Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.422
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Comput Biol ; 18(2): e1009732, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35202400

RESUMO

In vertebrates, the octopeptide angiotensin II (AngII) is an important in vivo regulator of the cardiovascular system. It acts mainly through two G protein-coupled receptors, AT1 and AT2. To better understand distinctive features of these receptors, we carried out a phylogenetic analysis that revealed a mirror evolution of AT1 and AT2, each one split into two clades, separating fish from terrestrial receptors. It also revealed that hallmark mutations occurred at, or near, the sodium binding site in both AT1 and AT2. Electrostatics computations and molecular dynamics simulations support maintained sodium binding to human AT1 with slow ingress from the extracellular side and an electrostatic component of the binding free energy around -3kT, to be compared to around -2kT for human AT2 and the δ opioid receptor. Comparison of the sodium binding modes in wild type and mutated AT1 and AT2 from humans and eels indicates that the allosteric control by sodium in both AT1 and AT2 evolved during the transition from fish to amniota. The unusual S7.46N mutation in AT1 is mirrored by a L3.36M mutation in AT2. In the presence of sodium, the N7.46 pattern in amniota AT1 stabilizes the inward orientation of N3.35 in the apo receptor, which should contribute to efficient N3.35 driven biased signaling. The M3.36 pattern in amniota AT2 favours the outward orientation of N3.35 and the receptor promiscuity. Both mutations have physiological consequences for the regulation of the renin-angiotensin system.


Assuntos
Angiotensina II , Receptor Tipo 2 de Angiotensina , Angiotensina II/genética , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Filogenia , Receptor Tipo 2 de Angiotensina/genética , Receptor Tipo 2 de Angiotensina/metabolismo , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Sódio , Vertebrados/genética
2.
J Biol Chem ; 295(29): 9986-9997, 2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32493776

RESUMO

G-protein-coupled receptors (GPCRs) are a ubiquitously expressed family of receptor proteins that regulate many physiological functions and other proteins. They act through two dissociable signaling pathways: the exchange of GDP to GTP by linked G-proteins and the recruitment of ß-arrestins. GPCRs modulate several members of the transient receptor potential (TRP) channel family of nonselective cation channels. How TRP channels reciprocally regulate GPCR signaling is less well-explored. Here, using an array of biochemical approaches, including immunoprecipitation and fluorescence, calcium imaging, phosphate radiolabeling, and a ß-arrestin-dependent luciferase assay, we characterize a GPCR-TRP channel pair, angiotensin II receptor type 1 (AT1R), and transient receptor potential vanilloid 4 (TRPV4), in primary murine choroid plexus epithelial cells and immortalized cell lines. We found that AT1R and TRPV4 are binding partners and that activation of AT1R by angiotensin II (ANGII) elicits ß-arrestin-dependent inhibition and internalization of TRPV4. Activating TRPV4 with endogenous and synthetic agonists inhibited angiotensin II-mediated G-protein-associated second messenger accumulation, AT1R receptor phosphorylation, and ß-arrestin recruitment. We also noted that TRPV4 inhibits AT1R phosphorylation by activating the calcium-activated phosphatase calcineurin in a Ca2+/calmodulin-dependent manner, preventing ß-arrestin recruitment and receptor internalization. These findings suggest that when TRP channels and GPCRs are co-expressed in the same tissues, many of these channels can inhibit GPCR desensitization.


Assuntos
Receptores de Angiotensina/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Calmodulina/genética , Calmodulina/metabolismo , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Receptores de Angiotensina/genética , Canais de Cátion TRPV/genética , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
3.
Pharmacogenet Genomics ; 31(4): 75-82, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33395026

RESUMO

In pharmacogenomics, variable receptor phenotypes, resulting from genetic polymorphisms, are often described as a change in protein function or regulation observed upon exposure to a drug. However, in some instances, phenotypes are defined using a class of medications rather than individual drugs. This paradigm assumes that a variation associated with a drug response phenotype will retain the magnitude and direction of the effect for other drugs with the same mechanism of action. However, nonsynonymous polymorphisms may have ligand-specific effects. The purpose of this study was to investigate the potential for point mutations to asymmetrically affect the binding of different drugs to a common target. Ligand binding data from site-directed mutagenesis studies on five G-protein coupled receptors (beta-1 and -2 adrenergic, dopamine D2, angiotensin II and mu-opioid receptor) were collected and analyzed. Binding data from 81 studies for 253 ligands with 447 mutant proteins, including 10 naturally occurring human variants, were analyzed, yielding 1989 mutation-ligand pairs. Fold change in binding affinity for mutant proteins, relative to the wild-type, for different drugs was examined for ligand-specific effects, with a fold-change difference of one or more orders of magnitude between agents considered significant. Of the mutations examined, 49% were associated with ligand-specific effects. One human variant (T164I, beta-2 adrenergic receptor) showed ligand-specific effects for antiasthmatic agents. These results indicate that ligand-specific changes in binding are a possible consequence of missense mutations. This implies that caution needs to be exercised when grouping drugs together during design or interpretation of genotype-phenotype association studies.


Assuntos
Mutagênese Sítio-Dirigida , Testes Farmacogenômicos , Receptores Acoplados a Proteínas G/genética , Receptores Opioides mu/genética , Antagonistas de Receptores de Angiotensina/farmacologia , Estudos de Associação Genética , Humanos , Ligantes , Polimorfismo Genético/genética , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Receptores de Angiotensina/genética , Receptores de Dopamina D2/genética , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Mutação Silenciosa/genética
4.
Biochem Biophys Res Commun ; 529(2): 140-147, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32703402

RESUMO

Cerebral stroke is a leading global cause for mortality and disability. However, its pathogenesis is still unclear. Most tripartite motif (TRIM) family proteins, including TRIM62, have E3 ubiquitin ligase activities, and have multiple functions in regulating cellular processes. Nevertheless, the effects of TRIM62 on cerebral stroke still remain vague. Here, we reported that TRIM62 expression was markedly up-regulated in oxygen and glucose deprivation (OGD)-treated microglial cells. After cerebral ischemia, significantly elevated expression of TRIM62 was detected in peri-infarct area of wild type (WT) mice. The TRIM62 knockout (KO) mice exhibited alleviated apoptosis and neuroinflammation in the ischemic brain, eventually attenuating the stroke outcomes. Both in vitro and in vivo studies showed that nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome was dramatically activated in cerebral ischemia/reperfusion (I/R) conditions, while being ameliorated in TRIM62-KO mice, contributing to the suppression of neuroinflammatory response. Importantly, the in vitro experiments showed that OGD could induce the K63-ubiquitination of TRIM62 and the interaction between TRIM62 and NLRP3. In addition, adenovirus-regulated TRIM62 over-expression promoted the NLRP3 and nuclear factor κB (NF-κB) signaling, along with elevated interleukin-1ß (IL-1ß) and IL-18 transcriptional activities. Together, our results demonstrated that TRIM62 suppression was strongly protective in ischemic stroke through inhibiting NLRP3-regulated neuroinflammation.


Assuntos
Isquemia Encefálica/genética , Inflamação/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptores de Angiotensina/genética , Receptores de Endotelina/genética , Animais , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Técnicas de Inativação de Genes , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Angiotensina/metabolismo , Receptores de Endotelina/metabolismo , Ubiquitinação
5.
BMC Cardiovasc Disord ; 20(1): 99, 2020 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-32106816

RESUMO

BACKGROUND: Few studies examined the effect of long-acting nitrates on renal function in chronic heart failure (CHF). Thus, we aimed to investigate the effect of long-acting nitrate on the expression of adrenoceptors (AR) and angiotensin II receptor (ATR) subtypes of the renal cortex, in rats with myocardial infarction-induced CHF. METHODS: Rats were randomly divided into the following groups: control, sham-operated, CHF, low- and high-dose nitrate, positive drug control (olmesartan), and high-dose of long-acting nitrate + olmesartan. Ultrasound echocardiography markers were compared, and the levels of AR subtypes, AT1R, and AT2R were measured using reverse transcription-polymerase chain reaction and western blot analysis. Histopathology of the kidney was determined on hematoxylin and eosin-stained sections. RESULTS: CHF significantly increased plasma renin activity (PRA) and angiotensin II levels, upregulated AT1R expression and downregulated α1A-, ß1-, ß2-AR, and AT2R expression compared to the sham control. High-dose nitrate or olmesartan alone, and especially in combination, decreased the levels of PRA and angiotensin II and downregulated the CHF-induced expression of AT1R, α1A-, ß1-, and ß2-AR, and AT2R. CHF resulted in significant impairment of the renal tissue, including inflammatory cells infiltration to the tubular interstitium and surrounding the renal glomerulus, and tubular necrosis, which was alleviated in all treatment groups to different degrees. CONCLUSIONS: Long-acting nitrates could reverse CHF-induced changes in AR and ATR subtypes in the kidney, and improve cardiac function to protect renal function. Compared with monotherapy, the combination of nitrates and olmesartan shows more significant benefits in regulating AR and ATR subtypes.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Dinitrato de Isossorbida/análogos & derivados , Córtex Renal/efeitos dos fármacos , Infarto do Miocárdio/complicações , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/metabolismo , Receptores de Angiotensina/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Imidazóis/farmacologia , Dinitrato de Isossorbida/farmacologia , Córtex Renal/metabolismo , Córtex Renal/fisiopatologia , Masculino , Ratos Wistar , Receptores Adrenérgicos alfa 1/genética , Receptores Adrenérgicos beta/genética , Receptores de Angiotensina/genética , Sistema Renina-Angiotensina/efeitos dos fármacos , Tetrazóis/farmacologia , Fatores de Tempo
6.
Am J Physiol Heart Circ Physiol ; 316(1): H70-H79, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30289294

RESUMO

Sex differences in the presentation, outcome, and responses to treatment of systolic heart failure (HF) have been reported. In the present study, we examined the effect of sex on central neural mechanisms contributing to neurohumoral excitation and its peripheral manifestations in rats with HF. Male and female Sprague-Dawley rats underwent coronary artery ligation (CL) to induce HF. Age-matched rats served as controls. Ischemic zone and left ventricular function were similar 24 h and 4 wk after CL. Female rats with HF had a lower mortality rate and less hemodynamic compromise, pulmonary congestion, and right ventricular remodeling 4 wk after CL. Plasma angiotensin II (ANG II), arginine vasopressin (AVP), and norepinephrine levels were increased in HF rats in both sexes, but AVP and norepinephrine levels increased less in female rats. In the hypothalamic paraventricular nucleus, a key cardiovascular-related nucleus contributing to neurohumoral excitation in HF, mRNA levels for the proinflammatory cytokines tumor necrosis factor-α and interleukin-1ß as well as cyclooxygenase-2 and the ANG II type 1a receptor were increased in HF rats of both sexes, but less so in female rats. Angiotensin-converting enzyme 2 protein levels increased in female HF rats but decreased in male HF rats. mRNA levels of AVP were lower in female rats in both control and HF groups compared with the respective male groups. Activation of extracellular signal-regulated protein kinases 1 and 2 increased similarly in both sexes in HF. The results suggest that female HF rats have less central neural excitation and less associated hemodynamic compromise than male HF rats with the same degree of initial ischemic cardiac injury. NEW & NOTEWORTHY Sex differences in the presentation and responses to treatment of heart failure (HF) are widely recognized, but the underlying mechanisms are poorly understood. The present study describes sex differences in the central nervous system mechanisms that drive neurohumoral excitation in ischemia-induced HF. Female rats had a less intense central neurochemical response to HF and experienced less hemodynamic compromise. Sex hormones may contribute to these differences in the central and peripheral adaptations to HF.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Hemodinâmica , Hipotálamo/metabolismo , Isquemia Miocárdica/fisiopatologia , Animais , Arginina Vasopressina/sangue , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Feminino , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Masculino , Isquemia Miocárdica/complicações , Isquemia Miocárdica/metabolismo , Norepinefrina/sangue , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Fatores Sexuais , Função Ventricular
7.
Biochem Biophys Res Commun ; 519(4): 868-873, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31563322

RESUMO

There is growing evidence that renin-angiotensin system (RAS) components have been involved in the development of various types of cancers, including prostate cancer. This article for the first time reports the impact of Ang1-9 and Ang3-7 on viability and proliferation, migration and invasion of epithelial prostate cells. The results of this study clearly show that Ang1-9 and Ang3-7 exert different/opposite effects on in vitro biological properties of prostate cells. It appears that Ang1-9 has pro-cancer activities via the ability to induce cell divisions, enhance cell motility and stimulate the expression of such genes as vascular endothelial growth factor (VEGF), hypoxia-inducible factors (HIF-1), vimentin (VIM) and REL proto-oncogene, NF-kB subunit (REL). On the contrary, Ang3-7 did not show any mitogenic activity. Furthermore, this peptide hormone limited the migration of PNT1A cells probably by downregulation of VEGF and VIM expression. Finally, it is worth noting that both angiotensins have the ability to modulate gene expression for angiotensin receptors. Unfortunately, we could not unequivocally identify the type of angiotensin receptor responsible for signal transduction pathway involved in PNT1A cell survival and proliferation. Undoubtedly, further research and testing in this area are necessary.


Assuntos
Angiotensina I/farmacologia , Células Epiteliais/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Próstata/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Epiteliais/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Próstata/citologia , Próstata/metabolismo , Proto-Oncogene Mas , Receptores de Angiotensina/genética , Fator A de Crescimento do Endotélio Vascular/genética , Vimentina/genética
8.
Biol Reprod ; 100(1): 139-148, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30102356

RESUMO

Sex hormones contribute to sex differences in blood pressure. Inappropriate activation of the renin-angiotensin system is involved in vascular dysfunction and hypertension. This study evaluated the role of androgens (testosterone) in angiotensin II (Ang II)-induced increase in blood pressure, vascular reactivity, and cardiac hypertrophy. Eight-week-old male Wistar rats underwent sham operation, castration, or castration with testosterone replacement. After 12 weeks of chronic changes in androgen status, Ang II (120 ng/kg per minute) or saline was infused for 28 days via subcutaneous miniosmotic pump, and changes in blood pressure was measured. Vascular reactivity and Ang II receptor levels were examined in mesenteric arteries. Heart weight, cardiac ANP mRNA levels, and fibrosis were also assessed. Ang II infusion increased arterial pressure in intact males. The Ang II-induced increase in hypertensive response was prevented in castrated males. Testosterone replacement in castrated males restored Ang II-induced hypertensive responses. Castration reduced vascular AT1R/AT2R ratio, an effect that was reversed by testosterone replacement. Ang II-induced hypertension was associated with increased contractile response of mesenteric arteries to Ang II and phenylephrine in intact and testosterone-replaced castrated males; these increases were prevented in castrated males. Ang II infusion induced increased left ventricle-to-body weight ratio and ANP mRNA expression, indicators of left ventricular hypertrophy, and fibrosis in intact and testosterone-replaced castrated males, and castration prevented the increase in these parameters caused by Ang II. This study demonstrates that testosterone plays a permissive role in development and maintenance of Ang II-induced vascular dysfunction, hypertension, and cardiac hypertrophy.


Assuntos
Angiotensina II/farmacologia , Cardiomegalia/induzido quimicamente , Hipertensão/induzido quimicamente , Testosterona/fisiologia , Animais , Fator Natriurético Atrial/genética , Fator Natriurético Atrial/metabolismo , Cardiomegalia/sangue , Cardiomegalia/complicações , Cardiomegalia/fisiopatologia , Hipertensão/sangue , Hipertensão/complicações , Hipertensão/patologia , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , Orquiectomia , Ratos , Ratos Wistar , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Testosterona/sangue
9.
Biol Reprod ; 100(5): 1193-1203, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30596890

RESUMO

Maternal high-fat diet (HFD) during pregnancy is linked to cardiovascular diseases in postnatal life. The current study tested the hypothesis that maternal HFD causes myocardial changes through angiotensin II receptor (AGTR) expression modulation in fetal and neonatal rat hearts. The control group of pregnant rats was fed a normal diet and the treatment group of pregnant rats was on a HFD (60% kcal fat). Hearts were isolated from embryonic day 21 fetuses (E21) and postnatal day 7 pups (PD7). Maternal HFD decreased the body weight of the offspring in both E21 and PD7. The ratio of heart weight to body weight was increased in E21, but not PD7, when compared to the control group. Transmission electron microscopy revealed disorganized myofibrils and effacement of mitochondria cristae in the treatment group. Maternal HFD decreased S-phase and increased G1-phase of the cellular cycle for fetal and neonatal cardiac cells. Molecular markers of cardiac hypertrophy, such as Nppa and Myh7, were found to be increased in the treatment group. There was an associated increase in Agtr2 mRNA and protein, whereas Agtr1a mRNA and AGTR1 protein were decreased in HFD fetal and neonatal hearts. Furthermore, maternal HFD decreased glucocorticoid receptors (GRs) binding to glucocorticoid response elements at the Agtr1a and Agtr2 promoter, which correlated with downregulation of GR in fetal and neonatal hearts. These findings suggest that maternal HFD may promote premature termination of fetal and neonatal cardiomyocyte proliferation and compensatory hypertrophy through intrauterine modulation of AGTR1 and AGTR2 expression via GR dependent mechanism.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Fenômenos Fisiológicos da Nutrição Materna , Miocárdio/metabolismo , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Receptores de Angiotensina/genética , Animais , Animais Recém-Nascidos , Cardiomegalia/congênito , Cardiomegalia/embriologia , Cardiomegalia/genética , Cardiomegalia/metabolismo , Células Cultivadas , Gorduras na Dieta/farmacologia , Feminino , Feto/efeitos dos fármacos , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Coração/efeitos dos fármacos , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/patologia , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo
10.
J Nucl Cardiol ; 26(4): 1298-1308, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-29344922

RESUMO

BACKGROUND: Renin-angiotensin-aldosterone system (RAAS) has an important role in atherosclerosis. We investigated the effects of six RAAS gene polymorphisms on myocardial perfusion. METHODS AND RESULTS: We examined 810 patients with known or suspected coronary artery disease (CAD) using stress-rest myocardial single-photon emission computed tomography. Summed stress score (SSS), summed rest score (SRS), summed difference score (SDS), transient ischemic dilation (TID), and lung/heart ratio (LHR) were recorded. The following gene polymorphisms were investigated: angiotensin-converting enzyme (ACE) insertion/deletion (I/D), angiotensinogen (AGT) M235T and T174M, angiotensin II type 1 receptor (AT1R) A1166C, renin (REN) C5312T, and angiotensin II type 2 receptor (AT2R) C3123A. The heterozygotes or homozygotes on ACE D allele were 7.54 times more likely to have abnormal SSS, while the AGT (T174M) heterozygotes were 5.19 times more likely to have abnormal SSS. The homozygotes of ACE D had significantly higher values on TID and LHR, while the AGT (T174M) heterozygotes had higher values on TID. The AT1R heterozygotes had greater odds for having SSS ≥ 3. The patients carried AT1R homozygosity of C allele had significantly higher values on TID, while heterozygotes of AT1R had significantly higher values on LHR. CONCLUSIONS: Among the polymorphisms investigated, ACE D allele had the strongest association with abnormal myocardial perfusion.


Assuntos
Angiotensinogênio/genética , Doença da Artéria Coronariana/diagnóstico por imagem , Peptidil Dipeptidase A/genética , Polimorfismo Genético/genética , Receptores de Angiotensina/genética , Renina/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/fisiopatologia , Circulação Coronária , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Imagem de Perfusão do Miocárdio , Sistema Renina-Angiotensina , Tomografia Computadorizada de Emissão de Fóton Único
11.
J Mol Cell Cardiol ; 125: 117-128, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30193956

RESUMO

AIMS: Angiotension II (Ang II) plays a central role in the pathogenesis of renin-angiotensin-aldosterone system (RAAS)-induced heart failure. Mst1 exerts its function in cardiomyocytes subjected to pathological stimuli via inhibiting autophagy and aggravating apoptosis, but its role in RAAS-mediated cardiac injury is still unknown. Here, we aimed to determine whether cardiomyocyte-specific Mst1 knockout can alleviate Ang II-induced cardiac injury by improving cardiomyocyte autophagy and whether these functions depend on Ang II receptors. RESULTS: Mst1 knockout alleviated Ang II-induced heart failure, without affecting blood pressure and compensatory concentric hypertrophy. Mst1 specific knockout improved the effects of Ang II on cardiomyocyte autophagy, as evidenced by further increased LC3-II expression and decreased P62 expression. More typical autophagosomes accompanied by less damaged mitochondria were also observed by electron microscopy in Ang II-treated Mst1Δ/Δ mice. In vitro, Mst1 knockdown promoted cardiomyocyte autophagic flux, as demonstrated by more GFP-mRFP-LC3 puncta per cell. Increased LC3-II and decreased P62 expression both in the presence and absence of chloroquine were observed in Mst1 knockdown cardiomyocytes administered with Ang II. Treatment with 3-MA, an inhibitor of autophagy, abolished the beneficial effects of Mst1 knockout against Ang II-induced cardiac dysfunction. The compensatory effects of Ang II on upregulated autophagy were associated with Mst1 inhibition. Interestingly, the knockdown or antagonization of AT1R inhibited cardiomyocyte autophagy, which may represent a threat to cardiac function. Importantly, Mst1 knockout consistently enhanced cardiomyocyte autophagy following the knockdown or blocking of AT1R and AT2R. CONCLUSION: Cardiomyocyte-specific Mst1 knockout alleviates Ang II-induced cardiac injury by enhancing cardiomyocyte autophagy. Mst1 inhibition may counteract the undesirable effects of Ang II receptors blockage on cardiomyocyte autophagy and represent a promising complementary treatment strategy against Ang II-induced cardiac injury.


Assuntos
Angiotensina II/toxicidade , Cardiomiopatias/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Angiotensina/metabolismo , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Autofagia/fisiologia , Western Blotting , Cardiomiopatias/induzido quimicamente , Células Cultivadas , Fator de Crescimento de Hepatócito/genética , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Proteínas Proto-Oncogênicas/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Angiotensina/genética
12.
Am J Physiol Heart Circ Physiol ; 315(3): H590-H601, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29979625

RESUMO

In the present study, the change in secretion of atrial natriuretic peptide (ANP) from the atria was defined in hypertension accompanied by ventricular hypertrophy and increased synthesis of ANP. To identify the change of the secretion and mechanisms involved, experiments were performed in isolated perfused beating atria from sham-operated normotensive and renovascular hypertensive rats. Expression of ANP, natriuretic peptide receptor (NPR)-C, components of the renin-angiotensin system, and muscarinic signaling pathway was measured in cardiac tissues. Basal levels of ANP secretion and acetylcholine (ACh)- and stretch-induced activation of ANP secretion were suppressed in the atria from hypertensive compared with normotensive rats. ACh increased ANP secretion via M2 muscarinic ACh receptor-ACh-sensitive K+ channel signaling. In hypertensive rats, ANP concentration increased in the left ventricle but decreased in the right ventricle. The atrial concentration of ANP was not changed in hypertensive compared with normotensive rats. ANP mRNA expression was accentuated in the left ventricle but suppressed in the other cardiac chambers in the hearts of hypertensive rats. NPR-C expression was inversely related to ANP mRNA levels. Angiotensin II type 1 receptor (AT1R) expression was accentuated in the cardiac chambers from hypertensive rats compared with normotensive rats, whereas angiotensin II type 2 receptor, M2 muscarinic receptor, and Kir3.4 channels were suppressed. AT1R blockade with losartan reversed the change observed in hypertensive rats. The present findings indicate that renovascular hypertension shifts the major site of ANP secretion and synthesis from the atria to the left ventricle through modulation of the expression of ANP, NPR-C, AT1R, and the M2 muscarinic signaling pathway. NEW & NOTEWORTHY Renovascular hypertension suppresses the atrial secretion of ANP and shifts the major site of the regulation of ANP secretion and synthesis from atria to the hypertrophied left ventricle possibly via modulation of the expression of ANP, natriuretic peptide receptor-C, angiotensin II subtype 1 receptor, and M2 muscarinic signaling pathway.


Assuntos
Fator Natriurético Atrial/metabolismo , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Hipertensão Renovascular/metabolismo , Animais , Fator Natriurético Atrial/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/genética , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo
13.
Wiad Lek ; 71(1 pt 2): 148-154, 2018.
Artigo em Russo | MEDLINE | ID: mdl-29602923

RESUMO

OBJECTIVE: Introduction: Intraventricular hemorrhage (IVH) causes increased morbidity and mortality in premature infants and is associated with adverse neurological outcomes. The aim of the research is to confirm the role of genes of renin-angiotensin system in the development of adverse outcomes of treatment of severe IVC in preterm. PATIENTS AND METHODS: Materials and methods: A prospective study was conducted, which included 58 premature infants (average birth weight 1016.8 ± 52.59 g, gestational age 26.96 ±0.33 weeks), who were treated at medical institutions in Poltava region during 2012-2015. I/D polymorphism of ACE gene, A/C polymorphism of AGT2R1 gene and 4a/b polymorphism of the eNOS gene were studied. The distribution of the genotype was compared between groups using the χ2 analysis. RESULTS: Results: Out of 58 children enrolled in the study, 33 (56.9%) of infants developed ventricular dilatation, which in 8 (13.8%) of the newborns progressed to hydrocephalus. Thirty-four (58.6%) infants with severe IVHs died. The average (median) time of death of infants was 10.6 days. Among children who died, a combination of genotypes ID & DD of the gene ACE +4ab & 4AA of the eNOS gene was found significantly more often than in children who survived, and at the border of statistical significance - the isolated genotype 4ab & 4 aa of the eNOS gene. The combination of genotype ID+DD of the ACE gene and the AC + CC genotype of the AGTR1 gene was significantly more frequent among children with ventricular dilatation than in children without it. The weight at birth have negative correlation trends with lethal outcomes and do not have with ventricular dilatation and hydrocephalus. CONCLUSION: Conclusions: Lethal outcomes in children with IVH are associated with weight at birth, as well as the presence of a combination of genotypes ID+DD of the ACE gene and 4ab+4aa of the eNOS gene.


Assuntos
Hemorragia Cerebral/terapia , Polimorfismo Genético , Sistema Renina-Angiotensina/genética , Hemorragia Cerebral/complicações , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/mortalidade , Ventrículos Cerebrais , Feminino , Humanos , Hidrocefalia/etiologia , Recém-Nascido Prematuro , Masculino , Óxido Nítrico Sintase Tipo III/genética , Peptidil Dipeptidase A/genética , Estudos Prospectivos , Receptores de Angiotensina/genética , Falha de Tratamento
14.
J Biol Chem ; 291(8): 3967-81, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26703465

RESUMO

Angiotensin II (Ang II) is a vasopressive hormone but is also a potent activator of cellular migration. We have previously shown that it can promote the activation of the GTPase ARF6 in a heterologous overexpressing system. The molecular mechanisms by which receptors control the activation of this small G protein remain, however, largely unknown. Furthermore, how ARF6 coordinates the activation of complex cellular responses needs to be further elucidated. In this study, we demonstrate that Ang II receptors engage ß-arrestin, but not Gq, to mediate ARF6 activation in HEK 293 cells. To further confirm the key role of ß-arrestin proteins, we overexpressed ß-arrestin2-(1-320), a dominant negative mutant known to block receptor endocytosis. We show that expression of this truncated construct does not support the activation of the GTPase nor cell migration. Interestingly, ß-arrestin2 can interact with the ARF guanine nucleotide exchange factor ARNO, although the C-terminally lacking mutant does not. We finally examined whether receptor endocytosis controlled ARF6 activation and cell migration. Although the clathrin inhibitor PitStop2 did not impact the ability of Ang II to activate ARF6, cell migration was markedly impaired. To further show that ARF activation regulates key signaling events leading to migration, we also examined MAPK activation. We demonstrate that this signaling axis is relevant in smooth muscle cells of the vasculature. Altogether, our findings show for the first time that Ang II receptor signaling to ß-arrestin regulates ARF6 activation. These proteins together control receptor endocytosis and ultimately cell migration.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Angiotensina II/metabolismo , Arrestinas/metabolismo , Movimento Celular/fisiologia , Endocitose/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Angiotensina II/genética , Animais , Arrestinas/genética , Movimento Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Ratos , Ratos Wistar , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Sulfonamidas/farmacologia , Tiazolidinas/farmacologia , beta-Arrestinas
15.
Am J Physiol Renal Physiol ; 313(2): F326-F338, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28515172

RESUMO

Kidney transplantation (KTX) is a life-saving procedure for patients with end-stage renal disease. Expression levels of many genes in the kidney vary between males and females, which may play an essential role in the sex differences in graft function. However, whether these differences are affected after cross-sex-KTX is unknown. In the present study, we assessed postoperative changes in genotype, function, and inflammatory responses of the grafts in same-sex- and cross-sex-KTX. Single kidney transplants were performed between same and different sex C57BL/6 mice paired into four combination groups: female donor/female recipient (F/F); male donor/male recipient (M/M); female donor/male recipient (F/M); and male donor/female recipient (M/F). The remnant native kidney was removed 4 days posttransplant. Expression levels of genes related to the contractility of the afferent arteriole and tubular sodium reabsorption were assessed. Same-sex-KTX did not significantly alter the magnitude or sex difference pattern of gene expression in male or female grafts. Cross-sex-KTX showed an attenuated sex difference in gene expressions. The measurements of endothelin 1, endothelin ETA receptor, Na+-K--2Cl cotransporter 2 (NKCC2), and epithelial Na+ channels (ENaC) subunits exhibited decreases in M/F compared with M/M and increases in F/M compared with F/F. There were no significant differences in hemodynamics or sodium excretion in response to acute volume expansion for any sex combinations. Cross-sex-KTX stimulated more robust inflammatory responses than same-sex-KTX. IL-6 and KC mRNA levels elevated 5- to 20-fold in cross-sex-KTX compared with same-sex-KTX. In conclusion, cross-sex-KTX alters gene expression levels and induces inflammatory responses, which might play an important role in long-term graft function.


Assuntos
Regulação da Expressão Gênica , Transplante de Rim/efeitos adversos , Rim/metabolismo , Rim/cirurgia , Nefrite/genética , Animais , Endotelina-1/genética , Endotelina-1/metabolismo , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Feminino , Interação Gene-Ambiente , Genótipo , Hemodinâmica , Mediadores da Inflamação/metabolismo , Rim/irrigação sanguínea , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais , Nefrite/metabolismo , Nefrite/patologia , Nefrite/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Receptores de Endotelina/genética , Receptores de Endotelina/metabolismo , Circulação Renal , Eliminação Renal , Fatores Sexuais , Sódio/metabolismo , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/genética , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/genética , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Fatores de Tempo
16.
BMC Nephrol ; 18(1): 183, 2017 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-28569136

RESUMO

BACKGROUND: Septic acute kidney injury affects 40-50% of all septic patients. Molecular differences between septic patients with and without acute kidney injury (AKI) are only poorly understood. Here, we investigated gene expression changes that differentiated the subjects who developed septic AKI from those who did not and coupled this approach with traditional parameters of renal physiology. METHODS: In 15 anesthetized, mechanically ventilated and instrumented pigs, progressive sepsis was induced either by peritonitis or by continuous intravenous infusion of Pseudomonas aeruginosa. Animals received standard intensive care including goal-directed hemodynamic management. Analyses were performed on kidneys from sham operated animals, septic pigs without AKI, and pigs with septic AKI. Before, and at 12, 18 and 22 h of progressive sepsis, systemic and renal hemodynamics, cortex microcirculation and plasma IL-6 and TNF-α were measured. At 22 h whole kidney expression of pre-selected genes was analyzed by quantitative Real Time PCR. RESULTS: Animals with septic AKI had systemic hemodynamic phenotype (normo- or hyperdynamic) comparable with non-AKI subjects, but demonstrated higher plasma levels of cytokines, an increase in renal vascular resistance and early fall in cortical microcirculatory blood flow. The genes whose expression discriminated septic AKI from non-AKI included Toll like receptor 4 (up-regulated 2.7-fold, P = 0.04); Cyclooxygenase-2 (up-regulated 14.6-fold, P = 0.01), Angiotensin II Receptor (up-regulated 8.1-fold, P = 0.01), Caspase 3 (up-regulated 5.1-fold, P = 0.02), Peroxisome Proliferator-Activated Receptor Gamma, Coactivator 1 Alpha (down-regulated 2-fold, P = 0.02). CONCLUSIONS: In this preliminary experimental study, kidney gene expression was profoundly different in animals that developed septic AKI as opposed to septic animals that did not. The biological functions of the genes differentially expressed support a role of inflammatory overstimulation coupled with metabolic and apoptotic molecular responses in early septic AKI. Cyclooxygenase-2 and angiotensin type 2 receptor-dependent downstream mechanisms appear fruitful targets for future mechanistic research.


Assuntos
Injúria Renal Aguda/genética , Expressão Gênica , Sepse/complicações , Injúria Renal Aguda/microbiologia , Injúria Renal Aguda/fisiopatologia , Animais , Caspase 3/genética , Ciclo-Oxigenase 2/genética , Modelos Animais de Doenças , Predisposição Genética para Doença , Hemodinâmica , Interleucina-6/sangue , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Receptores de Angiotensina/genética , Sepse/fisiopatologia , Suínos , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/sangue
17.
Int J Mol Sci ; 18(9)2017 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-28926934

RESUMO

Recent studies suggest that allicin may play a role in chronic kidney disease (CKD), reducing hypertension and oxidative stress and improving renal dysfunction. In the present study, CKD was induced by 5/6 nephrectomy and the animals were divided into four treatment groups as follows: control (C), CKD, CKD+allicin (40 mg/kg pathway oral) (CKDA), and CKD+Losartan (20 mg/kg) (CKDL). After CKD induction, the rats developed hypertension from week 3 to the end of the study. This was associated with increased creatinine and blood urea nitrogen (BUN) levels in serum, increased albuminuria, increased urinary excretion of N-acetyl-ß-d-glucosaminidase (NAG), increased nephrin expression, and incrased histological alterations in the cortex. The levels of angiotensin receptors and endothelial nitric oxide synthase (eNOS) were decreased in the renal cortex from the CKD group. Otherwise, lipid and protein oxidation were higher in the CKD group than in the control group. A disturbance was observed in the expression levels of the nuclear factor erythroid 2-related factor 2/Kelch ECH associating protein 1 system (Nrf2/keap1) and the antioxidant enzymes catalase, superoxide dismutase, and heme oxygenase-1. Allicin or losartan treatments relieved renal dysfunction, hypertension, and oxidative stress. In addition, both treatments showed the same efficacy on the expression of angiotensin receptors, the nephrin, Nrf2/keap1 pathway, and eNOS. Further in silico analyses suggest that allicin and losartan could have a common mechanism involving interaction with AT1 receptors. Allicin showed antihypertensive, antioxidant, and nephroprotective effects. The beneficial effects showed by allicin are similar, or even better, than those of losartan. In fact, the effect of allicin on blood pressure and renal function is comparable to reductions seen with losartan, a prescription drug commonly used as a first-line therapy.


Assuntos
Anti-Hipertensivos/uso terapêutico , Antioxidantes/uso terapêutico , Losartan/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Ácidos Sulfínicos/uso terapêutico , Acetilglucosaminidase/urina , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/efeitos adversos , Antioxidantes/administração & dosagem , Antioxidantes/efeitos adversos , Creatinina/sangue , Dissulfetos , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Rim/metabolismo , Losartan/administração & dosagem , Losartan/efeitos adversos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , Ratos , Ratos Wistar , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Ácidos Sulfínicos/administração & dosagem , Ácidos Sulfínicos/efeitos adversos , Ureia/sangue
18.
J Physiol ; 594(5): 1327-40, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26574162

RESUMO

A functioning placental renin-angiotensin system (RAS) appears necessary for uncomplicated pregnancy and is present during placentation, which occurs under low oxygen tensions. Placental RAS is increased in pre-eclampsia (PE), characterised by placental dysfunction and elevated oxidative stress. We investigated the effect of high altitude hypoxia on the RAS and hypoxia-inducible factors (HIFs) by measuring mRNA and protein expression in term placentae from normotensive (NT) and PE women who delivered at sea level or above 3100 m, using an explant model of hypoxia-reoxygenation to assess the impact of acute oxidative stress on the RAS and HIFs. Protein levels of prorenin (P = 0.049), prorenin receptor (PRR; P = 0.0004), and angiotensin type 1 receptor (AT1R, P = 0.006) and type 2 receptor (AT2R, P = 0.002) were all significantly higher in placentae from NT women at altitude, despite mRNA expression being unaffected. However, mRNA expression of all RAS components was significantly lower in PE at altitude than at sea level, yet PRR, angiotensinogen (AGT) and AT1R proteins were all increased. The increase in transcript and protein expression of all the HIFs and NADPH oxidase 4 seen in PE compared to NT at sea level was blunted at high altitude. Experimentally induced oxidative stress stimulated AGT mRNA (P = 0.04) and protein (P = 0.025). AT1R (r = 0.77, P < 0.001) and AT2R (r = 0.81, P < 0.001) mRNA both significantly correlated with HIF-1ß, whilst AT2R also correlated with HIF-1α (r = 0.512, P < 0.013). Our observations suggest that the placental RAS is responsive to changes in tissue oxygenation: this could be important in the interplay between reactive oxygen species as cell-signalling molecules for angiogenesis and hence placental development and function.


Assuntos
Altitude , Hipóxia Fetal/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Sistema Renina-Angiotensina , Angiotensinogênio/sangue , Estudos de Casos e Controles , Feminino , Humanos , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Estresse Oxidativo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Renina/genética , Renina/metabolismo
19.
Am J Physiol Heart Circ Physiol ; 310(11): H1834-41, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27199113

RESUMO

Preeclampsia (PE) is a severe complication of pregnancy associated with maternal and fetal morbidity and mortality. The underlying pathophysiology involves maternal systemic vascular and endothelial dysfunction associated with circulating antiangiogenic factors, although the specific etiology of the disease remains elusive. Our aim was to investigate the maternal endothelium in PE by exploring the expression of genes involved with endothelial function in a novel platform of maternal primary endothelial cells. Adipose tissue was sampled at the time of caesarean section from both normal and preeclamptic patients. Maternal microvascular endothelial cells were isolated by tissue digestion and CD31 magnetic Dynabeads. Cell purity was confirmed by immunofluorescence microscopy and flow cytometry. Western analyses revealed VEGF activation of VEGF receptor 2 (VEGFR2) and ERK in primary cells. Quantitative PCR analyses revealed significantly altered mRNA levels of various genes involved with angiogenesis and blood pressure control in preeclamptic cells, including soluble fms-like tyrosine kinase-1, endoglin, VEGFR2, angiotensin receptor 1, and endothelin compared with cells isolated from normal pregnancies. Overall, maternal endothelial cells from preeclamptic patients exhibit extensive alteration of expression of factors associated with antiangiogenic and vasoconstrictive phenotypes, shedding light on the underlying mechanisms associated with the vascular dysfunction characteristic of PE.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Pré-Eclâmpsia/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adulto , Endoglina/genética , Endoglina/metabolismo , Feminino , Expressão Gênica , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Pré-Eclâmpsia/genética , Gravidez , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
20.
World J Urol ; 34(1): 89-95, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25981400

RESUMO

INTRODUCTION AND OBJECTIVES: Reactive oxygen species (ROS) are produced during the interaction between oxalate/calcium oxalate monohydrate (COM) crystals and renal epithelial cells and are responsible for the various cellular responses through the activation of NADPH oxidase (Nox). Ox and COM also activate the renin-angiotensin-aldosterone system (RAAS). Aldosterone stimulates ROS production through activation of Nox with the involvement of mineralocorticoid receptor (MR), Rac1 and mitogen-activated protein kinases (MAPK). We investigated RAAS pathways in vivo in an animal model of hyperoxaluria and in vitro by exposing renal epithelial cells to COM crystals. METHODS: Hyperoxaluria was induced in male SD rats by administering ethylene glycol. One group of rats was additionally given spironolactone. Total RNA was extracted and subjected to genomic microarrays to obtain global transcriptome data. Normal rat kidney cell line (NRK-52E) was incubated with aldosterone(10(-7) M) and COM(67 µg/cm(2)) with or without spironolactone(10(-5) M), a selective inhibitor of SRC family of kinases; protein phosphatase 2(pp2) (10(-5) M) and Nox inhibitor; diphenylene iodonium (DPI) (10(-5) M). RESULTS: Relative expression of genes encoding for AGT, angiotensin receptors 1b and 2, Renin 1, Cyp11b, HSD11B2, Nr3c2, NOx4 and Rac1 was upregulated in the kidneys of rats with hyperoxaluria. Treatment with spironolactone reversed the effect of hyperoxaluria. Both aldosterone and COM crystals activated Nox and Rac1 expression in NRK52E, while spironolactone inhibited Nox and Rac1 expression. Increased Rac1 expression was significantly attenuated by treatment with PP2 and spironolactone. CONCLUSIONS: Results indicate that hyperoxaluria-induced production of ROS, injury and inflammation are in part associated with the activation of Nox through renin-angiotensin-aldosterone pathway.


Assuntos
Oxalato de Cálcio/metabolismo , Hiperoxalúria/genética , NADPH Oxidases/metabolismo , RNA Mensageiro/metabolismo , Sistema Renina-Angiotensina/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/efeitos dos fármacos , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Angiotensinogênio/efeitos dos fármacos , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Linhagem Celular , Citocromo P-450 CYP11B2/efeitos dos fármacos , Citocromo P-450 CYP11B2/genética , Citocromo P-450 CYP11B2/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Etilenoglicol/toxicidade , Perfilação da Expressão Gênica , Hiperoxalúria/induzido quimicamente , Hiperoxalúria/metabolismo , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacologia , NADPH Oxidase 4 , NADPH Oxidases/efeitos dos fármacos , NADPH Oxidases/genética , Oniocompostos/farmacologia , Proteína Fosfatase 2/farmacologia , RNA Mensageiro/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/efeitos dos fármacos , Receptores de Angiotensina/genética , Receptores de Angiotensina/metabolismo , Receptores de Mineralocorticoides/efeitos dos fármacos , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Renina/efeitos dos fármacos , Renina/genética , Renina/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Espironolactona/farmacologia , Esteroide 11-beta-Hidroxilase/efeitos dos fármacos , Esteroide 11-beta-Hidroxilase/genética , Esteroide 11-beta-Hidroxilase/metabolismo , Proteínas rac1 de Ligação ao GTP/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA