Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.120
Filtrar
1.
Pediatrics ; 153(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38098437

RESUMO

Infants with congenital bilateral renal agenesis are at significant risk for morbidity and mortality, despite substantial and continuing advances in fetal and neonatal therapeutics. Infants with bilateral renal agenesis may episodically develop severe hypotension that can be refractory to traditional vasopressors. Synthetic angiotensin-II has been successfully used in adult and a few pediatric patients with refractory hypotension but has not been extensively studied in infants. We describe the use of angiotensin-II in treating refractory hypotension in a premature infant with congenital bilateral renal agenesis admitted to the NICU. Within 48 hours, he no longer required other vasopressors. Subsequently, angiotensin-II was gradually weaned and discontinued over 10 days and the patient was ultimately discharged from the hospital. This case demonstrates that angiotensin-II may be a helpful agent to treat refractory hypotension in infants with bilateral renal agenesis.


Assuntos
Angiotensina II , Hipotensão , Nefropatias , Vasoconstritores , Hipotensão/tratamento farmacológico , Anormalidades Congênitas/tratamento farmacológico , Rim/anormalidades , Nefropatias/congênito , Nefropatias/tratamento farmacológico , Angiotensina II/administração & dosagem , Vasoconstritores/administração & dosagem , Humanos , Recém-Nascido , Lactente
2.
Biomolecules ; 13(6)2023 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-37371479

RESUMO

BACKGROUND: Gasdermin D, a molecule downstream of the nucleotide-binding oligomerization domain-like receptor family pyrin domain containing inflammasome, forms the membrane pore for the secretion of interleukin (IL)-1ß and IL-18, and also mediates pyroptosis. This study was to explore the influence of treatment with disulfiram, a small molecule inhibitor to gasdermin D, on the formation and progression of experimental abdominal aortic aneurysms (AAA). METHODS: AAAs were induced in 10-week-old male apolipoprotein E deficient mice by subcutaneous infusion of angiotensin II (1000 ng/min/kg body weight) for 28 days via osmotic minipumps. Three days prior to angiotensin II infusion, disulfiram (50 mg/kg) or an equal volume of saline as the vehicle control was administered daily via oral gavage. The influence on experimental AAAs was analyzed by serial measurements of aortic diameters via ultrasonography, grading AAA severity and histopathology at sacrifice. Serum IL-1ß and IL-18 levels, systolic blood pressure, total cholesterol, and triglyceride were also measured. Additional experiments assayed the influences on the cell viability and IL-1ß secretion of in vitro activated macrophages. RESULTS: Disulfiram significantly reduced the enlargement, incidence, and severity of angiotensin II-induced experimental AAAs with attenuation of medial elastin breaks, mural macrophage accumulation, and systolic blood pressure. The AAA suppression was also associated with reduced systemic levels of IL-1ß but not IL-18. However, disulfiram treatment had no impact on body weight gain and lipid levels in aneurysmal mice. Additionally, disulfiram treatment also markedly reduced the secretion of IL-1ß from activated macrophages with a limited effect on cell viability in vitro. CONCLUSIONS: Gasdermin D inhibition by disulfiram attenuated angiotensin II-induced experimental AAAs with reduced systemic IL-1ß levels and in vitro activated macrophage IL-1ß secretion. Our study suggests that pharmacological gasdermin D inhibition may have translational potential for limiting clinical AAA progression.


Assuntos
Angiotensina II , Aneurisma da Aorta Abdominal , Animais , Masculino , Camundongos , Angiotensina II/administração & dosagem , Angiotensina II/efeitos adversos , Angiotensina II/uso terapêutico , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/tratamento farmacológico , Aneurisma da Aorta Abdominal/patologia , Peso Corporal , Modelos Animais de Doenças , Dissulfiram/farmacologia , Gasderminas/antagonistas & inibidores , Camundongos Endogâmicos C57BL
3.
Peptides ; 153: 170786, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35304156

RESUMO

Fibroblast growth factors (FGFs) mediate key cardiac functions from development to homeostasis and disease. The current research was to explore the effects of FGF12 in the fibrosis of cardiac function and heart failure, and whether FGF12 alleviated cardiac fibrosis via inhibition of oxidative stress. Ligation of left coronary artery in mice induced heart failure and myocardial infarction (MI). Angiotensin II (Ang II) was administered to cardiac fibroblasts (CFs). FGF12 upregulation or FGF12 transgenic (Tg) mice could improve cardiac dysfunction of MI mice, and attenuated cardiac fibrosis of heart failure induced by MI in mice. FGF12 overexpression suppressed the increases of collagen I, collagen III and fibronectin which was induced by Ang II in CFs. The oxidative stress was enhanced in the heart of MI mice and CFs treated with Ang II, and these enhances were attenuated via FGF12 overexpression. Superoxide dismutase (SOD) overexpression inhibited the enhancements of collagen I, collagen III and fibronectin in the heart of MI mice, and in the CFs treated with Ang II. Overexpression of nicotinamide adenine dinucleotide phosphate oxidases (Nox1) reversed the attenuating influences of FGF12 on the enhancements of collagen I, collagen III and fibronectin in the CFs induced by Ang II. These outcomes showed that FGF12 upregulation can improve cardiac dysfunction and heart fibrosis of heart failure. FGF12 attenuates oxidative stress to suppress the cardiac fibrosis.


Assuntos
Fatores de Crescimento de Fibroblastos , Insuficiência Cardíaca , Infarto do Miocárdio , Miocárdio , Estresse Oxidativo , Remodelação Ventricular , Angiotensina II/administração & dosagem , Animais , Colágeno/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrose , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia
4.
Oxid Med Cell Longev ; 2022: 5044046, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35222798

RESUMO

Cardiac lymphatic vessel growth (lymphangiogenesis) and integrity play an essential role in maintaining tissue fluid balance. Inhibition of lymphatic lymphangiogenesis is involved in cardiac edema and cardiac remodeling after ischemic injury or pressure overload. However, whether lymphatic vessel integrity is disrupted during angiotensin II- (Ang II-) induced cardiac remodeling remains to be investigated. In this study, cardiac remodeling models were established by Ang II (1000 ng/kg/min) in VEGFR-3 knockdown (Lyve-1Cre VEGFR-3f/-) and wild-type (VEGFR-3f/f) littermates. Our results indicated that Ang II infusion not only induced cardiac lymphangiogenesis and upregulation of VEGF-C and VEGFR-3 expression in the time-dependent manner but also enhanced proteasome activity, MKP5 and VE-cadherin degradation, p38 MAPK activation, and lymphatic vessel hyperpermeability. Moreover, VEGFR-3 knockdown significantly inhibited cardiac lymphangiogenesis in mice, resulting in exacerbation of tissue edema, hypertrophy, fibrosis superoxide production, inflammation, and heart failure (HF). Conversely, administration of epoxomicin (a selective proteasome inhibitor) markedly mitigated Ang II-induced cardiac edema, remodeling, and dysfunction; upregulated MKP5 and VE-cadherin expression; inactivated p38 MAPK; and reduced lymphatic vessel hyperpermeability in WT mice, indicating that inhibition of proteasome activity is required to maintain lymphatic endothelial cell (LEC) integrity. Our results show that both cardiac lymphangiogenesis and lymphatic barrier hyperpermeability are implicated in Ang II-induced adaptive hypertrophic remodeling and dysfunction. Proteasome-mediated hyperpermeability of LEC junctions plays a predominant role in the development of cardiac remodeling. Selective stimulation of lymphangiogenesis or inhibition of proteasome activity may be a potential therapeutic option for treating hypertension-induced cardiac remodeling.


Assuntos
Angiotensina II/metabolismo , Cardiomegalia/metabolismo , Edema Cardíaco/metabolismo , Vasos Linfáticos/metabolismo , Angiotensina II/administração & dosagem , Animais , Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Edema Cardíaco/tratamento farmacológico , Edema Cardíaco/patologia , Edema Cardíaco/fisiopatologia , Células Endoteliais/metabolismo , Linfangiogênese/efeitos dos fármacos , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Permeabilidade/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/deficiência , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Arterioscler Thromb Vasc Biol ; 42(3): 277-288, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35045728

RESUMO

AngII (angiotensin II) infusion in mice has been used to provide mechanistic insight into human abdominal aortic aneurysms for over 2 decades. This is a technically facile animal model that recapitulates multiple facets of the human disease. Although numerous publications have reported abdominal aortic aneurysms with AngII infusion in mice, there remain many fundamental unanswered questions such as uniformity of describing the pathological characteristics and which cell type is stimulated by AngII to promote abdominal aortic aneurysms. Extrapolation of the findings to provide insight into the human disease has been hindered by the preponderance of studies designed to determine the effects on initiation of abdominal aortic aneurysms, rather than a more clinically relevant scenario of determining efficacy on the established disease. The purpose of this review is to enhance understanding of AngII-induced abdominal aortic pathologies in mice, thereby providing greater insight into the human disease.


Assuntos
Angiotensina II/toxicidade , Aneurisma da Aorta Abdominal/etiologia , Fatores Etários , Angiotensina II/administração & dosagem , Animais , Aneurisma da Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/terapia , Pressão Sanguínea/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Fatores de Risco de Doenças Cardíacas , Humanos , Hipercolesterolemia/complicações , Masculino , Camundongos , Camundongos Transgênicos , Modelos Cardiovasculares , Receptores de Angiotensina/efeitos dos fármacos , Fatores Sexuais
6.
Int J Mol Sci ; 22(24)2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34948475

RESUMO

Compound 21 (C21), an AT2 receptor agonist, and Angiotensin 1-7 (Ang 1-7), through Mas receptor, play an important role in the modulation of the protective arm of the renin-angiotensin system. The aim of this study was to investigate in an experimental model of angiotensin II-dependent hypertension whether the activation of the potentially protective arm of the renin-angiotensin system, through AT2 or Mas receptor stimulation, counteracts the onset of myocardial fibrosis and hypertrophy, and whether these effects are mediated by inflammatory mechanism and/or sympathetic activation. Sprague Dawley rats (n = 67) were treated for 1 (n = 25) and 4 (n = 42) weeks and divided in the following groups: (a) Angiotensin II (Ang II, 200 ng/kg/min, osmotic minipumps, sub cutis); (b) Ang II+Compound 21 (C21, 0.3 mg/kg/day, intraperitoneal); (c) Ang II+Ang 1-7 (576 µg/kg/day, intraperitoneal); (d) Ang II+Losartan (50 mg/kg/day, per os); (e) control group (physiological saline, sub cutis). Systolic blood pressure was measured by tail cuff method and, at the end of the experimental period, the rats were euthanized and the heart was excised to evaluate myocardial fibrosis, hypertrophy, inflammatory cell infiltration and tyrosine hydroxylase expression, used as marker of sympathetic activity. Ang II caused a significant increase of blood pressure, myocardial interstitial and perivascular fibrosis and myocardial hypertrophy, as compared to control groups. C21 or Ang 1-7 administration did not modify the increase in blood pressure in Ang II treated rats, but both prevented the development of myocardial fibrosis and hypertrophy. Treatment with losartan blocked the onset of hypertension and myocardial fibrosis and hypertrophy in Ang II treated rats. Activation of AT2 receptors or Mas receptors prevents the onset of myocardial fibrosis and hypertrophy in Ang II-dependent hypertension through the reduction of myocardial inflammatory cell infiltration and tyrosine hydroxylase expression. Unlike what happens in case of treatment with losartan, the antifibrotic and antihypertrophic effects that follow the activation of the AT2 or Mas receptors are independent on the modulation of blood pressure.


Assuntos
Angiotensina II/administração & dosagem , Angiotensina I/administração & dosagem , Cardiomegalia/prevenção & controle , Hipertensão/tratamento farmacológico , Imidazóis/administração & dosagem , Losartan/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Sulfonamidas/administração & dosagem , Tiofenos/administração & dosagem , Angiotensina I/farmacologia , Angiotensina II/farmacologia , Animais , Cardiomegalia/metabolismo , Modelos Animais de Doenças , Fibrose , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Imidazóis/farmacologia , Injeções Intraperitoneais , Losartan/farmacologia , Masculino , Fragmentos de Peptídeos/farmacologia , Proto-Oncogene Mas/metabolismo , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia , Tiofenos/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo
7.
Biomolecules ; 11(12)2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34944506

RESUMO

Renin-angiotensin systems produce angiotensin II (Ang II) and angiotensin 1-7 (Ang 1-7), which are able to induce opposite effects on circulation. This study in vivo assessed the effects induced by Ang II or Ang 1-7 on rat pial microcirculation during hypoperfusion-reperfusion, clarifying the mechanisms causing the imbalance between Ang II and Ang 1-7. The fluorescence microscopy was used to quantify the microvascular parameters. Hypoperfusion and reperfusion caused vasoconstriction, disruption of blood-brain barrier, reduction of capillary perfusion and an increase in reactive oxygen species production. Rats treated with Ang II showed exacerbated microvascular damage with stronger vasoconstriction compared to hypoperfused rats, a further increase in leakage, higher decrease in capillary perfusion and marker oxidative stress. Candesartan cilexetil (specific Ang II type 1 receptor (AT1R) antagonist) administration prior to Ang II prevented the effects induced by Ang II, blunting the hypoperfusion-reperfusion injury. Ang 1-7 or ACE2 activator administration, preserved the pial microcirculation from hypoperfusion-reperfusion damage. These effects of Ang 1-7 were blunted by a Mas (Mas oncogene-encoded protein) receptor antagonist, while Ang II type 2 receptor antagonists did not affect Ang 1-7-induced changes. In conclusion, Ang II and Ang 1-7 triggered different mechanisms through AT1R or MAS receptors able to affect cerebral microvascular injury.


Assuntos
Angiotensina II/administração & dosagem , Angiotensina I/administração & dosagem , Benzimidazóis/administração & dosagem , Compostos de Bifenilo/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Pia-Máter/irrigação sanguínea , Traumatismo por Reperfusão/metabolismo , Tetrazóis/administração & dosagem , Angiotensina I/efeitos adversos , Angiotensina II/efeitos adversos , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo/farmacologia , Feminino , Masculino , Microcirculação/efeitos dos fármacos , Microscopia de Fluorescência , Fragmentos de Peptídeos/efeitos adversos , Pia-Máter/efeitos dos fármacos , Pia-Máter/metabolismo , Proto-Oncogene Mas/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Tetrazóis/farmacologia
8.
Oxid Med Cell Longev ; 2021: 2860488, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34950416

RESUMO

Acute myocardial infarction (AMI) is a major cause of morbidity and mortality worldwide. Angiotensin (Ang) IV possesses many biological properties that are not yet completely understood. Therefore, we investigated the function and mechanism of Ang IV in AMI in in vivo and in vitro conditions. AMI was performed by ligation of the left anterior descending coronary artery (LAD) in male C57 mice. Ang IV was continuously infused by a minipump 3 d before AMI for 33 d. The neonatal rat ventricular myocytes (NRVCs) were stimulated with Ang IV and cultured under hypoxic conditions. In vivo, Ang IV infusion significantly reduced the mortality after AMI. By the 7th day after AMI, compared with the AMI group, Ang IV reduced the inflammatory cytokine expression. Moreover, terminal deoxyribonucleotidyl transferase- (TDT-) mediated dUTP nick-end labeling (TUNEL) assay showed that Ang IV infusion reduced AMI-induced cardiomyocyte apoptosis. Compared with AMI, Ang IV reduced autophagosomes in cardiomyocytes and improved mitochondrial swelling and disarrangement, as assessed by transmission electron microscopy. By 30th day after AMI, Ang IV significantly reduced the ratio of heart weight to body weight. Echocardiography showed that Ang IV improved impaired cardiac function. Hematoxylin and eosin (H&E) and Masson staining showed that Ang IV infusion reduced the infarction size and myocardial fibrosis. In vitro, dihydroethidium (DHE) staining and comet assay showed that, compared with the hypoxia group, Ang IV reduced oxidative stress and DNA damage. Enzyme-linked immunosorbent assay (ELISA) showed that Ang IV reduced hypoxia-induced secretion of the tumor necrosis factor- (TNF-) ɑ and interleukin- (IL-) 1ß. In addition, compared with the hypoxia group, Ang IV reduced the transformation of light chain 3- (LC3-) I to LC3-II but increased p62 expression and decreased cardiomyocyte apoptosis. Overall, the present study showed that Ang IV reduced the inflammatory response, autophagy, and fibrosis after AMI, leading to reduced infarction size and improved cardiac function. Therefore, administration of Ang IV may be a feasible strategy for the treatment of AMI.


Assuntos
Angiotensina II/análogos & derivados , Autofagia , Cardiomiopatias/prevenção & controle , Inflamação/tratamento farmacológico , Infarto do Miocárdio/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Angiotensina II/administração & dosagem , Angiotensina II/farmacologia , Animais , Apoptose , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Células Cultivadas , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Estresse Oxidativo , Ratos
9.
Peptides ; 146: 170669, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34624429

RESUMO

Alarin could alleviate myocardial infarction-induced heart failure. The present study was to explore whether alarin could alleviate myocardial hypertrophy via inhibiting cyclic adenosine monophosphate/protein kinase A (cAMP/PKA) signaling pathway to attenuate autophagy. Myocardial hypertrophy was induced by angiotensin (Ang) II infusion in vivo in mice and by Ang II treatment of neonatal rat cardiomyocytes (NRCMs) in vitro. The Ang II-induced hypertrophy and fibrosis of the heart were alleviated after alarin administration in mice. The increased atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and beta-myosin heavy chain (ß-MHC), and the decreased alpha-myosin heavy chain (α-MHC) induced by Ang II were reversed by alarin treatment in NRCMs. Alarin inhibited the increases of cAMP and PKA in NRCMs. Treatment with cAMP or overexpression of PKA blocked the attenuating effects of alarin on Ang II-induced hypertrophy in NRCMs. Alarin reduced the Ang II-induced increases of LC3, Beclin 1, autophagy-related gene (Atg)3 and Atg5 in NRCMs. The overexpression of cAMP and PKA reversed the alleviating effects of alarin on the increased autophagy induced by Ang II in NRCMs. These results indicated that alarin could moderate cardiac remodeling. Alarin improved myocardial hypertrophy via inhibiting the cAMP/PKA signaling pathway to attenuate autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Cardiomegalia/prevenção & controle , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Peptídeo Semelhante a Galanina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Angiotensina II/administração & dosagem , Animais , Linhagem Celular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Galanina/antagonistas & inibidores
10.
Sci Rep ; 11(1): 21193, 2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34707201

RESUMO

Endothelial dysfunction (ED) is a key factor for the development of cardiovascular diseases. Due to its chronic, life-threatening nature, ED only can be studied experimentally in animal models. Therefore, this work was aimed to characterize a murine model of ED induced by a daily intraperitoneal administration of angiotensin II (AGII) for 10 weeks. Oxidative stress, inflammation, vascular remodeling, hypertension, and damage to various target organs were evaluated in treated animals. The results indicated that a chronic intraperitoneal administration of AGII increases the production of systemic soluble VCAM, ROS and ICAM-1 expression, and the production of TNFα, IL1ß, IL17A, IL4, TGFß, and IL10 in the kidney, as well as blood pressure levels; it also promotes vascular remodeling and induces non-alcoholic fatty liver disease, glomerulosclerosis, and proliferative retinopathy. Therefore, the model herein proposed can be a representative model for ED; additionally, it is easy to implement, safe, rapid, and inexpensive.


Assuntos
Angiotensina II/administração & dosagem , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Doenças Vasculares/metabolismo , Angiotensina II/toxicidade , Animais , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Infusões Parenterais , Molécula 1 de Adesão Intercelular/metabolismo , Interleucinas/metabolismo , Rim/metabolismo , Rim/patologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Doenças Vasculares/etiologia , Doenças Vasculares/patologia , Remodelação Vascular
11.
Physiol Rep ; 9(17): e15000, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34491001

RESUMO

Autophagy is an evolutionarily conserved cellular catabolic process essential for cell homeostasis, and thus its failure is associated with several diseases. While autophagy has been reported to play a role in vascular smooth muscle cells (SMCs) in vascular disorders, its precise role in the pathogenesis of abdominal aortic aneurysm (AAA) has not yet been elucidated. In this study, we investigated the role of SMC autophagy in AAA formation. As a mouse model of AAA, we used control apolipoprotein E-deficient (apoeKO) mice and Atg7cKO (SMC-specific Atg7-deficient mice):apoeKO mice administered angiotensin II for 4 weeks. Intriguingly, Kaplan-Meier curves showed that the survival rates of Atg7cKO:apoeKO mice were significantly higher than those of apoeKO mice. The hematoma area in AAA of Atg7cKO:apoeKO mice was smaller than in apoeKO mice despite the lack of a significant difference in AAA incidence between the two groups. Furthermore, the amount of granulomatous tissues was significantly larger and the collagen-positive area within AAA was significantly larger in Atg7cKO:apoeKO mice than in apoeKO mice. In accordance with these findings, SMCs cultured from Atg7cKO mice showed increased expression of collagens, independent of angiotensin II action. Taken together, our data suggest that defective autophagy in SMCs elicits AAA healing that may underlie the better survival rate under dyslipidemia and angiotensin II infusion.


Assuntos
Angiotensina II/toxicidade , Aneurisma da Aorta Abdominal/patologia , Autofagia/fisiologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Angiotensina II/administração & dosagem , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Autofagia/efeitos dos fármacos , Células Cultivadas , Bombas de Infusão Implantáveis , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos
12.
Exp Cell Res ; 406(1): 112761, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34339675

RESUMO

Stresses, such as neurohumoral activation, induced pathological cardiac hypertrophy is the main risk factor for heart failure. The ubiquitin-proteasome system (UPS) plays a key role in maintaining protein homeostasis and cardiac function. However, research on the role and mechanism of deubiquitinating enzymes (DUBs) in cardiac hypertrophy is limited. Here, we observe that the deubiquitinating enzyme ubiquitin-specific protease 12(USP12) is upregulated in Ang II-induced hypertrophic hearts and primary neonatal rat cardiomyocytes (NRCMs). Inhibition of USP12 ameliorate Ang II-induced myocardial hypertrophy, while overexpression of USP12 have the opposite effect. USP12 deficiency also significantly attenuate the phenotype of Ang II-induced cardiac hypertrophy in vivo. Moreover, we demonstrate that USP12 aggravate Ang II-induced cardiac hypertrophy by enhancing METTL3, a methyltransferase which catalyze N6-methyladenosine (m6A) modification on messenger RNA and acts as a harmful factor in pathological cardiac hypertrophy. Upregulation of METTL3 reverse the reduction of myocardial hypertrophy induced by USP12 silencing in NRCMs. In contrast, knockdown of METTL3 attenuate the aggravation of myocardial hypertrophy in USP12-overexpressing NRCMs. Furthermore, we discover that USP12 promote the expression of METTL3 via upregulating p300. Mechanistically, USP12 binds and stabilizes p300, thereby activating the transcription of its downstream gene METTL3. Finally, our data show that USP12 is partially dependent on the stabilization of p300 to activate METTL3 expression and promote myocardial hypertrophy. Taken together, our results demonstrate that USP12 acts as a pro-hypertrophic deubiquitinating enzyme via enhancing p300/METTL3 axis, indicating that targeting USP12 could be a potential treatment strategy for pathological cardiac hypertrophy.


Assuntos
Cardiomegalia/genética , Proteína p300 Associada a E1A/genética , Metiltransferases/genética , Miócitos Cardíacos/metabolismo , Ubiquitina Tiolesterase/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Angiotensina II/administração & dosagem , Animais , Animais Recém-Nascidos , Cardiomegalia/induzido quimicamente , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Proteína p300 Associada a E1A/metabolismo , Regulação da Expressão Gênica , Masculino , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/citologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Ubiquitina Tiolesterase/metabolismo , Ubiquitinação
13.
J Cardiovasc Pharmacol ; 78(4): 551-559, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34269699

RESUMO

ABSTRACT: We have previously shown increased vascular reactivity to angiotensin (Ang) II in familial combined hyperlipidemia. However, this has not been well studied in familial hypercholesterolemia (FH), a condition with incipient endothelial dysfunction. This study aimed to examine microvascular and macrovascular responses to Ang II in FH. Therefore, we investigated the effects of a 3-hour infusion of Ang II on blood pressure and forearm skin microvascular function in 16 otherwise healthy patients with FH and matched healthy controls. Skin microvascular hyperemia was studied by laser Doppler fluxmetry during local heating. Microvascular resistance was determined by the ratio of mean arterial pressure to microvascular hyperemia. Macrovascular reactivity was assessed by changes in brachial blood pressure. Compared with the controls, the FH group had increased baseline systolic blood pressure (127 ± 14 vs. 115 ± 12 mm Hg; P = 0.02), while systolic blood pressure responses were similar (+24 ± 9 vs. +21 ± 7 mm Hg; P = 0.26) after 3 hours of Ang II infusion. At baseline, there were no group differences in microvascular hyperemia or resistance. However, after 3 hours of Ang II infusion, heat-induced microvascular hyperemia was less pronounced in FH (126 ± 95 vs. 184 ± 102 arbitrary units; P = 0.01), while microvascular resistance during heat-induced hyperemia was increased (1.9 ± 0.9 vs. 0.9 ± 0.8, P = 0.01), as compared to controls. Both these responses were further pronounced 1 hour after stopping Ang II. In conclusion, despite similar blood pressure responses to Ang II in the FH group and controls, microvascular dilatation capacity was impaired in the FH group, indicating endothelial dysfunction. These findings and increased microvascular resistance may lead to hypertension and microvascular complications in FH.


Assuntos
Angiotensina II/administração & dosagem , Pressão Arterial/efeitos dos fármacos , Artéria Braquial/efeitos dos fármacos , Hiperlipoproteinemia Tipo II/fisiopatologia , Microcirculação/efeitos dos fármacos , Pele/irrigação sanguínea , Vasodilatação/efeitos dos fármacos , Adulto , Angiotensina II/sangue , Artéria Braquial/fisiopatologia , Estudos de Casos e Controles , Feminino , Antebraço , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/diagnóstico , Infusões Intravenosas , Fluxometria por Laser-Doppler , Masculino , Pessoa de Meia-Idade , Fatores de Tempo
14.
Nephron ; 145(5): 518-527, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34062539

RESUMO

BACKGROUND: Increasing evidence suggests that angiotensin II (Ang II), the bioactive pro-oxidant in the renin-angiotensin system, aggravates fibrosis, and the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is involved in multiple diseases, such as renal fibrosis. However, the role and underlying mechanism of Ang II in renal fibrosis remain unclear. Here, we investigated whether the NLRP3 inflammasome mediated Ang II-induced renal fibrosis, as well as the downstream pathways involved in this process. METHODS: NLRP3-/- mice were used as a model to study Ang II-infused renal fibrosis. Mice were divided into 4 groups: sham wild type, Ang II-infused wild type, sham NLRP3-/-, and Ang II-infused NLRP3-/- groups. Ang II infusion-induced renal injury was confirmed by periodic acid-Schiff and Masson's staining, immunohistochemistry, and transmission electron microscopy (TEM). Mitochondrial morphology was presented on TEM micrographs, and mitochondrial function was reflected by the protein levels of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), mitochondrial transcription factor A (TFAM), dynamin-related protein 1 (DRP1), and mitofusin 2 (MFN2), as assessed by Western blotting. Endoplasmic reticulum (ER) stress was characterized by changes in the levels of ER chaperones, such as GRP94, BiP, CHOP, and caspase 12. RESULTS: Ang II infusion increased cell proliferation, extracellular matrix overproduction, inflammatory cell infiltration, and glomerulosclerosis and induced obvious morphological abnormalities in podocytes. Ang II infusion promoted mitochondrial damage, as indicated by TEM, and induced mitochondrial dysfunction, as evidenced by downregulation of PGC-1α, TFAM, and increased mitochondrial ROS. In addition, DRP1 expression was upregulated, while MFN2 expression was markedly decreased. The levels of GRP94, BiP, CHOP, and caspase 12 were significantly increased. However, all these detrimental effects were attenuated by NLRP3 deletion. CONCLUSIONS: NLRP3 deletion may attenuate angiotensin II-induced renal fibrosis by improving mitochondrial dysfunction and ER stress.


Assuntos
Angiotensina II/fisiologia , Estresse do Retículo Endoplasmático , Nefropatias/fisiopatologia , Mitocôndrias/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Angiotensina II/administração & dosagem , Animais , Humanos , Nefropatias/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo
15.
Nitric Oxide ; 113-114: 57-69, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34091009

RESUMO

Arterial hypertension is one of the major health risk factors leading to coronary artery disease, stroke or peripheral artery disease. Dietary uptake of inorganic nitrite (NO2-) and nitrate (NO3-) via vegetables leads to enhanced vascular NO bioavailability and provides antihypertensive effects. The present study aims to understand the underlying vasoprotective effects of nutritional NO2- and NO3- co-therapy in mice with angiotensin-II (AT-II)-induced arterial hypertension. High-dose AT-II (1 mg/kg/d, 1w, s. c.) was used to induce arterial hypertension in male C57BL/6 mice. Additional inorganic nitrite (7.5 mg/kg/d, p. o.) or nitrate (150 mg/kg/d, p. o.) were administered via the drinking water. Blood pressure (tail-cuff method) and endothelial function (isometric tension) were determined. Oxidative stress and inflammation markers were quantified in aorta, heart, kidney and blood. Co-treatment with inorganic nitrite, but not with nitrate, normalized vascular function, oxidative stress markers and inflammatory pathways in AT-II treated mice. Of note, the highly beneficial effects of nitrite on all parameters and the less pronounced protection by nitrate, as seen by improvement of some parameters, were observed despite no significant increase in plasma nitrite levels by both therapies. Methemoglobin levels tended to be higher upon nitrite/nitrate treatment. Nutritional nitric oxide precursors represent a non-pharmacological treatment option for hypertension that could be applied to the general population (e.g. by eating certain vegetables). The more beneficial effects of inorganic nitrite may rely on superior NO bioactivation and stronger blood pressure lowering effects. Future large-scale clinical studies should investigate whether hypertension and cardiovascular outcome in general can be influenced by dietary inorganic nitrite therapy.


Assuntos
Anti-Hipertensivos/farmacologia , Hipertensão/tratamento farmacológico , Nitratos/farmacologia , Nitritos/farmacologia , Administração Oral , Angiotensina II/administração & dosagem , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/sangue , Pressão Sanguínea/efeitos dos fármacos , Hipertensão/induzido quimicamente , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nitratos/administração & dosagem , Nitratos/sangue , Nitritos/administração & dosagem , Nitritos/sangue , Estresse Oxidativo/efeitos dos fármacos
17.
J Am Heart Assoc ; 10(9): e018716, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33870711

RESUMO

Background Increased potassium intake lowers blood pressure in patients with hypertension, but increased potassium intake also elevates plasma concentrations of the blood pressure-raising hormone aldosterone. Besides its well-described renal effects, aldosterone is also believed to have vascular effects, acting through mineralocorticoid receptors present in endothelial and vascular smooth muscle cells, although mineralocorticoid receptors-independent actions are also thought to be involved. Methods and Results To gain further insight into the effect of increased potassium intake and potassium-stimulated hyperaldosteronism on the human cardiovascular system, we conducted a randomized placebo-controlled double-blind crossover study in 25 healthy normotensive men, where 4 weeks treatment with a potassium supplement (90 mmol/day) was compared with 4 weeks on placebo. At the end of each treatment period, we measured potassium and aldosterone in plasma and performed an angiotensin II (AngII) infusion experiment, during which we assessed the aldosterone response in plasma. Hemodynamics were also monitored during the AngII infusion using ECG, impedance cardiography, finger plethysmography (blood pressure-monitoring), and Doppler ultrasound. The study showed that higher potassium intake increased plasma potassium (mean±SD, 4.3±0.2 versus 4.0±0.2 mmol/L; P=0.0002) and aldosterone (median [interquartile range], 440 [336-521] versus 237 [173-386] pmol/L; P<0.0001), and based on a linear mixed model for repeated measurements, increased potassium intake potentiated AngII-stimulated aldosterone secretion (P=0.0020). In contrast, the hemodynamic responses (blood pressure, total peripheral resistance, cardiac output, and renal artery blood flow) to AngII were similar after potassium and placebo. Conclusions Increased potassium intake potentiates AngII-stimulated aldosterone secretion without affecting systemic cardiovascular hemodynamics in healthy normotensive men. Registration EudraCT Number: 2013-004460-66; URL: https://www.ClinicalTrials.gov; Unique identifier: NCT02380157.


Assuntos
Angiotensina II/administração & dosagem , Pressão Sanguínea/fisiologia , Hipertensão/terapia , Potássio na Dieta/farmacocinética , Potássio/sangue , Adulto , Aldosterona/sangue , Biomarcadores/sangue , Estudos Cross-Over , Método Duplo-Cego , Feminino , Seguimentos , Voluntários Saudáveis , Humanos , Hipertensão/fisiopatologia , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Vasoconstritores/administração & dosagem , Adulto Jovem
18.
Nat Commun ; 12(1): 2417, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33893295

RESUMO

SARS-CoV-2 uses ACE2, an inhibitor of the Renin-Angiotensin-Aldosterone System (RAAS), for cellular entry. Studies indicate that RAAS imbalance worsens the prognosis in COVID-19. We present a consecutive retrospective COVID-19 cohort with findings of frequent pulmonary thromboembolism (17%), high pulmonary artery pressure (60%) and lung MRI perfusion disturbances. We demonstrate, in swine, that infusing angiotensin II or blocking ACE2 induces increased pulmonary artery pressure, reduces blood oxygenation, increases coagulation, disturbs lung perfusion, induces diffuse alveolar damage, and acute tubular necrosis compared to control animals. We further demonstrate that this imbalanced state can be ameliorated by infusion of an angiotensin receptor blocker and low-molecular-weight heparin. In this work, we show that a pathophysiological state in swine induced by RAAS imbalance shares several features with the clinical COVID-19 presentation. Therefore, we propose that severe COVID-19 could partially be driven by a RAAS imbalance.


Assuntos
COVID-19/fisiopatologia , Pulmão/fisiopatologia , Sistema Renina-Angiotensina/fisiologia , SARS-CoV-2/isolamento & purificação , Angiotensina II/administração & dosagem , Angiotensina II/metabolismo , Antagonistas de Receptores de Angiotensina/administração & dosagem , Enzima de Conversão de Angiotensina 2/antagonistas & inibidores , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , COVID-19/metabolismo , COVID-19/virologia , Feminino , Humanos , Pulmão/diagnóstico por imagem , Pulmão/virologia , Imageamento por Ressonância Magnética/métodos , Ligação Proteica/efeitos dos fármacos , Estudos Retrospectivos , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Suínos , Internalização do Vírus/efeitos dos fármacos
19.
Clin Sci (Lond) ; 135(7): 911-924, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33782695

RESUMO

Hypertension is a complex and multifactorial disorder caused by lifestyle and environmental factors, inflammation and disease-related genetic factors and is a risk factor for stroke, ischemic heart disease and renal failure. Although circulating monocytes and tissue macrophages contribute to the pathogenesis of hypertension, the underlying mechanisms are poorly understood. Cysteine rich protein 1 (CRIP1) is highly expressed in immune cells, and CRIP1 mRNA expression in monocytes associates with blood pressure (BP) and is up-regulated by proinflammatory modulation suggesting a link between CRIP1 and BP regulation through the immune system. To address this functional link, we studied CRIP1 expression in immune cells in relation to BP using a human cohort study and hypertensive mouse models. CRIP1 expression in splenic monocytes/macrophages and in circulating monocytes was significantly affected by angiotensin II (Ang II) in a BP-elevating dose (2 mg/kg/day). In the human cohort study, monocytic CRIP1 expression levels were associated with elevated BP, whereas upon differentiation of monocytes to macrophages this association along with the CRIP1 expression level was diminished. In conclusion, CRIP1-positive circulating and splenic monocytes seem to play an important role in hypertension related inflammatory processes through endogenous hormones such as Ang II. These findings suggest that CRIP1 may affect the interaction between the immune system, in particular monocytes, and the pathogenesis of hypertension.


Assuntos
Proteínas de Transporte/metabolismo , Hipertensão/fisiopatologia , Monócitos/metabolismo , Angiotensina II/administração & dosagem , Animais , Pressão Sanguínea , Proteínas de Transporte/genética , Diferenciação Celular , Estudos de Coortes , Modelos Animais de Doenças , Humanos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Macrófagos , Masculino , NG-Nitroarginina Metil Éster/administração & dosagem , Baço , Transcriptoma
20.
FASEB J ; 35(3): e21419, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33566370

RESUMO

In the early phase of the Coronavirus disease 2019 (COVID-19) pandemic, it was postulated that the renin-angiotensin-system inhibitors (RASi) increase the infection risk. This was primarily based on numerous reports, which stated that the RASi could increase the organ Angiotensin-converting enzyme 2 (ACE2), the receptor of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in rodents. RASi can theoretically antagonize the potential influence of angiotensin II (Ang II) on ACE2. However, while Ang II decreases the ACE2 levels in cultured cells, there is little evidence that supports this phenomenon in living animals. In this study, we tested whether Ang II or Ang II combined with its antagonist would alter the ACE2 and other molecules associated with the infection of SARS-CoV-2. Male C57BL6/J mice were administered vehicle, Ang II (400 ng/kg/min), or Ang II with losartan (10 mg/kg/min) for 2 weeks. ACE2 knockout mice were used as a negative control for the ACE2 assay. We found that both Ang II, which elevated blood pressure by 30 mm Hg, and Ang II with losartan, had no effect on the expression or protein activity of ACE2 in the lung, left ventricle, kidney, and ileum. Likewise, these interventions had no effect on the expression of Transmembrane Protease Serine 2 (TMPRSS2) and Furin, proteases that facilitate the virus-cell fusion, and the expression or activity of Tumor Necrosis Factor α-Convertase (TACE) that cleaves cell-surface ACE2. Collectively, physiological concentrations of Ang II do not modulate the molecules associated with SARS-CoV-2 infection. These results support the recent observational studies suggesting that the use of RASi is not a risk factor for COVID-19.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Angiotensina II/farmacologia , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/metabolismo , Losartan/farmacologia , SARS-CoV-2 , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Angiotensina II/administração & dosagem , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Enzima de Conversão de Angiotensina 2/genética , Animais , Furina/genética , Furina/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Losartan/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Vasoconstritores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...