Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Mol Psychiatry ; 28(7): 2878-2893, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36316366

RESUMO

Coronavirus disease-2019 (COVID-19) is primarily a respiratory disease, however, an increasing number of reports indicate that SARS-CoV-2 infection can also cause severe neurological manifestations, including precipitating cases of probable Parkinson's disease. As microglial NLRP3 inflammasome activation is a major driver of neurodegeneration, here we interrogated whether SARS-CoV-2 can promote microglial NLRP3 inflammasome activation. Using SARS-CoV-2 infection of transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) as a COVID-19 pre-clinical model, we established the presence of virus in the brain together with microglial activation and NLRP3 inflammasome upregulation in comparison to uninfected mice. Next, utilising a model of human monocyte-derived microglia, we identified that SARS-CoV-2 isolates can bind and enter human microglia in the absence of viral replication. This interaction of virus and microglia directly induced robust inflammasome activation, even in the absence of another priming signal. Mechanistically, we demonstrated that purified SARS-CoV-2 spike glycoprotein activated the NLRP3 inflammasome in LPS-primed microglia, in a ACE2-dependent manner. Spike protein also could prime the inflammasome in microglia through NF-κB signalling, allowing for activation through either ATP, nigericin or α-synuclein. Notably, SARS-CoV-2 and spike protein-mediated microglial inflammasome activation was significantly enhanced in the presence of α-synuclein fibrils and was entirely ablated by NLRP3-inhibition. Finally, we demonstrate SARS-CoV-2 infected hACE2 mice treated orally post-infection with the NLRP3 inhibitory drug MCC950, have significantly reduced microglial inflammasome activation, and increased survival in comparison with untreated SARS-CoV-2 infected mice. These results support a possible mechanism of microglial innate immune activation by SARS-CoV-2, which could explain the increased vulnerability to developing neurological symptoms akin to Parkinson's disease in COVID-19 infected individuals, and a potential therapeutic avenue for intervention.


Assuntos
COVID-19 , Doença de Parkinson , Humanos , Camundongos , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Microglia/metabolismo , alfa-Sinucleína/metabolismo , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo , COVID-19/metabolismo , Camundongos Transgênicos
2.
Clin Sci (Lond) ; 134(20): 2755-2769, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-33034619

RESUMO

Heart failure (HF) is associated with impaired L-arginine transport. In the present study, we tested the hypothesis that augmented L-arginine transport prevents the loss of kidney function in HF. Renal function was assessed in wildtype mice (WT), transgenic mice with HF (dilated cardiomyopathy, DCM) and double transgenic mice (double transgenic mice with DCM and CAT-1 overexpression, HFCAT-1) with HF and endothelial-specific overexpression of the predominant L-arginine transporter, cationic amino acid transporter-1 (CAT-1) (n=4-8/group). Cardiac function was assessed via echocardiography and left ventricular catheterisation. Renal function was assessed via quantification of albuminuria and creatinine clearance. Plasma nitrate and nitrite levels together with renal fibrosis and inflammatory markers were also quantified at study end. Albumin/creatinine ratio was two-fold greater in DCM mice than in WT mice (P=0.002), and tubulointerstitial and glomerular fibrosis were approximately eight- and three-fold greater, respectively, in DCM mice than in WT mice (P≤0.02). Critically, urinary albumin/creatinine ratio and tubulointerstitial and glomerular fibrosis were less in HFCAT-1 mice than in DCM mice (P<0.05). Renal CAT-1 expression and plasma nitrate and nitrite levels were less in DCM mice compared with WT (P≤0.03) but was greater in HFCAT-1 mice than in DCM mice (P≤0.009). Renal expression of IL-10 was less in DCM mice compared with WT (P<0.001) but was greater in HFCAT-1 mice compared with DCM mice (P=0.02). Our data provide direct evidence that augmented L-arginine transport prevents renal fibrosis, inflammation and loss of kidney function in HF.


Assuntos
Transportador 1 de Aminoácidos Catiônicos/metabolismo , Células Endoteliais/metabolismo , Insuficiência Cardíaca/fisiopatologia , Testes de Função Renal , Rim/fisiopatologia , Animais , Pressão Sanguínea , Peso Corporal , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Transportador 1 de Aminoácidos Catiônicos/genética , Fibrose , Regulação da Expressão Gênica , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/genética , Inflamação/genética , Inflamação/patologia , Rim/imunologia , Rim/patologia , Masculino , Camundongos Transgênicos , Miocárdio/patologia , Nitratos/sangue , Nitritos/sangue , Tamanho do Órgão , Especificidade de Órgãos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Clin Sci (Lond) ; 133(20): 2061-2067, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31654065

RESUMO

The precise mechanisms underlying resistant hypertension remain elusive. Reduced nitric oxide (NO) bioavailability is frequently documented in chronic kidney disease, obesity, diabetes and advanced age, all of which are risk factors for resistant hypertension. Sympathetic overactivity and chronic activation of the renin-angiotensin system are salient features of resistant hypertension. Interestingly, recent data indicate that renal sympathetic overactivity can reduce the expression of neuronal nitric oxide synthase in the paraventricular nucleus. Reduced NO levels in the paraventricular nucleus can increase sympathetic outflow and this can create a vicious cycle contributing to resistant hypertension. Angiotensin II can reduce l-arginine transport and hence NO production. Reduced NO levels may reduce the formation of angiotensin 1-7 dampening the cardio-protective effects of the renin-angiotensin system contributing to resistant hypertension. In addition, interleukin-6 (IL-6) is demonstrated to be independently associated with resistant hypertension, and IL-6 can reduce NO synthesis. Despite this, NO levels have not been quantified in resistant hypertension. Findings from a small proof of concept study indicate that NO donors can reduce blood pressure in patients with resistant hypertension but more studies are required to validate these preliminary findings. In the present paper, we put forward the hypothesis that reduced NO bioavailability contributes substantially to the development of resistant hypertension.


Assuntos
Arginina/fisiologia , Hipertensão/fisiopatologia , Óxido Nítrico/fisiologia , Disponibilidade Biológica , Endotélio Vascular/fisiopatologia , Humanos , Hipertensão/etiologia , Hipertensão/terapia , Inflamação/complicações , Óxido Nítrico/deficiência , Óxido Nítrico/farmacocinética , Sistema Renina-Angiotensina/fisiologia , Transdução de Sinais/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Falha de Tratamento , Rigidez Vascular/fisiologia
4.
Circulation ; 135(10): 964-977, 2017 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-27927713

RESUMO

BACKGROUND: Dietary intake of fruit and vegetables is associated with lower incidence of hypertension, but the mechanisms involved have not been elucidated. Here, we evaluated the effect of a high-fiber diet and supplementation with the short-chain fatty acid acetate on the gut microbiota and the prevention of cardiovascular disease. METHODS: Gut microbiome, cardiorenal structure/function, and blood pressure were examined in sham and mineralocorticoid excess-treated mice with a control diet, high-fiber diet, or acetate supplementation. We also determined the renal and cardiac transcriptome of mice treated with the different diets. RESULTS: We found that high consumption of fiber modified the gut microbiota populations and increased the abundance of acetate-producing bacteria independently of mineralocorticoid excess. Both fiber and acetate decreased gut dysbiosis, measured by the ratio of Firmicutes to Bacteroidetes, and increased the prevalence of Bacteroides acidifaciens. Compared with mineralocorticoid-excess mice fed a control diet, both high-fiber diet and acetate supplementation significantly reduced systolic and diastolic blood pressures, cardiac fibrosis, and left ventricular hypertrophy. Acetate had similar effects and markedly reduced renal fibrosis. Transcriptome analyses showed that the protective effects of high fiber and acetate were accompanied by the downregulation of cardiac and renal Egr1, a master cardiovascular regulator involved in cardiac hypertrophy, cardiorenal fibrosis, and inflammation. We also observed the upregulation of a network of genes involved in circadian rhythm in both tissues and downregulation of the renin-angiotensin system in the kidney and mitogen-activated protein kinase signaling in the heart. CONCLUSIONS: A diet high in fiber led to changes in the gut microbiota that played a protective role in the development of cardiovascular disease. The favorable effects of fiber may be explained by the generation and distribution of one of the main metabolites of the gut microbiota, the short-chain fatty acid acetate. Acetate effected several molecular changes associated with improved cardiovascular health and function.


Assuntos
Acetato de Desoxicorticosterona/farmacologia , Fibras na Dieta/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Hipertensão/prevenção & controle , Animais , Bactérias/genética , Bactérias/isolamento & purificação , Pressão Sanguínea/efeitos dos fármacos , Acetato de Desoxicorticosterona/uso terapêutico , Fibras na Dieta/uso terapêutico , Suplementos Nutricionais , Modelos Animais de Doenças , Fibrose , Trato Gastrointestinal/microbiologia , Hipertensão/patologia , Hipertensão/veterinária , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Miocárdio/patologia , Tamanho do Órgão/efeitos dos fármacos , Análise de Componente Principal , RNA Ribossômico 16S/química , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Transcriptoma/efeitos dos fármacos
5.
Exp Physiol ; 103(12): 1593-1602, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30311699

RESUMO

NEW FINDINGS: What is the central question of this study? The aim was to determine the renoprotective effects of serelaxin in the setting of chronic heart failure. What are the main findings and its importance? Our data indicate that serelaxin can reduce renal fibrosis and inflammation in experimental heart failure. Currently, there are no effective treatments to rescue renal function in heart failure patients, and our data suggest that serelaxin might have the potential to reduce renal fibrosis and inflammation in heart failure. ABSTRACT: Serelaxin has been demonstrated to attenuate renal fibrosis and inflammation in cardiorenal disease. In the present study, we tested the hypothesis that serelaxin can prevent the decline in renal function in dilated cardiomyopathy (DCM) by targeting renal fibrosis and inflammation. Male transgenic mice with DCM (n = 16) and their wild-type littermates (WT; n = 20) were administered either vehicle or serelaxin (500 µg kg-1  day-1 ; subcutaneous minipumps; 8 weeks). Cardiac function was assessed via echocardiography before and during the eighth week of serelaxin treatment. Renal function and inflammation as well as cardiac and renal fibrosis were assessed at the end of the study. Serelaxin had minimal effect on cardiac function (P ≥ 0.99). Tubulointerstitial and glomerular fibrosis were ∼3-fold greater in vehicle-treated DCM mice compared with vehicle-treated WT mice (P ≤ 0.001). Renal mRNA expression of Tnfα and Il1α were ∼4- and ∼3-fold greater, respectively, in vehicle-treated DCM mice compared with vehicle-treated WT mice (P ≤ 0.05). Tubulointerstitial and glomerular fibrosis were 46 and 45% less, respectively, in serelaxin-treated DCM mice than in vehicle-treated DCM mice (P ≤ 0.01). Renal cortical mRNA expression of Tnfα and Il1α were 56 and 58% less, respectively, in the former group compared with the latter (P ≤ 0.05). The urinary albumin:creatinine ratio was ∼3-fold greater in vehicle-treated DCM mice compared with vehicle-treated WT mice (P = 0.02). The urinary albumin:creatinine ratio was not significantly different between vehicle-treated DCM mice and serelaxin-treated DCM mice (P = 0.38). These data suggest that serelaxin can attenuate renal fibrosis and inflammation and has the potential to exert renoprotective effects in DCM.


Assuntos
Anti-Inflamatórios/farmacologia , Síndrome Cardiorrenal/tratamento farmacológico , Cardiomiopatia Dilatada/tratamento farmacológico , Insuficiência Cardíaca/tratamento farmacológico , Rim/efeitos dos fármacos , Nefrite/prevenção & controle , Relaxina/farmacologia , Animais , Síndrome Cardiorrenal/patologia , Síndrome Cardiorrenal/fisiopatologia , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/fisiopatologia , Colágeno/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Fibrose , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Masculino , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Nefrite/genética , Nefrite/metabolismo , Nefrite/fisiopatologia , Óxido Nítrico/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
6.
Heart Lung Circ ; 25(8): 874-80, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27132623

RESUMO

Renal dysfunction and heart failure commonly co-exist; it is termed the cardiorenal syndrome (CRS). This combination of renal and cardiac impairment presents a substantial clinical challenge and is associated with adverse prognosis. The pathogenesis of the CRS is complex, including chronic activation of the renin-angiotensin-aldosterone system (RAAS) and the sympathetic nervous system, together with reduced renal perfusion. Chronic activation of the RAAS can impair mitochondrial function, and increase mitochondrial derived oxidative stress which in turn can lead to renal injury and sodium and water retention. For example, it has been shown that exogenous Ang II augments renal mitochondrial oxidative stress, reduces GFR and induces albuminuria in rats with heart failure. Administration of Ang II also augmented renal mitochondrial dysfunction in aged mice. Current treatments for CRS, including angiotensin-converting enzyme inhibitors, exert limited renal protection if any at all. Therefore, novel treatments particularly those that can target renal mechanisms downstream to chronic activation of the renal renin-angiotensin system are likely to exert renoprotection in the setting of CRS.


Assuntos
Síndrome Cardiorrenal/metabolismo , Rim/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Sistema Renina-Angiotensina , Equilíbrio Hidroeletrolítico , Animais , Síndrome Cardiorrenal/patologia , Síndrome Cardiorrenal/fisiopatologia , Taxa de Filtração Glomerular , Humanos , Rim/patologia , Rim/fisiopatologia , Camundongos , Mitocôndrias/patologia , Ratos
7.
Exp Physiol ; 100(7): 796-804, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25958845

RESUMO

NEW FINDINGS: What is the central question of this study? What is the potential role of endothelial NO production via overexpression of the l-arginine transporter, CAT1, as a mitigator of cardiac hypertrophy? What is the main finding and its importance? Augmentation of endothelium-specific l-arginine transport via CAT1 can attenuate pressure-overload-dependent cardiac hypertrophy and fibrosis. Our findings support the conclusion that interventions that improve endothelial l-arginine transport may provide therapeutic utility in the setting of myocardial hypertrophy. Such modifications may be introduced by exercise training or locally delivered gene therapy, but further experimental and clinical studies are required. Endothelial dysfunction has been postulated to play a central role in the development of cardiac hypertrophy, probably as a result of reduced NO bioavailability. We tested the hypothesis that increased endothelial NO production, mediated by increased l-arginine transport, could attenuate pressure-overload-induced cardiac hypertrophy. Echocardiography and blood pressure measurements were performed 15 weeks after transverse aortic constriction (TAC) in wild-type (WT) mice (n = 12) and in mice with endothelium-specific overexpression of the l-arginine transporter, CAT1 (CAT+; n = 12). Transverse aortic constriction induced greater increases in heart weight to body weight ratio in WT (by 47%) than CAT+ mice (by 25%) compared with the respective controls (P ≤ 0.05). Likewise, the increase in left ventricular wall thickness induced by TAC was significantly attenuated in CAT+ mice (P = 0.05). Cardiac collagen type I mRNA expression was greater in WT mice with TAC (by 22%; P = 0.03), but not in CAT+ mice with TAC, compared with the respective controls. Transverse aortic constriction also induced lesser increases in ß-myosin heavy chain mRNA expression in CAT+ mice compared with WT (P ≤ 0.05). Left ventricular systolic pressure after TAC was 36 and 39% greater in WT and CAT+ mice, respectively, compared with the respective controls (P ≤ 0.001). Transverse aortic constriction had little effect on left ventricular end-diastolic pressure in both genotypes. Taken together, these data indicate that augmenting endothelial function by overexpression of l-arginine transport can attenuate pressure-overload-induced cardiac hypertrophy.


Assuntos
Cardiomegalia/fisiopatologia , Miocárdio/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Animais , Arginina/metabolismo , Cardiomegalia/genética , Cardiomegalia/patologia , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Hipertrofia Ventricular Esquerda/genética , Hipertrofia Ventricular Esquerda/metabolismo , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Pressão , Disfunção Ventricular Esquerda/genética
8.
Am J Physiol Renal Physiol ; 306(9): F1026-38, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24598805

RESUMO

Tissue hypoxia has been demonstrated, in both the renal cortex and medulla, during the acute phase of reperfusion after ischemia induced by occlusion of the aorta upstream from the kidney. However, there are also recent clinical observations indicating relatively well preserved oxygenation in the nonfunctional transplanted kidney. To test whether severe acute kidney injury can occur in the absence of widespread renal tissue hypoxia, we measured cortical and inner medullary tissue Po2 as well as total renal O2 delivery (Do2) and O2 consumption (Vo2) during the first 2 h of reperfusion after 60 min of occlusion of the renal artery in anesthetized rats. To perform this experiment, we used a new method for measuring kidney Do2 and Vo2 that relies on implantation of fluorescence optodes in the femoral artery and renal vein. We were unable to detect reductions in renal cortical or inner medullary tissue Po2 during reperfusion after ischemia localized to the kidney. This is likely explained by the observation that Vo2 (-57%) was reduced by at least as much as Do2 (-45%), due to a large reduction in glomerular filtration (-94%). However, localized tissue hypoxia, as evidence by pimonidazole adduct immunohistochemistry, was detected in kidneys subjected to ischemia and reperfusion, particularly in, but not exclusive to, the outer medulla. Thus, cellular hypoxia, particularly in the outer medulla, may still be present during reperfusion even when reductions in tissue Po2 are not detected in the cortex or inner medulla.


Assuntos
Injúria Renal Aguda/metabolismo , Rim/metabolismo , Consumo de Oxigênio , Oxigênio/metabolismo , Traumatismo por Reperfusão/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Animais , Biomarcadores/metabolismo , Hipóxia Celular , Modelos Animais de Doenças , Fluorescência , Taxa de Filtração Glomerular , Hemoglobinas/metabolismo , Imuno-Histoquímica , Rim/irrigação sanguínea , Rim/patologia , Rim/fisiopatologia , Fluxometria por Laser-Doppler , Masculino , Modelos Cardiovasculares , Nitroimidazóis/metabolismo , Ratos , Ratos Sprague-Dawley , Circulação Renal , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Reprodutibilidade dos Testes , Índice de Gravidade de Doença , Fatores de Tempo
9.
Biochem Biophys Res Commun ; 446(2): 423-7, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24495806

RESUMO

A non-membrane bound form of Neprilysin (NEP) with catalytic activity has the potential to cleave substrates throughout the circulation, thus leading to systemic effects of NEP. We used the endothelial cell line Ea.hy926 to identify the possible role of exosomes and A Disintegrin and Metalloprotease 17 (ADAM-17) in the production of non-membrane bound NEP. Using a bradykinin based quenched fluorescent substrate (40 µM) assay, we determined the activity of recombinant human NEP (rhNEP; 12 ng), and NEP in the media of endothelial cells (10% v/v; after 24 h incubation with cells) to be 9.35±0.70 and 6.54±0.41 µmols of substrate cleaved over 3h, respectively. The presence of NEP in the media was also confirmed by Western blotting. At present there are no commercially available inhibitors specific for ADAM-17. We therefore synthesised two inhibitors TPI2155-14 and TPI2155-17, specific for ADAM-17 with IC50 values of 5.36 and 4.32 µM, respectively. Treatment of cells with TPI2155-14 (15 µM) and TPI2155-17 (4.3 µM) resulted in a significant decrease in NEP activity in media (62.37±1.43 and 38.30±4.70, respectively as a % of control; P<0.0001), implicating a possible role for ADAM-17 in NEP release. However, centrifuging media (100,000g for 1 h at 4 °C) removed all NEP activity from the supernatant indicating the likely role of exosomes in the release of NEP. Our data therefore indicated for the first time that NEP is released from endothelial cells via exosomes, and that this process is dependent on ADAM-17.


Assuntos
Proteínas ADAM/metabolismo , Células Endoteliais/metabolismo , Exossomos/metabolismo , Neprilisina/biossíntese , Proteína ADAM17 , Catálise , Linhagem Celular , Humanos , Neprilisina/química , Transdução de Sinais/fisiologia , Solubilidade
10.
Mol Cell Biochem ; 396(1-2): 49-54, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25226840

RESUMO

This study examined the effect of nitric oxide on the production of soluble ECE-1. Activity of ECE-1 in media was measured using a quenched fluorescent substrate assay, and expressed as a percentage of control. Endothelial cells were incubated with the nitric oxide donor Diethylenetriamine NONOate (DETA; 250-800 µM), NOS substrate L-Arg (200-1,000 µM), a L-Arg transport inhibitor (L-Lys; 10 µM) and NOS inhibitors (L-Gln and N5-[imino(nitroamino)methyl]-L-ornithine, methyl ester, monohydrochloride (L-NAME); 10-100 µM). The effect of L-Arg (1,000 µM) was also tested in the presence of L-Lys (10 µM), L-Gln (100 µM) and L-NAME (10-100 µM). Ultracentrifugation (100,000×g, 4 °C, 1 h) completely removed ECE-1 activity from the supernatant. In addition, fractionation of concentrated media on a sucrose density gradient indicated that ECE-1 activity was localised to the mid portion of the gradient, thus suggesting the possible role of exosomes in ECE-1 release. Production of soluble ECE-1 by Ea.hy926 cells was inhibited significantly (P < 0.05, unpaired t test, n = 4) in the presence of DETA (75.31 ± 3.59; 800 µM) and L-Arg (60.97 ± 9.22; 1,000 µM). L-Arg-mediated reduction in the release of soluble ECE-1 was blocked by the inhibition of NOS using L-NAME (100 µM; 99.19 ± 0.58) and L-Gln (100 µM; 104.41 ± 0.65). In addition, the presence of L-Lys (10 µM) significantly blocked the L-Arg (1,000 µM)-induced reduction in soluble ECE-1 levels (122.38 ± 13.16). These treatments had no effect on the expression of ECE-1 on the cell surface. Our data provide evidence that NO can inhibit the production of soluble ECE-1 by endothelial cells.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Células Endoteliais/metabolismo , Metaloendopeptidases/metabolismo , Óxido Nítrico/metabolismo , Arginina/farmacologia , Ácido Aspártico Endopeptidases/química , Linhagem Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Endoteliais/efeitos dos fármacos , Enzimas Conversoras de Endotelina , Humanos , Lisina/farmacologia , Metaloendopeptidases/química , NG-Nitroarginina Metil Éster/farmacologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Compostos Nitrosos/farmacologia , Solubilidade , Ultracentrifugação
11.
Clin Exp Pharmacol Physiol ; 41(12): 1031-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25115333

RESUMO

Observational studies indicate that psychological stress may contribute to the pathogenesis of hypertension and this may be further accentuated by factors such as endothelial dysfunction. On this basis, we aimed to determine whether oxidative stress enhances pressor responses to stressful stimuli and whether augmenting endothelial function by increasing the transport of L-arginine can counter the effects of oxidative stress. Telemetry probes were used to measure mean arterial pressure (MAP) in wild-type (WT; n = 6) and endothelial cationic amino acid transporter-1 (CAT-1)-overexpressing (CAT+) mice (n = 6) before and during an aversive (restraint) and non-aversive (almond feeding) stressor. The superoxide dismutase inhibitor diethyldithiocarbamic acid (DETCA; 30 mg/kg per day; 14 days) was then administered via a minipump to induce oxidative stress. Stress responses to feeding and restraint were repeated during Days 11-12 of DETCA infusion. In WT mice, pressor responses to restraint and feeding were augmented during infusion of DETCA (35 ± 1 and 28 ± 1 mmHg, respectively) compared with respective pretreatment responses (28 ± 2 and 24 ± 1 mmHg, respectively; P ≤ 0.01). In CAT+ mice, pressor responses to feeding were blunted during DETCA (20 ± 1 mmHg) compared with the control response (23 ± 1 mmHg; P = 0.03). In these mice, pressor responses to restraint were similar before (28 ± 1 mmHg) and during (26 ± 1 mmHg) DETCA infusion (P = 0.26). We conclude that endothelial CAT-1 overexpression can counter the ability of oxidative stress to augment pressor responses to behavioural stress.


Assuntos
Pressão Sanguínea/genética , Transportador 1 de Aminoácidos Catiônicos/genética , Células Endoteliais/metabolismo , Estresse Oxidativo/genética , Animais , Arginina/genética , Pressão Sanguínea/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Hipertensão/genética , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos , Superóxido Dismutase/genética , Vasoconstritores/farmacologia
12.
Pflugers Arch ; 465(7): 929-34, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23328864

RESUMO

Endothelin is one of the most potent peptide vasoconstrictors thus far characterised. It is produced by the cleavage of its precursor big endothelin-1 by endothelin-converting enzyme-1 (ECE-1). The endothelin system which includes endothelin-1 (ET-1), ET receptors and ECE-1 is well characterised in the kidney and is known to play a key role in the pathogenesis of end-stage renal disease (ESRD). Therefore, inhibition of ECE-1 and antagonism of ET receptors represent potential therapeutic approaches for the treatment of ESRD. Here, we review the current literature on the localisation of ECE-1 in the normal kidney and how ECE-1 expression is altered in pathological conditions leading to ESRD. We also discuss the roles of neutral endopeptidase (NEP) and chymase in mediating the production of ET-1 in the kidney in ESRD. As such, we also discuss that complete inhibition of ET-1 production in the kidney requires the inhibition of ECE-1, NEP and chymase.


Assuntos
Ácido Aspártico Endopeptidases/antagonistas & inibidores , Falência Renal Crônica/metabolismo , Metaloendopeptidases/antagonistas & inibidores , Animais , Ácido Aspártico Endopeptidases/metabolismo , Quimases/metabolismo , Enzimas Conversoras de Endotelina , Endotelinas/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Falência Renal Crônica/tratamento farmacológico , Falência Renal Crônica/enzimologia , Metaloendopeptidases/metabolismo , Neprilisina/metabolismo
13.
Curr Opin Nephrol Hypertens ; 22(1): 45-50, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23095292

RESUMO

PURPOSE OF REVIEW: L-Arginine (L-Arg) is the substrate for nitric oxide (NO) formation. Reduced NO bioavailability, particularly within the renal circulation, has been identified as a key factor in the pathogenesis of hypertension. This review focuses on the pathogenic role of abnormal L-Arg transport, particularly within the kidney, in hypertension. RECENT FINDINGS: Most recent studies have attempted to restore NO bioavailability in cardiovascular diseases with the use of antioxidants to reduce NO inactivation, but this approach has failed to provide beneficial effects in the clinical setting. We argue that this may be due to reduced NO formation in hypertension, which has largely been overlooked as a means of restoring NO bioavailability in cardiovascular diseases. Recent data indicate that renal L-Arg transport plays an important role in regulating both renal perfusion and function and the long-term set point of arterial pressure in health. Perturbations in the renal L-Arg transport system can give rise to abnormal renal perfusion and function, initiating hypertension and related renal damage. SUMMARY: Accordingly, we propose that L-Arg transporters are a new treatment target in hypertension and in disease states where renal NO bioavailability is disturbed.


Assuntos
Arginina/metabolismo , Pressão Sanguínea , Hipertensão/metabolismo , Óxido Nítrico/biossíntese , Circulação Renal , Endotélio/fisiopatologia , Humanos , Hipertensão/fisiopatologia
15.
J Alzheimers Dis ; 85(3): 943-955, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34897090

RESUMO

Hypertension is a major risk factor for the pathogenesis of vascular dementia and Alzheimer's disease. Chronic activation of the renin-angiotensin system (RAS) contributes substantially to neuroinflammation. We propose that neuroinflammation arising from chronic RAS activation can initiate and potentiate the onset of hypertension and related dementia. Neuroinflammation induced by chronic activation of the RAS plays a key role in the pathogenesis of dementia. Increased levels of pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and transforming growth factor (TGF)-ß have been reported in brain tissue of vascular dementia patients and animal models of vascular dementia induced by either angiotensin II infusion or transverse aortic coarctation. It is proposed that neuronal cell death and synaptic dysfunction induced by neuroinflammation lead to cognitive impairment in dementia. The neuroprotective RAS pathway, regulated by angiotensin-converting enzyme 2 (ACE2) which converts angiotensin II into angiotensin-(1-7), can attenuate hypertension and dementia. Furthermore, the use of anti-hypertensive medications in preventing dementia or cognitive decline in hypertensive patients and animal models of dementia have mostly been beneficial. Current evidence suggests a strong link between RAS induced neuroinflammation and the onset of hypertension and dementia, which warrants further investigation. Strategies to counteract an overactive RAS and enhance the neuroprotective arm of the RAS may help prevent or improve cognitive impairment associated with hypertension.


Assuntos
Citocinas/metabolismo , Demência Vascular/fisiopatologia , Hipertensão/complicações , Inflamação/complicações , Sistema Renina-Angiotensina/fisiologia , Angiotensina I , Angiotensina II/metabolismo , Animais , Anti-Hipertensivos/efeitos adversos , Anti-Hipertensivos/uso terapêutico , Encéfalo/metabolismo , Citocinas/imunologia , Humanos , Hipertensão/tratamento farmacológico , Fragmentos de Peptídeos
16.
Front Physiol ; 12: 813012, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35087423

RESUMO

Despite current therapies for diabetic nephropathy, many patients continue to progress to end-stage renal disease requiring renal replacement therapy. While the precise mechanisms underlying diabetic nephropathy remain to be determined, it is well established that chronic activation of the renin angiotensin aldosterone system (RAAS) plays a substantial role in the pathogenesis of diabetic nephropathy. Angiotensin converting enzyme 2 (ACE2), the enzyme responsible for activating the reno-protective arm of the RAAS converts angiotensin (Ang) II into Ang 1-7 which exerts reno-protective effects. Chronic RAAS activation leads to kidney inflammation and fibrosis, and ultimately lead to end-stage kidney disease. Currently, angiotensin converting enzyme inhibitors and Ang II receptor blockers are approved for renal fibrosis and inflammation. Targeting the reno-protective arm of the RAAS should therefore, provide further treatment options for kidney fibrosis and inflammation. In this review, we examine how targeting the reno-protective arm of the RAAS can ameliorate kidney inflammation and fibrosis and rescue kidney function in diabetic nephropathy. We argue tissue ACE2 stimulation provides a unique and promising therapeutic approach for diabetic nephropathy.

17.
Clin Exp Pharmacol Physiol ; 37(2): e58-69, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19566838

RESUMO

1. Angiotensin (Ang) II has multiple actions in the renal medullary circulation. It can induce vasodilatation and blunt the response of medullary blood flow (MBF) to renal nerve activation through AT(1) receptor-mediated release of nitric oxide (NO) and/or vasodilator prostaglandins. These actions require high intravascular and/or intratubular AngII concentrations, so are not apparent under physiological conditions. 2. Nevertheless, these mechanisms blunt the responsiveness of MBF to AT(1) receptor-mediated vasoconstriction. When these protective mechanisms fail, as when oxidative stress reduces NO bioavailability in the medullary circulation, AngII reduces MBF. If sustained, reduced MBF leads to the development of hypertension. 3. Chronic activation of the renin-angiotensin system (RAS) induces oxidative stress in the kidney. Therefore, MBF may be reduced in models of hypertension associated with RAS activation both because AngII levels per se are increased and because of increased responsiveness of MBF to AngII-induced vasoconstriction. 4. Endogenous AngII enhances the responsiveness of MBF to renal nerve stimulation, whereas NO blunts it. Chronic RAS activation and/or oxidative stress should therefore be expected to enhance MBF responses to renal nerve stimulation. Consistent with this, reductions in MBF induced by renal nerve stimulation are enhanced in rabbits with AngII-induced hypertension, renovascular hypertension or after 9 weeks of fat feeding. 5. We conclude that the ability of endogenous AngII to reduce MBF and enhance the response of MBF to activation of the renal nerves could contribute to the development of hypertension under conditions of RAS activation, especially if accompanied by increased renal sympathetic nerve activity.


Assuntos
Angiotensina II/fisiologia , Hipertensão Renal/fisiopatologia , Medula Renal/irrigação sanguínea , Medula Renal/fisiologia , Circulação Renal/fisiologia , Animais , Cães , Medula Renal/inervação , Camundongos , Óxido Nítrico/fisiologia , Estresse Oxidativo/fisiologia , Prostaglandinas/fisiologia , Coelhos , Ratos , Circulação Renal/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia , Sistema Nervoso Simpático/fisiologia , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia
18.
Clin Exp Pharmacol Physiol ; 36(3): 249-55, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19076168

RESUMO

1. l-Arginine is the substrate for vascular nitric oxide (NO) formation. Under normal physiological conditions, intracellular l-arginine levels far exceed the K(m) of NO synthase for l-arginine. However, endogenous NO formation is dependent on extracellular l-arginine concentrations, giving rise to the concept of the 'l-arginine paradox'. 2. Nitric oxide production in epithelial and endothelial cells is closely coupled to cellular l-arginine uptake, indicating that l-arginine transport mechanisms play a major role in the regulation of NO-dependent function. 3. Consistent with the data in endothelial and epithelial cells are functional data indicating that exogenous l-arginine can increase renal vascular and tubular NO bioavailability and thereby influence kidney perfusion, function and arterial pressure. The integrated effect of increased cellular l-arginine transport is to lower arterial pressure. Therefore, the use of l-arginine in the treatment of hypertension warrants investigation. 4. Low NO bioavailability is central to the development and maintenance of hypertension and to related endothelial dysfunction and target organ damage. We propose that l-arginine can interrupt the vicious cycle that initiates and maintains low NO in hypertension by increasing the formation of NO.


Assuntos
Arginina/metabolismo , Hipertensão/metabolismo , Óxido Nítrico/metabolismo , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Anti-Hipertensivos/uso terapêutico , Arginina/uso terapêutico , Pressão Sanguínea , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Humanos , Hipertensão/fisiopatologia , Cinética , Óxido Nítrico Sintase/metabolismo , Equilíbrio Hidroeletrolítico
19.
Artigo em Inglês | MEDLINE | ID: mdl-28971608

RESUMO

Endothelin-1 (ET-1) and nitric oxide (NO) are two highly potent vasoactive molecules with opposing effects on the vasculature. Endothelin-converting enzyme (ECE) and nitric oxide synthase (NOS) catalyse the production of ET-1 and NO, respectively. It is well established that these molecules play a crucial role in the initiation and progression of cardiovascular diseases and have therefore become targets of therapy. Many studies have examined the mechanism(s) by which NO regulates ET-1 production. Expression and localization of ECE-1 is a key factor that determines the rate of ET-1 production. ECE-1 can either be membrane bound or be released from the cell surface to produce a soluble form. NO has been shown to reduce the expression of both membrane-bound and soluble ECE-1. Several studies have examined the mechanism(s) behind NO-mediated inhibition of ECE expression on the cell membrane. However, the precise mechanism(s) behind NO-mediated inhibition of soluble ECE production are unknown. We hypothesize that both exogenous and endogenous NO, inhibits the production of soluble ECE-1 by preventing its release via extracellular vesicles (e.g., exosomes), and/or by inhibiting the activity of A Disintegrin and Metalloprotease-17 (ADAM17). If this hypothesis is proven correct in future studies, these pathways represent targets for the therapeutic manipulation of soluble ECE-1 production.


Assuntos
Proteína ADAM17/metabolismo , Enzimas Conversoras de Endotelina/metabolismo , Óxido Nítrico/farmacologia , Animais , Membrana Celular/metabolismo , Regulação para Baixo , Endotelina-1/metabolismo , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Humanos
20.
Sci Rep ; 7(1): 17718, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29255249

RESUMO

Mechanisms underlying the renal pathology in cardiorenal syndrome (CRS) type 2 remain elusive. We hypothesised that renal glutathione deficiency is central to the development of CRS type 2. Glutathione precursor, N-acetylcysteine (NAC;40 mg/kg/day; 8 weeks) or saline were administered to transgenic mice with dilated cardiomyopathy (DCM) and wild-type (WT) controls. Cardiac structure, function and glutathione levels were assessed at the end of this protocol. Renal fibrosis, glutathione content, expression of inflammatory and fibrotic markers, and function were also evaluated. In both genotypes, NAC had minimal effect on cardiac glutathione, structure and function (P ≥ 0.20). In NAC treated DCM mice, loss of glomerular filtration rate (GFR), tubulointerstitial and glomerular fibrosis and renal oxidised glutathione levels were attenuated by 38%, 99%, 70% and 52% respectively, compared to saline treated DCM mice (P ≤ 0.01). Renal expression of PAI-1 was greater in saline treated DCM mice than in WT mice (P < 0.05). Renal PAI-1 expression was less in NAC treated DCM mice than in vehicle treated DCM mice (P = 0.03). Renal IL-10 expression was greater in the former cohort compared to the latter (P < 0.01). These data indicate that normalisation of renal oxidized glutathione levels attenuates PAI-1 expression and renal inflammation preventing loss of GFR in experimental DCM.


Assuntos
Acetilcisteína/metabolismo , Síndrome Cardiorrenal/fisiopatologia , Fibrose/prevenção & controle , Acetilcisteína/farmacologia , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Modelos Animais de Doenças , Taxa de Filtração Glomerular , Glutationa/metabolismo , Rim/metabolismo , Rim/fisiopatologia , Nefropatias/patologia , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Nefrite/metabolismo , Estresse Oxidativo , Sistema Urinário/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA