Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
J Cell Mol Med ; 24(13): 7393-7404, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32420690

RESUMO

Clinical and experimental studies show that angiotensin II (AngII) promotes vascular pathology via activation of AngII type 1 receptors (AT1Rs). We recently reported that NP-6A4, a selective peptide agonist for AngII type 2 receptor (AT2R), exerts protective effects on human vascular cells subjected to serum starvation or doxorubicin exposure. In this study, we investigated whether NP-6A4-induced AT2R activation could mitigate AngII-induced abdominal aortic aneurism (AAA) using AngII-treated Apoe-/- mice. Male Apoe-/- mice were infused with AngII (1 µg/kg/min) by implanting osmotic pumps subcutaneously for 28 days. A subset of mice was pre-treated subcutaneously with NP-6A4 (2.5 mg/kg/day) or vehicle for 14 days prior to AngII, and treatments were continued for 28 days. NP-6A4 significantly reduced aortic stiffness of the abdominal aorta induced by AngII as determined by ultrasound functional analyses and histochemical analyses. NP-6A4 also increased nitric oxide bioavailability in aortic tissues and suppressed AngII-induced increases in monocyte chemotactic protein-1, osteopontin and proteolytic activity of the aorta. However, NP-6A4 did not affect maximal intraluminal aortic diameter or AAA incidences significantly. These data suggest that the effects of AT2R agonist on vascular pathologies are selective, affecting the aortic stiffness and proteolytic activity without affecting the size of AAA.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/fisiopatologia , Proteólise , Receptor Tipo 2 de Angiotensina/agonistas , Rigidez Vascular , Angiotensina II , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos Knockout , Osteopontina/metabolismo , Fenótipo , Receptor Tipo 2 de Angiotensina/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 39(2): 212-223, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30580570

RESUMO

Objective- Abdominal aortic aneurysm is caused by the accumulation of inflammatory cells in the aortic wall. Our recent studies demonstrated that inhibition of Notch signaling attenuates abdominal aortic aneurysm formation by shifting the macrophage balance towards anti-inflammatory (M2) phenotype. Using IL12p40-/- (interleukin 12 p40) mice, we investigated the effects of M2-predominant macrophages on the development of abdominal aortic aneurysm. Approach and Results- Male (8-10 week-old) wild-type and IL12p40-/- mice (n=15) on C57BL/6 background were infused with Ang II (angiotensin II, 1000 ng/kg per minute) by implanting osmotic pumps subcutaneously for 28 days. In the IL12p40-/- mice, Ang II significantly increased the maximal intraluminal diameter (9/15) as determined by transabdominal ultrasound imaging. In addition, IL12p40-deletion significantly increased aortic stiffness in response to Ang II as measured by pulse wave velocity and atomic force microscopy. Histologically, IL12p40-/- mice exhibited increased maximal external diameter of aorta and aortic lesions associated with collagen deposition and increased elastin fragmentation compared with wild-type mice infused with Ang II. Mechanistically, IL12p40 deficiency by siRNA (small interfering RNA) augmented the Tgfß2-mediated Mmp2 expression in wild-type bone marrow-derived macrophages without affecting the expression of Mmp9. No such effects of IL12p40 deficiency on MMP2/MMP9 was observed in human aortic smooth muscle cells or fibroblasts. Depletion of macrophages in IL12p40-/- mice by clodronate liposomes significantly decreased the maximal external diameter of aorta and aortic stiffness in response to Ang II as determined by imaging and atomic force microscopy. Conclusions- IL12p40 depletion promotes the development of abdominal aortic aneurysm, in part, by facilitating recruitment of M2-like macrophages and potentiating aortic stiffness and fibrosis mediated by Tgfß2.


Assuntos
Angiotensina II/farmacologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Subunidade p40 da Interleucina-12/fisiologia , Animais , Colágeno/metabolismo , Subunidade p40 da Interleucina-12/deficiência , Macrófagos/fisiologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator de Crescimento Transformador beta2/fisiologia , Rigidez Vascular
3.
Can J Physiol Pharmacol ; 95(3): 281-287, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28177677

RESUMO

Mammalian target for rapamycin complex 1 (mTORC1) is a common target for the action of immunosuppressant macrolide rapamycin and glucose-lowering metformin. Inhibition of mTORC1 can exert both beneficial and detrimental effects in different pathologies. Here, we investigated the differential effects of rapamycin (1.2 mg/kg per day delivered subcutaneously for 6 weeks) and metformin (300 mg/kg per day delivered orally for 11 weeks) treatments on male Zucker diabetic fatty (ZDF) rats that mimic the cardiorenal pathology of type 2 diabetic patients and progress to insulin insufficiency. Rapamycin and metformin improved proteinuria, and rapamycin also reduced urinary gamma glutamyl transferase (GGT) indicating improvement of tubular health. Metformin reduced food and water intake, and urinary sodium and potassium, whereas rapamycin increased urinary sodium. Metformin reduced plasma alkaline phosphatase, but induced transaminitis as evidenced by significant increases in plasma AST and ALT. Metformin also induced hyperinsulinemia, but did not suppress fasting plasma glucose after ZDF rats reached 17 weeks of age, and worsened lipid profile. Rapamycin also induced mild transaminitis. Additionally, both rapamycin and metformin increased plasma uric acid and creatinine, biomarkers for cardiovascular and renal disease. These observations define how rapamycin and metformin differentially modulate metabolic profiles that regulate cardiorenal pathology in conditions of severe type 2 diabetes.


Assuntos
Doenças Cardiovasculares/prevenção & controle , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Resistência à Insulina , Metformina/farmacologia , Complexos Multiproteicos/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Biomarcadores/sangue , Biomarcadores/urina , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/etiologia , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/enzimologia , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/prevenção & controle , Modelos Animais de Doenças , Progressão da Doença , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , Proteinúria/enzimologia , Proteinúria/etiologia , Proteinúria/prevenção & controle , Ratos Zucker , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Fatores de Tempo
4.
Can J Physiol Pharmacol ; 95(3): 305-309, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28051329

RESUMO

Both circulating adiponectin (APN) and cardiac APN exert cardioprotective effects and improve insulin sensitivity and mitochondrial function. Low circulating APN serves as a biomarker for cardiovascular risk. Ablation of adiponectin receptor 1 (AdipoR1) causes myocardial mitochondrial dysfunction. Although high salt intake is a contributor to cardiovascular disease, how it modulates the expression of APN or AdipoR1 in cardiomyocytes is not known. We report that APN mRNA expression was attenuated in a dose-dependent manner in mouse cardiomyocyte cell line HL-1 exposed to salt concentrations ranging from 0.75% to 1.5% for 12 h. High-salt exposure (0.88% and 1.25% for 12 h) also suppressed APN and AdipoR1 protein expression significantly in rat cardiac muscle H9c2 cells. Co-immunostaining for AdipoR1 and mitochondrial complex 1 indicated that AdipoR1 may be co-localized with mitochondria. These data show for the first time that high salt is an important suppressor of cardiovascular protective APN and AdipoR1.


Assuntos
Adiponectina/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Receptores de Adiponectina/metabolismo , Cloreto de Sódio/farmacologia , Adiponectina/genética , Angiotensina II/farmacologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Regulação para Baixo , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores de Adiponectina/genética , Transdução de Sinais/efeitos dos fármacos
5.
Am J Physiol Renal Physiol ; 304(8): F1105-13, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23389452

RESUMO

Blockade of the angiotensin (ANG) II receptor type 1 (AT(1)R) with angiotensin receptor blockers (ARBs) is widely used in the treatment of hypertension. However, ARBs are variably effective in reducing blood pressure, likely due, in part, to polymorphisms in the ARB binding pocket of the AT(1)R. Therefore, we need a better understanding of variations/polymorphisms that alter binding of ARBs in heterogeneous patient populations. The opossum proximal tubule cell (OKP) line is commonly used in research to evaluate renal sodium handling and therefore blood pressure. Investigating this issue, we found natural sequence variations in the opossum AT(1)R paralleling those observed in the human AT(1)R. Therefore, we posited that these sequence variations may explain ARB resistance. We demonstrate that OKP cells express AT(1)R mRNA, bind (125)I-labeled ANG II, and exhibit ANG II-induced phosphorylation of Jak2. However, Jak2 phosphorylation is not inhibited by five different ARBs commonly used to treat hypertension. Additionally, nonradioactive ANG II competes (125)I-ANG II efficiently, whereas a 10-fold molar excess of olmesartan and the ANG II receptor type 2 blocker PD-123319 is unable to block (125)I-ANG II binding. In contrast, ANG II binding to OKP cells stably expressing rat AT(1A)Rs, which have a conserved AT(1)R-binding pocket with human AT(1)R, is efficiently inhibited by olmesartan. A novel observation was that resistance to ARB binding to opossum AT(1)Rs correlates with variations from the human receptor at positions 108, 163, 192, and 198 within the ARB-binding pocket. These observations highlight the potential utility of evaluating AT(1)R polymorphisms within the ARB-binding pocket in various hypertensive populations.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Imidazóis/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Gambás/genética , Receptor Tipo 1 de Angiotensina/química , Receptor Tipo 1 de Angiotensina/genética , Tetrazóis/farmacologia , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Sítios de Ligação , Linhagem Celular , Resistência a Medicamentos/genética , Humanos , Radioisótopos do Iodo , Janus Quinase 2/metabolismo , Túbulos Renais Proximais/citologia , Filogenia , Polimorfismo Genético/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , RNA Mensageiro/genética , Ratos , Receptor Tipo 1 de Angiotensina/metabolismo , Especificidade da Espécie , Vasoconstritores/metabolismo , Vasoconstritores/farmacologia
6.
Front Endocrinol (Lausanne) ; 14: 1108449, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36909327

RESUMO

Healthy, premenopausal women have the advantage of female-specific cardiovascular protection compared to age-matched healthy men. However, pathologies such as obesity and Type 2 diabetes mellitus (T2DM) cause losing of this female-specific cardiovascular protection in young, obese and diabetic females. Molecular mechanisms underlying this loss of female-specific cardiovascular protection in young, obese and diabetic females are not clearly elucidated. This review takes a close look at the latest advances in our understanding of sex differences in adult cardiac gene expression patterns in health and disease. Based on the emerging data, this review proposes that female biased gene expression patterns in healthy adult hearts of human and pre-clinical models support the existence of active fetal gene program in healthy, premenopausal female heart compared to age-matched healthy male heart. However, the misalignment of gene expression pattern in this female-specific active cardiac fetal gene program caused by pathologies such as obesity and T2DM may contribute to the loss of female-specific cardiovascular protection in young, obese and diabetic females.


Assuntos
Diabetes Mellitus Tipo 2 , Adulto , Gravidez , Feminino , Humanos , Masculino , Diabetes Mellitus Tipo 2/metabolismo , Coração , Obesidade , Cuidado Pré-Natal , Expressão Gênica
7.
Cells ; 12(10)2023 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-37408206

RESUMO

BACKGROUND: Obese and pre-diabetic women have a higher risk for cardiovascular death than age-matched men with the same symptoms, and there are no effective treatments. We reported that obese and pre-diabetic female Zucker Diabetic Fatty (ZDF-F) rats recapitulate metabolic and cardiac pathology of young obese and pre-diabetic women and exhibit suppression of cardio-reparative AT2R. Here, we investigated whether NP-6A4, a new AT2R agonist with the FDA designation for pediatric cardiomyopathy, mitigate heart disease in ZDF-F rats by restoring AT2R expression. METHODS: ZDF-F rats on a high-fat diet (to induce hyperglycemia) were treated with saline, NP-6A4 (10 mg/kg/day), or NP-6A4 + PD123319 (AT2R-specific antagonist, 5 mg/kg/day) for 4 weeks (n = 21). Cardiac functions, structure, and signaling were assessed by echocardiography, histology, immunohistochemistry, immunoblotting, and cardiac proteome analysis. RESULTS: NP-6A4 treatment attenuated cardiac dysfunction, microvascular damage (-625%) and cardiomyocyte hypertrophy (-263%), and increased capillary density (200%) and AT2R expression (240%) (p < 0.05). NP-6A4 activated a new 8-protein autophagy network and increased autophagy marker LC3-II but suppressed autophagy receptor p62 and autophagy inhibitor Rubicon. Co-treatment with AT2R antagonist PD123319 suppressed NP-6A4's protective effects, confirming that NP-6A4 acts through AT2R. NP-6A4-AT2R-induced cardioprotection was independent of changes in body weight, hyperglycemia, hyperinsulinemia, or blood pressure. CONCLUSIONS: Cardiac autophagy impairment underlies heart disease induced by obesity and pre-diabetes, and there are no drugs to re-activate autophagy. We propose that NP-6A4 can be an effective drug to reactivate cardiac autophagy and treat obesity- and pre-diabetes-induced heart disease, particularly for young and obese women.


Assuntos
Cardiomiopatias , Cardiopatias , Hiperglicemia , Estado Pré-Diabético , Feminino , Ratos , Animais , Ratos Zucker , Obesidade/complicações , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/etiologia
8.
PLoS One ; 18(3): e0282859, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36928870

RESUMO

Chemotherapy-induced impairment of autophagy is implicated in cardiac toxicity induced by anti-cancer drugs. Imperfect translation from rodent models and lack of in vitro models of toxicity has limited investigation of autophagic flux dysregulation, preventing design of novel cardioprotective strategies based on autophagy control. Development of an adult heart tissue culture technique from a translational model will improve investigation of cardiac toxicity. We aimed to optimize a canine cardiac slice culture system for exploration of cancer therapy impact on intact cardiac tissue, creating a translatable model that maintains autophagy in culture and is amenable to autophagy modulation. Canine cardiac tissue slices (350 µm) were generated from left ventricular free wall collected from euthanized client-owned dogs (n = 7) free of cardiovascular disease at the Foster Hospital for Small Animals at Tufts University. Cell viability and apoptosis were quantified with MTT assay and TUNEL staining. Cardiac slices were challenged with doxorubicin and an autophagy activator (rapamycin) or inhibitor (chloroquine). Autophagic flux components (LC3, p62) were quantified by western blot. Cardiac slices retained high cell viability for >7 days in culture and basal levels of autophagic markers remained unchanged. Doxorubicin treatment resulted in perturbation of the autophagic flux and cell death, while rapamycin co-treatment restored normal autophagic flux and maintained cell survival. We developed an adult canine cardiac slice culture system appropriate for studying the effects of autophagic flux that may be applicable to drug toxicity evaluations.


Assuntos
Cardiotoxicidade , Miócitos Cardíacos , Animais , Cães , Miócitos Cardíacos/metabolismo , Cardiotoxicidade/metabolismo , Autofagia , Doxorrubicina/farmacologia , Doxorrubicina/metabolismo , Sirolimo/farmacologia
9.
Am J Physiol Heart Circ Physiol ; 302(11): H2341-51, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22447938

RESUMO

Angiotensin II contributes to myocardial tissue remodeling and interstitial fibrosis through NADPH oxidase-mediated generation of oxidative stress in the progression of heart failure. Recent data have suggested that nebivolol, a third-generation ß-blocker, improves diastolic dysfunction by targeting nitric oxide (NO) and metabolic pathways that decrease interstitial fibrosis. We sought to determine if targeting NO would improve diastolic function in a model of tissue renin-angiotensin system overactivation. We used the transgenic (TG) (mRen2)27 rat, which overexpresses the murine renin transgene and manifests insulin resistance and left ventricular dysfunction. We treated 6- to 7-wk-old TG (mRen2)27 rats and age-matched Sprague-Dawley control rats with nebivolol (10 mg·kg(-1)·day(-1)) or placebo via osmotic minipumps for a period of 21 days. Compared with Sprague-Dawley control rats, TG (mRen2)27 rats displayed a prolonged diastolic relaxation time and reduced initial filling rate associated with increased interstitial fibrosis and left ventricular hypertrophy. These findings were temporally related to increased NADPH oxidase activity and subunits p47(phox) and Rac1 and increased total ROS and peroxynitrite formation in parallel with reductions in the antioxidant heme oxygenase as well as the phosphorylation/activation of endothelial NO synthase and PKB/Akt. Treatment with nebivolol restored diastolic function and interstitial fibrosis through increases in the phosphorylation of 5'-AMP-activated protein kinase, Akt, and endothelial NO synthase and reductions in oxidant stress. These results support that targeting NO with nebivolol treatment improves diastolic dysfunction through reducing myocardial oxidative stress by enhancing 5'-AMP-activated protein kinase and Akt activation of NO biosynthesis.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Benzopiranos/farmacologia , Diástole/efeitos dos fármacos , Etanolaminas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Renina/genética , Renina/fisiologia , Remodelação Ventricular/efeitos dos fármacos , Quinases Proteína-Quinases Ativadas por AMP , Animais , Diástole/fisiologia , Modelos Animais de Doenças , Fibrose , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , NADPH Oxidases/metabolismo , Nebivolol , Óxido Nítrico/metabolismo , Estresse Oxidativo/fisiologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia , Remodelação Ventricular/fisiologia
10.
Am J Physiol Heart Circ Physiol ; 302(8): H1667-82, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22345570

RESUMO

The statistical association between endurance exercise capacity and cardiovascular disease suggests that impaired aerobic metabolism underlies the cardiovascular disease risk in men and women. To explore this connection, we applied divergent artificial selection in rats to develop low-capacity runner (LCR) and high-capacity runner (HCR) rats and found that disease risks segregated strongly with low running capacity. Here, we tested if inborn low aerobic capacity promotes differential sex-related cardiovascular effects. Compared with HCR males (HCR-M), LCR males (LCR-M) were overweight by 34% and had heavier retroperitoneal, epididymal, and omental fat pads; LCR females (LCR-F) were 20% heavier than HCR females (HCR-F), and their retroperitoneal, but not perireproductive or omental, fat pads were heavier as well. Unlike HCR-M, blood pressure was elevated in LCR-M, and this was accompanied by left ventricular (LV) hypertrophy. Like HCR-F, LCR-F exhibited normal blood pressure and LV weight as well as increased spontaneous cage activity compared with males. Despite normal blood pressures, LCR-F exhibited increased myocardial interstitial fibrosis and diastolic dysfunction, as indicated by increased LV stiffness, a decrease in the initial filling rate, and an increase in diastolic relaxation time. Although females exhibited increased arterial stiffness, ejection fraction was normal. Increased interstitial fibrosis and diastolic dysfunction in LCR-F was accompanied by the lowest protein levels of phosphorylated AMP-actived protein kinase [phospho-AMPK (Thr(172))] and silent information regulator 1. Thus, the combination of risk factors, including female sex, intrinsic low aerobic capacity, and overweightness, promote myocardial stiffness/fibrosis sufficient to induce diastolic dysfunction in the absence of hypertension and LV hypertrophy.


Assuntos
Miocárdio/patologia , Sobrepeso/fisiopatologia , Consumo de Oxigênio/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Aerobiose/genética , Aerobiose/fisiologia , Animais , Barorreflexo/genética , Barorreflexo/fisiologia , Pressão Sanguínea/fisiologia , Western Blotting , Cateterismo Cardíaco , Citrato (si)-Sintase/metabolismo , Diástole/fisiologia , Feminino , Fibrose , Testes de Função Cardíaca , Hemodinâmica/fisiologia , Hidroximetilglutaril-CoA Redutases/metabolismo , Imuno-Histoquímica , Imageamento por Ressonância Magnética , Masculino , Microscopia Eletrônica de Transmissão , Miocárdio/ultraestrutura , Sobrepeso/genética , Condicionamento Físico Animal/fisiologia , Resistência Física/fisiologia , Ratos , Corrida/fisiologia , Telemetria , Remodelação Ventricular/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa