Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Cardiovasc Magn Reson ; 20(1): 75, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30463565

RESUMO

BACKGROUND: Current guidelines for assessing the risk of experiencing a hospitalized cardiovascular (CV) event discourage stress testing of asymptomatic individuals; however, these recommendations are based on evidence gathered primarily from those aged < 60 years, and do not address the possibility of unrecognized "silent myocardial ischemia" in middle aged and older adults. METHODS: We performed dobutamine cardiovascular magnetic resonance (CMR) stress testing in 327 consecutively recruited participants aged > 55 years without CV-related symptoms nor known coronary artery disease, but otherwise at increased risk for a future CV event due to pre-existing hypertension or diabetes mellitus for at least 5 years. After adjusting for the demographics and CV risk factors, log-rank test and Cox proportional hazards models determined the additional predictive value of the stress test results for forecasting hospitalized CV events/survival. Either stress-induced LV wall motion abnormalities or perfusion defects were used to indicate myocardial ischemia. RESULTS: Participants averaged 68 ± 8 years in age; 39% men, 75% Caucasian. There were 38 hospitalized CV events or deaths which occurred during a mean follow-up of 58 months. Using Kaplan-Meier analyses, myocardial ischemia identified future CV events/survival (p <  0.001), but this finding was more evident in men (p <  0.001) versus women (p = 0.27). The crude hazard ratio (HR) of myocardial ischemia for CV events/survival was 3.13 (95% CI: 1.64-5.93; p < 0.001). After accounting for baseline demographics, CV risk factors, and left ventricular ejection fraction/mass, myocardial ischemia continued to be associated with CV events/survival [HR: 4.07 (95% CI: 1.95-8.73) p < 0.001]. CONCLUSIONS: Among asymptomatic middle-aged individuals with risk factors for a sentinel CV event, the presence of myocardial ischemia during dobutamine CMR testing forecasted a future hospitalized CV event or death. Further studies are needed in middle aged and older individuals to more accurately characterize the prevalence, significance, and management of asymptomatic myocardial ischemia. TRIAL REGISTRATION: ( ClinicalTrials.gov identifier): NCT00542503 and was retrospectively registered on October 11th, 2007.


Assuntos
Agonistas de Receptores Adrenérgicos beta 1/administração & dosagem , Dobutamina/administração & dosagem , Imageamento por Ressonância Magnética/métodos , Isquemia Miocárdica/diagnóstico por imagem , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Doenças Assintomáticas , Progressão da Doença , Feminino , Hospitalização , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/mortalidade , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia , Valor Preditivo dos Testes , Prognóstico , Estudos Prospectivos , Medição de Risco , Fatores de Risco , Fatores de Tempo
2.
Am J Physiol Renal Physiol ; 298(5): F1214-21, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20053790

RESUMO

Mitochondrial dysfunction is involved in pathopysiology of ischemia-reperfusion-induced acute kidney injury (AKI). The p66shc adaptor protein is a newly recognized mediator of mitochondrial dysfunction, which might play a role in AKI-induced renal tubular injury. Oxidative stress-mediated Serine36 phosphorylation of p66shc facilitates its transportation to the mitochondria where it oxidizes cytochrome c and generates excessive amount of reactive oxygen species (ROS). The consequence is mitochondrial depolarization and injury. Earlier we determined that p66shc plays an essential role in injury of cultured mouse renal proximal tubule cells during oxidative stress. Here, we studied the role of p66shc in ROS generation and consequent mitochondrial dysfunction during oxidative injury in renal proximal tubule cells. We employed p66shc knockdown renal proximal tubule cells and cells that overexpress wild-type, Serine phosphorylation (S36A), or cytochrome c-binding (W134F) mutants of p66shc. Inhibition of the mitochondrial electron transport chain or the mitochondrial permeability transition revealed that hydrogen peroxide-induced injury is mitochondrial ROS and consequent mitochondrial depolarization dependent. We also found that through Ser36 phosphorylation and mitochondria/cytochrome c binding, p66shc mediates those effects. We propose a similar mechanism in vivo as we demonstrated mitochondrial binding of p66shc as well as its association with cytochrome c in the postischemic kidneys of mice. Thus, manipulating p66shc might offer a new therapeutic modality to ameliorate renal ischemic injury.


Assuntos
Injúria Renal Aguda/fisiopatologia , Túbulos Renais Proximais/fisiopatologia , Mitocôndrias/fisiologia , Estresse Oxidativo/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Animais , Linhagem Celular , Citocromos c/metabolismo , Peróxido de Hidrogênio/farmacologia , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Serina/metabolismo
3.
Am J Physiol Regul Integr Comp Physiol ; 299(4): R1044-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20668235

RESUMO

We have previously demonstrated that moderate hyperbilirubinemia decreases blood pressure in ANG II-dependent hypertension through mechanisms that decrease oxidative stress and increase nitric oxide levels. Since decreases in renal hemodynamics play an important role in mediating the hypertensive actions of ANG II, the goal of the present study was to examine the effect of moderate hyperbilirubinemia on glomerular filtration rate (GFR) and renal blood flow (RBF) in a mouse model of ANG II hypertension. Mice were made moderately hyperbilirubinemic by two methods: indinavir or specific morpholino antisense oligonucleotides against UGT1A1, which is the enzyme responsible for the conjugation of bilirubin in the liver. GFR and RBF were measured in mice after implantation of an osmotic minipump delivering ANG II at a rate of 1 µg·kg(-1)·min(-1). GFR was measured by continuous infusion of I(125)-labeled iothalamate on days 5 and 6 of ANG II infusion in conscious mice. RBF was measured on day 7 of ANG II infusion in anesthetized mice. Blood levels of unconjugated bilirubin were significantly increased in mice treated with indinavir or anti-UGT1A1 (P = 0.002). ANG II decreased GFR by 33% of control (n = 9, P = 0.004), and this was normalized by moderate hyperbilirubinemia (n = 6). Next, we examined the effect of moderate hyperbilirubinemia on RBF in ANG II-infused mice. ANG II infusion significantly decreased RBF by 22% (P = 0.037) of control, and this decrease was normalized by moderate hyperbilirubinemia (n = 6). These results indicate that improvement of renal hemodynamics may be one mechanism by which moderate hyperbilirubinemia lowers blood pressure in this model.


Assuntos
Angiotensina II/fisiologia , Hiperbilirrubinemia/fisiopatologia , Hipertensão/fisiopatologia , Circulação Renal/fisiologia , Animais , Bilirrubina/sangue , Western Blotting , Taxa de Filtração Glomerular/fisiologia , Glucuronosiltransferase/metabolismo , Indinavir/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfolinas/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Inibidores da Síntese de Proteínas/farmacologia
4.
Pharmacol Res ; 61(6): 525-30, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20338241

RESUMO

Recent studies have identified imidazole-dioxolane based compounds as novel heme oxygenase (HO) inhibitors. While these compounds have been demonstrated to be specific HO inhibitors in vitro, they have yet to be used to inhibit renal HO activity in vivo. The goal of this study was to determine the effectiveness of the imidazole-dioxolane HO-1 inhibitor, QC-13, in the inhibition of renal HO activity in vivo. HO-1 was induced in mice by treatment with cobalt protoporphyrin (CoPP). After 5 days, QC-13 was delivered either by continuous intrarenal medullary interstitial infusion (IRMI) into one kidney at several concentrations for 72 h or by two intraperitoneal injections over a 48-h period. IRMI infusion of QC-13 at a concentration of 25 microM resulted in a significant decrease in medullary but not cortical HO activity as compared to CoPP treated kidneys. IRMI infusion of QC-13 at a lower concentration (2.5 microM) had no effect on either medullary or cortical HO activity in CoPP treated mice. In contrast, administration of QC-13 at a higher concentration (250 microM) resulted in a significant decrease in both medullary and cortical HO activity in CoPP treated mice. Systemic administration of QC-13 resulted in significant decrease both renal cortical and medullary HO activity in CoPP treated mice. In contrast to classical porphyrin based HO inhibitors, IRMI infusion of QC-13 did not induce HO-1 protein levels as determined by Western blot analysis of medullary protein samples. Our results demonstrated that imidazole-dioxolane inhibitors are renal HO inhibitors in vivo and can inhibit HO activity independent of HO-1 induction. These inhibitors may be useful tools to elucidate the role of renal HO-1 in numerous physiologic and pathophysiologic conditions.


Assuntos
Dioxolanos/farmacologia , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Heme Oxigenase (Desciclizante)/metabolismo , Imidazóis/farmacologia , Rim/enzimologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R738-43, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19571206

RESUMO

Population studies indicate that moderate hyperbilirubinemia is associated with reduced incidence of cardiovascular diseases, including hypertension. Despite this correlative evidence, no studies have directly tested the hypothesis that moderate increases in plasma bilirubin levels can attenuate the development of hypertension. This hypothesis was tested by treating mice with Indinavir, a drug that competes with bilirubin for metabolism by UDP-glucuronosyltransferase 1A1 (UGT1A1). Treatment of mice with Indinavir (500 mg x kg(-1) x day(-1), gavage) resulted in a twofold increase in plasma unconjugated bilirubin levels. Next, we determined the effect of Indinavir-induced changes in plasma bilirubin on the development of ANG II-dependent hypertension. Moderate hyperbilirubinemia was induced 3 days before the implantation of an osmotic minipump that delivered ANG II at a rate of 1 microg x kg(-1) x min(-1). ANG II infusion increased mean arterial pressure (MAP) by 20 mmHg in control mice but by only 6 mmHg in mice treated with Indinavir (n = 6). Similar to Indinavir treatment, direct infusion of bilirubin (37.2 mg x kg(-1) x day(-1) i.v.) resulted in a twofold increase in plasma bilirubin levels and also attenuated the development of ANG II-dependent hypertension. Moderate hyperbilirubinemia resulted in an increase in plasma nitrate/nitrite levels, which averaged 36 +/- 2 vs. 50 +/- 7 microM in ANG II vehicle vs. Indinavir-treated mice (n = 5). Moderate hyperbilirubinemia resulted in attenuation of vascular oxidative stress as determined by dihydroethidium staining of aortic segments. These results indicate that moderate hyperbilirubinemia prevents ANG II-dependent hypertension by a mechanism that may involve decreases in vascular oxidative stress.


Assuntos
Bilirrubina/metabolismo , Hiperbilirrubinemia/metabolismo , Hipertensão/prevenção & controle , Angiotensina II , Animais , Bilirrubina/sangue , Pressão Sanguínea , Cardiomegalia/metabolismo , Cardiomegalia/prevenção & controle , Modelos Animais de Doenças , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/metabolismo , Hiperbilirrubinemia/induzido quimicamente , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Indinavir , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nitratos/sangue , Nitritos/sangue , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Estresse Oxidativo
6.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1822-8, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19846746

RESUMO

Heme oxygenase (HO) is the enzyme responsible for the breakdown of heme-generating carbon monoxide (CO) and biliverdin in this process. HO-2 is the constitutively expressed isoform in most tissues, such as the kidney and vasculature. CO generated by HO is believed to be an important vasodilator in the renal circulation along with another gas, nitric oxide (NO). To determine the importance of HO-2 in the regulation of blood pressure and renal blood flow (RBF), we treated HO-2 knockout (KO) mice chronically with either ANG II or N(G)-nitroarginine methyl ester (l-NAME). Basal blood pressures were not different between wild-type (WT), heterozygous (HET), or KO mice and averaged 113 +/- 3 vs. 115 +/- 2 vs. 116 +/- 2 mmHg. Similar increases in blood pressure to chronic ANG II as well as l-NAME treatment were observed in all groups with blood pressures increasing an average of 30 mmHg in response to ANG II and 15 mmHg in response to l-NAME. Basal RBFs were not different between the groups averaging 6.0 +/- 0.5 (n = 6) vs. 4.8 +/- 0.6 (n = 10) vs. 5.8 +/- 0.7 (n = 6) ml*min(-1)*g(-1) kidney weight in WT, HET, and KO mice. HO-2 KO and HET mice exhibited an attenuated decrease in RBF in response to acute administration of ANG II, while no differences were observed with l-NAME. Our data indicate that blood pressure and RBF responses to increased ANG II or inhibition of nitric oxide are not significantly enhanced in HO-2 KO mice.


Assuntos
Pressão Sanguínea , Heme Oxigenase (Desciclizante)/deficiência , Hipertensão/enzimologia , Rim/irrigação sanguínea , Circulação Renal , Administração Oral , Angiotensina II/administração & dosagem , Animais , Cardiomegalia/enzimologia , Cardiomegalia/genética , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Feminino , Heme Oxigenase (Desciclizante)/genética , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/fisiopatologia , Bombas de Infusão Implantáveis , Infusões Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NG-Nitroarginina Metil Éster/administração & dosagem , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Resistência Vascular , Vasoconstrição
7.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1546-53, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19759334

RESUMO

Induction of heme oxygenase-1 (HO-1) in the renal medulla increases carbon monoxide and bilirubin production and decreases ANG II-mediated superoxide production. The goal of this study was to determine the importance of increases in bilirubin to the antioxidant effects of HO-1 induction in cultured mouse thick ascending loop of Henle (TALH) and inner medullary collecting duct (IMCD3) cells. Bilirubin levels were decreased by using small interfering RNAs (siRNAs) targeted to biliverdin reductase (BVR), which is the cellular enzyme responsible for the conversion of biliverdin to bilirubin. Treatment of cultured TALH or IMCD-3 cells with BVR siRNA (50 or 100 nM) resulted in an 80% decrease in the level of BVR protein and decreased cellular bilirubin levels from 46 +/- 5 to 23 +/- 4 nM (n = 4). We then determined the effects of inhibition of BVR on ANG II-mediated superoxide production. Superoxide production induced by ANG II (10(-9) M) significantly increased in both TALH and IMCD-3 cells. Treatment of TALH cells with BVR siRNA resulted in a significant increase in ouabain-sensitive rubidium uptake from 95 +/- 6 to 122 +/- 5% control (n = 4, P < 0.05). Lastly, inhibition of BVR with siRNA did not prevent the decrease in superoxide levels observed in cells pretreated with the HO-1 inducer, hemin. We conclude that decreased levels of cellular bilirubin increase ANG II-mediated superoxide production and sodium transport; however, increases in bilirubin are not necessary for HO-1 induction to attenuate ANG II-mediated superoxide production.


Assuntos
Angiotensina II/metabolismo , Células Epiteliais/metabolismo , Túbulos Renais Distais/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Superóxidos/metabolismo , Animais , Bilirrubina/metabolismo , Biliverdina/farmacologia , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Hemina/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Túbulos Renais Distais/citologia , Túbulos Renais Distais/efeitos dos fármacos , Camundongos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , RNA Interferente Pequeno/farmacologia
8.
Am J Hypertens ; 21(2): 189-93, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18174886

RESUMO

BACKGROUND: Induction of heme oxygenase-1 (HO-1) attenuates the development of angiotensin II (Ang II)-dependent hypertension in mice. However, the mechanism by which HO-1 lowers blood pressure in this model is not clear. This study was designed to determine whether induction of HO-1 results in an improvement in vascular relaxation in Ang II hypertensive mice. METHODS: Mice were treated with either of the vehicles (control), the HO-1 inducer cobalt protoporphyrin (CoPP;50 mg/kg), Ang II(1 microg/kg/min, 14 days), or Ang II + CoPP. CoPP was administered as a single bolus dose 2 days prior to subcutaneous implantation of the osmotic minipump containing Ang II. Vascular relaxation was examined in isolated carotid arteries precontracted with the thromboxane mimetic U46619 (0.4 microg/ml). RESULTS: Endothelial dependent relaxation to acetylcholine (ACh; 1 micromol/l) was significantly impaired in Ang II-treated mice compared to control mice (56 +/- 3% vs. 40 +/- 4%, P < 0.05, n > or = 6). Similarly, endothelial independent relaxation to sodium nitroprusside (SNP; 1 micromol/l) was significantly impaired in Ang II mice (56 +/- 6% vs. 28 +/- 6%, P < 0.05, n > or = 6). Relaxation in response to the carbon monoxide donor, CORM-A1 (100 micromol/l), was attenuated after Ang II treatment (75 +/- 7% vs. 59 +/- 7%,P < 0.05, n > or = 6). CoPP treatment induced HO-1 but not HO-2 protein in the aorta, as measured by western blot analysis. CoPP treatment had no effect on vascular responses in control mice and did not improve ACh (26 +/- 5%, n = 15), SNP (23 +/- 4%, n = 15), or CORM-A1 (46 +/- 7%, n = 10) dependent relaxation in Ang II treated mice. CONCLUSIONS: These results suggest that induction of HO-1 lowers Ang II-dependent hypertension through a mechanism independent of improved vascular relaxation.


Assuntos
Heme Oxigenase-1/metabolismo , Hipertensão/tratamento farmacológico , Hipertensão/fisiopatologia , Protoporfirinas/farmacologia , Vasodilatação/fisiologia , Angiotensina II/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Interações Medicamentosas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Ativação Enzimática/efeitos dos fármacos , Hipertensão/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos
9.
Am J Cardiol ; 116(11): 1752-5, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26433273

RESUMO

Myocardial injury because of oxidative stress manifesting through reductions in left ventricular ejection fraction (LVEF) may occur after the administration of anthracycline-based chemotherapy (A-bC). We hypothesized that bilirubin, an effective endogenous antioxidant, may attenuate the reduction in LVEF that sometimes occurs after receipt of A-bC. We identified 751 consecutively treated patients with cancer who underwent a pre-A-bC LVEF measurement, exhibited a serum total bilirubin level <2 mg/dl, and then received a post-A-bC LVEF assessment because of symptomatology associated with heart failure. Analysis of variance, Tukey's Studentized range test, and chi-square tests were used to evaluate an association between bilirubin and LVEF changes. The LVEF decreased by 10.7 ± 13.7%, 8.9 ± 11.8%, and 7.7 ± 11.5% in group 1 (bilirubin at baseline ≤0.5 mg/dl), group 2 (bilirubin 0.6 to 0.8 mg/dl), and group 3 (bilirubin 0.9 to 1.9 mg/dl), respectively. More group 1 patients experienced >15% decrease in LVEF compared with those in group 3 (p = 0.039). After adjusting for age, coronary artery disease/myocardial infarction, diabetes mellitus, hematocrit, and the use of cardioactive medications, higher precancer treatment bilirubin levels and lesser total anthracycline doses were associated with LVEF preservation (p = 0.047 and 0.011, respectively). In patients treated with anthracyclines who subsequently develop symptoms associated with heart failure, pre-anthracycline treatment serum bilirubin levels inversely correlate with subsequent deterioration in post-cancer treatment LVEF. In conclusion, these results suggest that increased levels of circulating serum total bilirubin, an intrinsic antioxidant, may facilitate preservation of LVEF in patients receiving A-bC for cancer.


Assuntos
Antraciclinas/uso terapêutico , Antineoplásicos/uso terapêutico , Bilirrubina/sangue , Neoplasias Hematológicas/tratamento farmacológico , Volume Sistólico , Adulto , Idoso , Antraciclinas/efeitos adversos , Antraciclinas/farmacologia , Antibióticos Antineoplásicos/farmacologia , Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Neoplasias da Mama/sangue , Neoplasias da Mama/tratamento farmacológico , Daunorrubicina/farmacologia , Daunorrubicina/uso terapêutico , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Feminino , Neoplasias Hematológicas/sangue , Humanos , Idarubicina/farmacologia , Idarubicina/uso terapêutico , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Estudos Retrospectivos , Volume Sistólico/efeitos dos fármacos , Disfunção Ventricular Esquerda/induzido quimicamente
10.
Curr Pharm Biotechnol ; 13(6): 819-26, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22201605

RESUMO

Carbon monoxide (CO) is an endogenously produced gas resulting from the degradation of heme by heme oxygense or from fatty acid oxidation. Heme oxygenase (HO) enzymes are constitutively expressed in the kidney (HO-2) and HO-1 is induced in the kidney in response to several physiological and pathological stimuli. While the beneficial actions of HO in the kidney have been recognized for some time, the important role of CO in mediating these effects has not been fully examined. Recent studies using CO inhalation therapy and carbon monoxide releasing molecules (CORMs) have demonstrated that increases in CO alone can be beneficial to the kidney in several forms of acute renal injury by limiting oxidative injury, decreasing cell apoptosis, and promoting cell survival pathways. Renal CO is also emerging as a major regulator of renal vascular and tubular function acting to protect the renal vasculature against excessive vasoconstriction and to promote natriuresis by limiting sodium reabsorption in tubule cells. Within this review, recent studies on the physiological actions of CO in the kidney will be explored as well as the potential therapeutic avenues that are being developed targeting CO in the kidney which may be beneficial in diseases such as acute renal failure and hypertension.


Assuntos
Monóxido de Carbono/fisiologia , Rim/fisiologia , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Animais , Pressão Sanguínea , Monóxido de Carbono/farmacologia , Monóxido de Carbono/uso terapêutico , Humanos
11.
Int J Hypertens ; 2012: 392890, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22506099

RESUMO

Angiotensin II (AngII) causes hypertension (HTN) and promotes renal injury while simultaneously inducing reno-protective enzymes like heme oxygenase-1 (HO-1). We examined the modulatory role of HO on sub-pressor angiotensin II (SP-AngII) induced renal inflammation and injury. We first tested whether the SP-AngII-induced renal dysfunction, inflammation and injury are exacerbated by either preventing (chronic HO-1 inhibition) or reversing (late HO-1 inhibition) SP-AngII-induced HO (using tin protoporphyrin; SnPP). We next examined whether additional chronic or late induction of SP-AngII-induced HO (using cobalt protoporphyrin; CoPP), prevents or ameliorates renal damage. We found that neither chronic nor late SnPP altered blood pressure. Chronic SnPP worsened SP-AngII-induced renal dysfunction, inflammation, injury and fibrosis, whereas late SnPP worsened renal dysfunction but not inflammation. Chronic CoPP prevented HTN, renal dysfunction, inflammation and fibrosis, but surprisingly, not the NGAL levels (renal injury marker). Late CoPP did not significantly alter SP-AngII-induced HTN, renal inflammation or injury, but improved renal function. Thus, we conclude (a) endogenous HO may be an essential determining factor in SP-AngII induced renal inflammation, injury and fibrosis, (b) part of HO's renoprotection may be independent of blood pressure changes; and (c) further induction of HO-1 protects against renal injury, suggesting a possible therapeutic target.

12.
Hypertension ; 52(4): 660-5, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18695146

RESUMO

The main goal of this study was to determine whether kidney-specific induction of heme oxygenase-1 (HO-1) can prevent the development of angiotensin (Ang) II-dependent hypertension. To test this hypothesis, intrarenal medullary interstitial catheters were implanted into the left kidney of uninephrectomized mice. Infusion of cobalt protoporphyrin (CoPP; 250 microg/mL; at 50 microL/h for 48 hours) resulted in significant induction of HO-1 in the renal medulla when examined 2 weeks after the infusion with no induction observed in other organs, such as the heart or liver. Next, we examined the effect of renal-specific induction of HO-1 on the development of Ang II-dependent hypertension. CoPP or vehicle (0.1 mol/L NaOH [pH 8.3]) was infused as indicated above 2 days before implantation of an osmotic minipump, which delivered Ang II or saline vehicle at a rate of 1 microg/kg per minute. Mean arterial pressure was measured in conscious, unrestrained mice for 3 consecutive days starting on day 7 after implantation of the minipumps. Mean arterial pressure averaged 114+/-5, 122+/-4, 162+/-2, and 125+/-6 mm Hg in vehicle-, intrarenal medullary interstitial CoPP-, Ang II-, and Ang II + intrarenal medullary interstitial CoPP-treated mice, respectively (n=6 or 7). These results demonstrate that kidney-specific induction of HO-1 prevents the development of Ang II-dependent hypertension and that induction of HO-1 in the kidney may be the mechanism by which systemic delivery of CoPP lowers blood pressure in Ang II-dependent hypertension.


Assuntos
Heme Oxigenase-1/biossíntese , Hipertensão/prevenção & controle , Medula Renal/enzimologia , Protoporfirinas/administração & dosagem , Angiotensina II/toxicidade , Animais , Pressão Sanguínea/fisiologia , Western Blotting , Modelos Animais de Doenças , Indução Enzimática , Heme/metabolismo , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Bombas de Infusão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espectrofotometria , Resultado do Tratamento
13.
Am J Physiol Renal Physiol ; 295(4): F1158-65, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18701634

RESUMO

Heme oxygenase (HO)-1 induction can attenuate the development of angiotensin II (ANG II)-dependent hypertension. However, the mechanism by which HO-1 lowers blood pressure is not clear. The goal of this study was to test the hypothesis that induction of HO-1 can reduce the ANG II-mediated increase in superoxide production in cultured thick ascending loop of Henle (TALH) cells. Studies were performed on an immortalized cell line of mouse TALH (mTALH) cells. HO-1 was induced in cultured mTALH cells by treatment with cobalt protoporphyrin (CoPP, 10 microM) or hemin (50 microM) or by transfection with a plasmid containing the human HO-1 isoform. Treatment of mTALH cells with 10(-9) M ANG II increased dihydroethidium (DHE) fluorescence (an index of superoxide levels) from 35.5+/-5 to 136+/-18 relative fluorescence units (RFU)/microm2. Induction of HO-1 via CoPP, hemin, or overexpression of the human HO-1 isoform significantly reduced ANG II-induced DHE fluorescence to 64+/-5, 64+/-8, and 41+/-4 RFU/microm2, respectively. To determine which metabolite of HO-1 is responsible for reducing ANG II-mediated increases in superoxide production in mTALH cells, cells were preincubated with bilirubin or carbon monoxide (CO)-releasing molecule (CORM)-A1 (each at 100 microM) before exposure to ANG II. DHE fluorescence averaged 80+/-7 RFU/microm2 after incubation with ANG II and was significantly decreased to 55+/-7 and 53+/-4 RFU/microm2 after pretreatment with bilirubin and CORM-A1. These results demonstrate that induction of HO-1 in mTALH cells reduces the levels of ANG II-mediated superoxide production through the production of both bilirubin and CO.


Assuntos
Angiotensina II/farmacologia , Heme Oxigenase-1/metabolismo , Alça do Néfron/enzimologia , Superóxidos/metabolismo , Vasoconstritores/farmacologia , Animais , Bilirrubina/metabolismo , Bilirrubina/farmacologia , Monóxido de Carbono/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Guanilato Ciclase/metabolismo , Heme Oxigenase-1/genética , Hemina/metabolismo , Hemina/farmacologia , Alça do Néfron/citologia , Alça do Néfron/efeitos dos fármacos , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Protoporfirinas/metabolismo , Protoporfirinas/farmacologia , RNA Interferente Pequeno , Receptores Citoplasmáticos e Nucleares/metabolismo , Guanilil Ciclase Solúvel
14.
Ren Fail ; 29(5): 543-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17654315

RESUMO

Recent studies have suggested that carbon monoxide (CO) inhalation can reduce ischemia-reperfusion injury of kidneys. The purpose of the present study was to determine whether the direct application of CO using tricarbonylchloro (glycinato) ruthenium II (CORM3) would reduce cold-rewarm-associated apoptosis in renal tubular epithelial (RPTE) cells. RPTE cells were subjected to 48 hours of cold followed by 24 hours of rewarming with increasing concentrations (0-500 microM) of CORM3. CORM3 (100 microM) reduced apoptosis as determined by the TUNEL method from 21.6 +/- 5.2 to 5.8 +/- 1.1 % (untreated vs. treated, n = 5; p < 0.001). We subsequently observed that the incubation of RPTE cells with CORM3 induced heme oxygenase (HO)-1 gene expression. As HO-1 itself can confer protection against cold rewarm injury, we investigated the role of HO-1 in the protective actions of CORM3 using siRNA oligonucleotides directed against HO-1. CORM3 treatment of RPTE cells caused a 4.9- fold increase in HO-1 gene expression as determined by real time PCR. Prior treatment of RPTE cells with siRNAs against HO-1 was able to completely abolish the CORM3 mediated induction of HO-1 mRNA and protein. The abolition of HO-induction with siRNAs did reduce CORM3-mediated protection against cold rewarm-induced apoptosis; however, CORM3 was able to significantly protect RPTE cells against cold-rewarm injury: apoptosis was 33.7 +/- 0.9% vs. 15.4 +/- 0.5% vs. 62.8 +/- 1.5% vs. 23.5 +/- 3.4 in control cold-rewarm vs. cold-rewarm + CORM3 (100 microM) vs. cold-rewarm + HO-1 siRNA vs. cold-rewarm + CORM3 (100 microM) + HO-1 siRNA (n = 4). These results suggest that increased levels of CO alone can protect against cold-rewarm-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Monóxido de Carbono/farmacologia , Células Epiteliais/efeitos dos fármacos , Túbulos Renais/citologia , Monóxido de Carbono/uso terapêutico , Células Cultivadas , Temperatura Baixa , Humanos , Compostos Organometálicos/farmacologia , Compostos Organometálicos/uso terapêutico , Reaquecimento
15.
Am J Physiol Regul Integr Comp Physiol ; 292(4): R1472-8, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17194725

RESUMO

Heme oxygenase-1 (HO-1) induction can attenuate the development of angiotensin II (ANG II)-dependent hypertension. However, the mechanism by which HO-1 lowers blood pressure in this model is not clear. The goal of this study was to test the hypothesis that induction of HO-1 in the kidney can attenuate the increase in reactive oxygen species (ROS) generation in the kidney that occurs during ANG II-dependent hypertension. Mice were divided into four groups, control (Con), cobalt protoporphyrin (CoPP), ANG II, and ANG II + CoPP. CoPP treatment (50 mg/kg) was administered in a single subcutaneous injection 2 days prior to implantation of an osmotic minipump that infused ANG II at a rate of 1 microg x kg(-1) x min(-1). At the end of this period, mean arterial blood pressure (MAP) averaged 93 +/- 5, 90 +/- 5, 146 +/- 8, and 105 +/- 6 mmHg in Con, CoPP-, ANG II-, and ANG II + CoPP-treated mice. To determine whether HO-1 induction resulted in a decrease in ANG II-stimulated ROS generation in the renal medulla, superoxide production was measured. Medullary superoxide production was increased by ANG II infusion and normalized in mice pretreated with CoPP. The reduction in ANG II-mediated superoxide production in the medulla with CoPP was associated with a decrease in extracellular superoxide dismutase protein but an increase in catalase protein and activity. These results suggest that reduction in superoxide and possibly hydrogen peroxide production in the renal medulla may be a potential mechanism by which induction of HO-1 with CoPP lowers blood pressure in ANG-II dependent hypertension.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/fisiologia , Heme Oxigenase-1/biossíntese , Hipertensão Renal/metabolismo , Medula Renal , Superóxidos/metabolismo , Vasoconstritores/farmacologia , Angiotensina II/administração & dosagem , Animais , Indução Enzimática , Heme Oxigenase-1/genética , Hipertensão Renal/induzido quimicamente , Hipertensão Renal/fisiopatologia , Implantes Experimentais , Infusões Intravenosas , Injeções Subcutâneas , Medula Renal/enzimologia , Medula Renal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Protoporfirinas/administração & dosagem , Protoporfirinas/farmacologia , Vasoconstritores/administração & dosagem
16.
Hypertension ; 45(4): 730-5, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15699464

RESUMO

Previous studies have indicated that the production of 20-hydroxyecisatatraenoic acid (20-HETE) is similar in the liver of C57/B6 mice and rats, but the renal production of 20-HETE is very low in this strain of mice. The present study examined the effects of induction of the renal production of 20-HETE with fenofibrate (FF) on the development of angiotensin II (Ang II)-dependent hypertension in C57BL/6J mice. The mice were divided into 4 groups and treated with vehicle (control), FF (90 mg/kg per day, IP), Ang II (1000 ng/kg per minute, SC), and Ang II plus FF. Mean arterial blood pressure (MAP) averaged 109+/-4 and 106+/-2 mm Hg in control and FF-treated mice (n=7). MAP was significantly increased in the Ang II-treated mice to 144+/-4 mm Hg (n=7). However, FF treatment prevented the development of Ang II-dependent hypertension, with MAP averaging 115+/-5 mm Hg in mice treated with both Ang II plus FF (n=7). Renal production of 20-HETE was very low in control (n=7) and Ang II-treated (n=7) mice and was increased by >2-fold in FF-treated (n=7) and Ang II plus FF-treated (n=7) mice. The levels of Cyp4A proteins were markedly increased in the kidneys of mice treated with FF and Ang II plus FF but not in the renal vasculature. These results suggest that upregulation of the production of 20-HETE in renal tubules may contribute to the blood pressure-lowering effects of FF treatment in Ang II-dependent hypertension in C57BL/6J mice.


Assuntos
Angiotensina II , Fenofibrato/farmacologia , Hipertensão/induzido quimicamente , Hipertensão/prevenção & controle , Vasoconstritores , Angiotensina II/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Citocromo P-450 CYP4A/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vasoconstritores/farmacologia
17.
J Am Soc Nephrol ; 16(4): 950-8, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15728782

RESUMO

Heme oxygenase (HO) induction has been demonstrated to be beneficial in limiting the extent of cellular damage after ischemia-induced acute renal failure (ARF). Because increased HO activity is associated with the production of carbon monoxide (CO) as well as the potent antioxidant bilirubin, it is unclear which of the two is of greater importance in the protective effects of HO induction. The purpose of this study was to determine the protective role of CO alone in ischemia-induced ARF. Bilateral clamping of the renal pedicle for 40 min was associated with a ninefold increase in the levels of plasma creatinine 24 h after reperfusion as compared with normal plasma creatinine levels; however, administration of CO donor compounds tricarbonyldichlororuthenium(II) dimer, ([Ru(CO)(3)Cl(2)](2), 10 mg/kg) or tricarbonylchloro(glycinato)ruthenium(II) ([Ru(CO)(3)Cl(glycinate)], (CORM-3) 1 h before the onset of ischemia significantly decreased the levels of plasma creatinine 24 h after reperfusion as compared with vehicle-treated mice. Surprising, treatment with the CO donors was associated with an increase in HO activity 24 h after ischemia. For determining whether the protective effects of the CO donors were due to CO or HO-1 induction, experiments were performed in which HO was inhibited before administration of the CO donors. Pretreatment with the HO inhibitor had no effect on the level of plasma creatinine 24 h after reperfusion after treatment with the CO donor compounds. These results suggest that CO itself may be protective and limit renal damage in ischemia induced ARF.


Assuntos
Injúria Renal Aguda/patologia , Monóxido de Carbono/metabolismo , Citoproteção , Isquemia/complicações , Rim/irrigação sanguínea , Compostos Organometálicos/farmacologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Animais , Creatinina/sangue , Inibidores Enzimáticos/farmacologia , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Isquemia/sangue , Isquemia/enzimologia , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/patologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo , Zinco/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA