockers (ARBs), led us to propose that peripheral overexpression of AT(2)R would improve the antihypertensive action of losartan (Los) in Sprague-Dawley (SD) rats made hypertensive via chronic infusion of ANG II. Here we utilized adenoviral vector-mediated AT(2)R gene transfer to test this hypothesis. A single intracardiac injection of adenoviral vector containing genomic AT(2)R (G-AT(2)R) DNA and enhanced green fluorescent protein (EGFP) gene controlled by cytomegalovirus (CMV) promoters (Ad-G-AT(2)R-EGFP; 5 x 10(9) infectious units) into adult SD rats produced robust AT(2)R overexpression in cardiovascular tissues (kidney, lung, heart, aorta, mesenteric artery, and renal artery) that persisted for 3-5 days postinjection. By 7 days post viral injection, the overexpressed AT(2)R are reduced toward basal values in certain tissues (lung, kidney, and heart) and are undetectable in others (kidney and blood vessels). In two separate protocols, we demonstrated that the hypotensive effect of Los (0.125, 0.5, and 1.0 mg/kg iv) was significantly greater in the AT(2)R-overexpressing animals (-40.7 +/- 4.3, -41.8 +/- 4.8, and -48.1 +/- 2.6 mmHg, respectively) compared with control vector (Ad-CMV-EGFP)-treated rats (-12.4 +/- 2.2, -20.2 +/- 3.4, and -27.3 +/- 3.4 mmHg, respectively). These results provide support for a depressor role of AT(2)R and the proposal that combined AT(2)R agonist and ARB treatment may be an improved therapeutic strategy for controlling hypertension.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Losartan/farmacologia , Receptor Tipo 2 de Angiotensina/efeitos dos fármacos , Adenoviridae/genética , Angiotensina II/metabolismo , Animais , Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Vetores Genéticos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Imidazóis/farmacologia , Masculino , Miocárdio/citologia , Miocárdio/metabolismo , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor Tipo 2 de Angiotensina/biossíntese , Receptor Tipo 2 de Angiotensina/genética , Fatores de Tempo , Transdução Genética , Regulação para Cima
16.
Am J Physiol Heart Circ Physiol ; 290(6): H2417-23, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16415071

RESUMO

Cardiac remodeling is a hallmark hypertension-induced pathophysiology. In the current study, the role of the angiotensin-(1-7) fragment in modulating cardiac remodeling was examined. Sprague-Dawley rats underwent uninephrectomy surgery and were implanted with a deoxycorticosterone acetate (DOCA) pellet. DOCA animals had their drinking water replaced with 0.9% saline solution. A subgroup of DOCA-salt animals was implanted with osmotic minipumps, which delivered angiotensin-(1-7) chronically (100 ng.kg(-1).min(-1)). Control animals underwent sham surgery and were maintained on normal drinking water. Blood pressure was measured weekly with the use of the tail-cuff method, and after 4 wk of treatment, blood pressure responses to graded doses of angiotensin II were determined by direct carotid artery cannulation. Ventricle size was measured, and cross sections of the heart ventricles were paraffin embedded and stained using Masson's Trichrome to measure interstitial and perivascular collagen deposition and myocyte diameter. DOCA-salt treatment caused significant increases in blood pressure, cardiac hypertrophy, and myocardial and perivascular fibrosis. Angiotensin-(1-7) infusion prevented the collagen deposition effects without any effect on blood pressure or cardiac hypertrophy. These results indicate that angiotensin-(1-7) selectively prevents cardiac fibrosis independent of blood pressure or cardiac hypertrophy in the DOCA-salt model of hypertension.


Assuntos
Angiotensina I/uso terapêutico , Desoxicorticosterona , Cardiopatias/prevenção & controle , Hipertensão/induzido quimicamente , Hipertensão/complicações , Fragmentos de Peptídeos/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Cardiomegalia/induzido quimicamente , Cardiomegalia/prevenção & controle , Colágeno/metabolismo , Vasos Coronários/patologia , Relação Dose-Resposta a Droga , Fibrose , Cardiopatias/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Remodelação Ventricular/efeitos dos fármacos
17.
Regul Pept ; 133(1-3): 62-7, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16219374

RESUMO

Estrogen's suggested cardio-protective effects have come into question following the results of recent clinical trials. Two major components of the renin-angiotensin system (RAS) that are modulated by estrogen are angiotensin converting enzyme, and the angiotensin II type 1 receptor. Further research has revealed several new components of the RAS, including angiotensin converting enzyme 2, its peptide product angiotensin 1-7 (Ang 1-7), and that peptide's receptor, Mas. These components appear to oppose the classical effects of the RAS, and may act to buffer the RAS in vivo. Recent work has shown that during pregnancy, when estradiol levels are elevated, renal and urinary Ang 1-7 are greatly increased. This study examined the effects of estradiol on the efficacy of Ang 1-7 in the rat aorta. Female Sprague-Dawley rats were ovariectomized and a subgroup was chronically treated with subcutaneous pellets of estradiol for 3 weeks. Thoracic aortas were harvested for assessment of in vitro vascular reactivity to Ang 1-7. The results demonstrated that increased estradiol exposure attenuated the relaxation response to Ang 1-7 in a dose-dependent manner. These findings are in contrast to recent work showing potentiated responses to Ang 1-7 in mesenteric arteries from estrogen-manipulated rats, and may suggest a regional specificity in estradiol-mediated changes in the RAS.


Assuntos
Angiotensina I/metabolismo , Anti-Hipertensivos/metabolismo , Aorta/efeitos dos fármacos , Estradiol/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Angiotensina I/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Aorta/metabolismo , Estradiol/sangue , Estradiol/metabolismo , Feminino , Músculo Liso Vascular/metabolismo , Fragmentos de Peptídeos/farmacologia , Gravidez , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA