Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 325(4): R337-R343, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37486069

RESUMO

Diabetes-induced glomerular hyperfiltration is an early alteration in kidney function in diabetes. Previous studies have shown that reduced adenosine A2a receptor signaling contributes to diabetes-induced glomerular hyperfiltration. The present study investigated the effects of enhanced interstitial adenosine concentration by inhibition of cellular adenosine reuptake, thereby promoting endogenous adenosine signaling. Insulinopenic diabetes was induced by streptozotocin in adult male Sprague-Dawley rats. Two weeks after diabetes induction, kidney function in terms of glomerular filtration rate, and total, cortical, and medullary renal blood flows were evaluated under thiobutabarbital anesthesia during baseline and after renal artery infusion of two doses of the adenosine reuptake inhibitor dilazep. Dilazep did not affect mean arterial pressure indicating that the effects of the interventions were intrarenal. Diabetics had increased glomerular filtration rate compared with controls and dilazep dose-dependently decreased glomerular filtration rate in diabetics, whereas it had no significant effect in controls. Dilazep increased cortical renal blood flows in controls, whereas medullary blood flow was not significantly changed. Dilazep did not affect total renal blood flow in any of the groups but decreased cortical blood flow in diabetics, resulting in decreased filtration fraction by dilazep in diabetics. Pretreatment with the adenosine A2a antagonist ZM241385 prevented intrarenal dilazep-mediated effects on glomerular filtration rate and filtration fraction in diabetics. In conclusion, enhancing intrarenal adenosine signaling by dilazep normalizes diabetes-induced glomerular hyperfiltration at least in part by activation of adenosine A2a receptors.


Assuntos
Diabetes Mellitus , Nefropatias , Ratos , Animais , Masculino , Ratos Sprague-Dawley , Dilazep/farmacologia , Adenosina/farmacologia , Glomérulos Renais , Rim , Taxa de Filtração Glomerular
2.
J Physiol ; 598(23): 5573-5587, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32857872

RESUMO

KEY POINTS: Reducing Na+ intake reduces the partial pressure of oxygen in the renal cortex and activates the renin-angiotensin-aldosterone system. In the absence of high blood pressure, these consequences of dietary Na+ reduction may be detrimental for the kidney. In a normotensive animal experimental model, reducing Na+ intake for 2 weeks increased renal oxygen consumption, which was normalized by mineralocorticoid receptor blockade. Furthermore, blockade of the angiotensin II AT1 receptor restored cortical partial pressure of oxygen by improving oxygen delivery. This shows that increased activity of the renin-angiotensin-aldosterone system contributes to increased oxygen metabolism in the kidney after 2 weeks of a low Na+ diet. The results provide insights into dietary Na+ restriction in the absence of high blood pressure, and its consequences for the kidney. ABSTRACT: Reduced Na+ intake reduces the PO2 (partial pressure of oxygen) in the renal cortex. Upon reduced Na+ intake, reabsorption along the nephron is adjusted with activation of the renin-angiotensin-aldosterone system (RAAS). Thus, we studied the effect of reduced Na+ intake on renal oxygen homeostasis and function in rats, and the impact of intrarenal angiotensin II AT1 receptor blockade using candesartan and mineralocorticoid receptor blockade using canrenoic acid potassium salt (CAP). Male Sprague-Dawley rats were fed standard rat chow containing normal (0.25%) and low (0.025%) Na+ for 2 weeks. The animals were anaesthetized (thiobutabarbital 120 mg kg-1 ) and surgically prepared for kidney oxygen metabolism and function studies before and after acute intrarenal arterial infusion of candesartan (4.2 µg kg-1 ) or intravenous infusion of CAP (20 mg kg-1 ). Baseline mean arterial pressure and renal blood flow were similar in both dietary groups. Fractional Na+ excretion and cortical oxygen tension were lower and renal oxygen consumption was higher in low Na+ groups. Neither candesartan nor CAP affected arterial pressure. Renal blood flow and cortical oxygen tension increased in both groups after candesartan in the low Na+ group. Fractional Na+ excretion was increased and oxygen consumption reduced in the low Na+ group after CAP. These results suggest that blockade of angiotensin II AT1 receptors has a major impact upon oxygen delivery during normal and low Na+ conditions, while aldosterone receptors mainly affect oxygen metabolism following 2 weeks of a low Na+ diet.


Assuntos
Angiotensina II , Receptores de Mineralocorticoides , Aldosterona/metabolismo , Angiotensina II/metabolismo , Animais , Pressão Sanguínea , Dieta , Rim/metabolismo , Masculino , Oxigênio/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Sistema Renina-Angiotensina
3.
Am J Physiol Renal Physiol ; 314(3): F439-F444, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29092848

RESUMO

About one-third of patients with type 1 diabetes develops kidney disease. The mechanism is largely unknown, but intrarenal hypoxia has been proposed as a unifying mechanism for chronic kidney disease, including diabetic nephropathy. The endothelin system has recently been demonstrated to regulate oxygen availability in the diabetic kidney via a pathway involving endothelin type A receptors (ETA-R). These receptors mainly mediate vasoconstriction and tubular sodium retention, and inhibition of ETA-R improves intrarenal oxygenation in the diabetic kidney. Endothelin type B receptors (ETB-R) can induce vasodilation of the renal vasculature and also regulate tubular sodium handling. However, the role of ETB-R in kidney oxygen homeostasis is unknown. The effects of acute intrarenal ETB-R activation (sarafotoxin 6c for 30-40 min; 0.78 pmol/h directly into the renal artery) on kidney function and oxygen metabolism were investigated in normoglycemic controls and insulinopenic male Sprague-Dawley rats administered streptozotocin (55 mg/kg) 2 wk before the acute experiments. Intrarenal activation of ETB-R improved oxygenation in the hypoxic diabetic kidney. However, the effects on diabetes-induced increased kidney oxygen consumption could not explain the improved oxygenation. Rather, the improved kidney oxygenation was due to hemodynamic effects increasing oxygen delivery without increasing glomerular filtration or tubular sodium load. In conclusion, increased ETB-R signaling in the diabetic kidney improves intrarenal tissue oxygenation due to increased oxygen delivery secondary to increased renal blood flow.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Nefropatias Diabéticas/tratamento farmacológico , Hemodinâmica/efeitos dos fármacos , Rim/efeitos dos fármacos , Oxigênio/sangue , Receptor de Endotelina B/agonistas , Circulação Renal/efeitos dos fármacos , Venenos de Víboras/farmacologia , Animais , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/fisiopatologia , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/fisiopatologia , Rim/metabolismo , Rim/fisiopatologia , Masculino , Ratos Sprague-Dawley , Receptor de Endotelina B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estreptozocina
4.
Am J Physiol Renal Physiol ; 314(5): F864-F872, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28971989

RESUMO

An increased kidney oxygen consumption causing tissue hypoxia has been suggested to be a common pathway toward chronic kidney disease. The mammalian target of rapamycin (mTOR) regulates cell proliferation and mitochondrial function. mTOR inhibitors (e.g., rapamycin) are used clinically to prevent graft rejection. mTOR has been identified as a key player in diabetes, which has stimulated the use of mTOR inhibitors to counter diabetic nephropathy. However, the effect of mTOR inhibition on kidney oxygen consumption is unknown. Therefore, we investigated the effects of mTOR inhibition on in vivo kidney function, oxygen homeostasis, and glomerular permeability. Control and streptozotocin-induced diabetic rats were chronically treated with rapamycin, and the functional consequences were studied 14 days thereafter. In both groups, mTOR inhibition induced mitochondrial uncoupling, resulting in increased total kidney oxygen consumption and decreased intrarenal oxygen availability. Concomitantly, mTOR inhibition induced tubular injury, as estimated from urinary excretion of kidney injury molecule-1 (KIM-1) and reduced urinary protein excretion. The latter corresponded to reduced sieving coefficient for large molecules. In conclusion, mTOR inhibition induces mitochondrial dysfunction leading to decreased oxygen availability in normal and diabetic kidneys, which translates into increased KIM-1 in the urine. Reduced proteinuria after mTOR inhibition is an effect of reduced glomerular permeability for large molecules. Since hypoxia has been suggested as a common pathway in the development of chronic kidney disease, mTOR inhibition to patients with preexisting nephropathy should be used with caution, since it may accelerate the progression of the disease.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Nefropatias Diabéticas/complicações , Taxa de Filtração Glomerular/efeitos dos fármacos , Glomérulos Renais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Inibidores de Proteínas Quinases/toxicidade , Insuficiência Renal Crônica/induzido quimicamente , Sirolimo/toxicidade , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Moléculas de Adesão Celular/metabolismo , Hipóxia Celular , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Progressão da Doença , Glomérulos Renais/enzimologia , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/enzimologia , Túbulos Renais/patologia , Masculino , Mitocôndrias/enzimologia , Estresse Oxidativo/efeitos dos fármacos , Ratos Sprague-Dawley , Insuficiência Renal Crônica/enzimologia , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
5.
Am J Physiol Renal Physiol ; 312(2): F278-F283, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27927650

RESUMO

Diabetes mellitus is associated with decreased nitric oxide bioavailability thereby affecting renal blood flow regulation. Previous reports have demonstrated that cellular uptake of l-arginine is rate limiting for nitric oxide production and that plasma l-arginine concentration is decreased in diabetes. We therefore investigated whether regional renal blood flow regulation is affected by cellular l-arginine uptake in streptozotocin-induced diabetic rats. Rats were anesthetized with thiobutabarbital, and the left kidney was exposed. Total, cortical, and medullary renal blood flow was investigated before and after renal artery infusion of increasing doses of either l-homoarginine to inhibit cellular uptake of l-arginine or Nω-nitro- l-arginine methyl ester (l-NAME) to inhibit nitric oxide synthase. l-Homoarginine infusion did not affect total or cortical blood flow in any of the groups, but caused a dose-dependent reduction in medullary blood flow. l-NAME decreased total, cortical and medullary blood flow in both groups. However, the reductions in medullary blood flow in response to both l-homoarginine and l-NAME were more pronounced in the control groups compared with the diabetic groups. Isolated cortical tubular cells displayed similar l-arginine uptake capacity whereas medullary tubular cells isolated from diabetic rats had increased l-arginine uptake capacity. Diabetics had reduced l-arginine concentrations in plasma and medullary tissue but increased l-arginine concentration in cortical tissue. In conclusion, the reduced l-arginine availability in plasma and medullary tissue in diabetes results in reduced nitric oxide-mediated regulation of renal medullary hemodynamics. Cortical blood flow regulation displays less dependency on extracellular l-arginine and the upregulated cortical tissue l-arginine may protect cortical hemodynamics in diabetes.


Assuntos
Arginina/metabolismo , Diabetes Mellitus Experimental/metabolismo , Medula Renal/irrigação sanguínea , Circulação Renal/fisiologia , Animais , Transporte Biológico/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Homoarginina/farmacologia , Medula Renal/efeitos dos fármacos , Medula Renal/metabolismo , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos
6.
J Am Soc Nephrol ; 26(2): 328-38, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25183809

RESUMO

Hyperglycemia results in increased oxygen consumption and decreased oxygen tension in the kidney. We tested the hypothesis that activation of hypoxia-inducible factors (HIFs) protects against diabetes-induced alterations in oxygen metabolism and kidney function. Experimental groups consisted of control and streptozotocin-induced diabetic rats treated with or without chronic cobalt chloride to activate HIFs. We elucidated the involvement of oxidative stress by studying the effects of acute administration of the superoxide dismutase mimetic tempol. Compared with controls, diabetic rats displayed tissue hypoxia throughout the kidney, glomerular hyperfiltration, increased oxygen consumption, increased total mitochondrial leak respiration, and decreased tubular sodium transport efficiency. Diabetic kidneys showed proteinuria and tubulointerstitial damage. Cobalt chloride activated HIFs, prevented the diabetes-induced alterations in oxygen metabolism, mitochondrial leak respiration, and kidney function, and reduced proteinuria and tubulointerstitial damage. The beneficial effects of tempol were less pronounced after activation of HIFs, indicating improved oxidative stress status. In conclusion, activation of HIFs prevents diabetes-induced alteration in kidney oxygen metabolism by normalizing glomerular filtration, which reduces tubular electrolyte load, preventing mitochondrial leak respiration and improving tubular transport efficiency. These improvements could be related to reduced oxidative stress and account for the reduced proteinuria and tubulointerstitial damage. Thus, pharmacologic activation of the HIF system may prevent development of diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/prevenção & controle , Nefropatias Diabéticas/fisiopatologia , Fator 1 Induzível por Hipóxia/fisiologia , Animais , Antioxidantes/farmacologia , Cobalto/farmacologia , Óxidos N-Cíclicos/farmacologia , Modelos Animais de Doenças , Taxa de Filtração Glomerular/efeitos dos fármacos , Taxa de Filtração Glomerular/fisiologia , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Ratos , Ratos Sprague-Dawley , Marcadores de Spin
7.
Am J Physiol Renal Physiol ; 309(3): F227-34, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26041448

RESUMO

Early stage diabetic nephropathy is characterized by glomerular hyperfiltration and reduced renal tissue Po2. Recent observations have indicated that increased tubular Na(+)-glucose linked transport (SGLT) plays a role in the development of diabetes-induced hyperfiltration. The aim of the present study was to determine how inhibition of SLGT impacts upon Po2 in the diabetic rat kidney. Diabetes was induced by streptozotocin in Sprague-Dawley rats 2 wk before experimentation. Renal hemodynamics, excretory function, and renal O2 homeostasis were measured in anesthetized control and diabetic rats during baseline and after acute SGLT inhibition using phlorizin (200 mg/kg ip). Baseline arterial pressure was similar in both groups and unaffected by SGLT inhibition. Diabetic animals displayed reduced baseline Po2 in both the cortex and medulla. SGLT inhibition improved cortical Po2 in the diabetic kidney, whereas it reduced medullary Po2 in both groups. SGLT inhibition reduced Na(+) transport efficiency [tubular Na(+) transport (TNa)/renal O2 consumption (Qo2)] in the control kidney, whereas the already reduced TNa/Qo2 in the diabetic kidney was unaffected by SGLT inhibition. In conclusion, these data demonstrate that when SGLT is inhibited, renal cortex Po2 in the diabetic rat kidney is normalized, which implies that increased proximal tubule transport contributes to the development of hypoxia in the diabetic kidney. The reduction in medullary Po2 in both control and diabetic kidneys during the inhibition of proximal Na(+) reabsorption suggests the redistribution of active Na(+) transport to less efficient nephron segments, such as the medullary thick ascending limb, which results in medullary hypoxia.


Assuntos
Nefropatias Diabéticas/metabolismo , Hipóxia/metabolismo , Córtex Renal/metabolismo , Medula Renal/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Proteínas de Transporte de Sódio-Glucose/antagonistas & inibidores , Anestesia , Animais , Pressão Arterial/efeitos dos fármacos , Diabetes Mellitus Experimental , Córtex Renal/efeitos dos fármacos , Masculino , Florizina/farmacologia , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo , Urodinâmica/efeitos dos fármacos
8.
Am J Physiol Renal Physiol ; 306(10): F1171-8, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24623147

RESUMO

One-third of diabetes mellitus patients develop diabetic nephropathy, and with underlying mechanisms unknown it is imperative that diabetic animal models resemble human disease. The present study investigated the susceptibility to develop diabetic nephropathy in four commonly used and commercially available mouse strains with type 1 diabetes to determine the suitability of each strain. Type 1 diabetes was induced in C57Bl/6, NMRI, BALB/c, and 129Sv mice by alloxan, and conscious glomerular filtration rate, proteinuria, and oxidative stress levels were measured in control and diabetic animals at baseline and after 5 and 10 wk. Histological alterations were analyzed using periodic acid-Schiff staining. Diabetic C57Bl/6 displayed increased glomerular filtration rate, i.e., hyperfiltration, whereas all other parameters remained unchanged. Diabetic NMRI developed the most pronounced hyperfiltration as well as increased oxidative stress and proteinuria but without glomerular damage. Diabetic BALB/c did not develop hyperfiltration but presented with pronounced proteinuria, increased oxidative stress, and glomerular damage. Diabetic 129Sv displayed proteinuria and increased oxidative stress without glomerular hyperfiltration or damage. However, all strains displayed intrastrain correlation between oxidative stress and proteinuria. In conclusion, diabetic C57Bl/6 and NMRI both developed glomerular hyperfiltration but neither presented with histological damage, although NMRI developed low-degree proteinuria. Thus these strains may be suitable when investigating the mechanism causing hyperfiltration. Neither BALB/c nor 129Sv developed hyperfiltration although both developed pronounced proteinuria. However, only BALB/c developed detectable histological damage. Thus BALB/c may be suitable when studying the roles of proteinuria and histological alterations for the progression of diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 1/complicações , Nefropatias Diabéticas/epidemiologia , Nefropatias Diabéticas/fisiopatologia , Suscetibilidade a Doenças/epidemiologia , Suscetibilidade a Doenças/fisiopatologia , Aloxano/efeitos adversos , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Tipo 1/induzido quimicamente , Modelos Animais de Doenças , Taxa de Filtração Glomerular/fisiologia , Incidência , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Estresse Oxidativo/fisiologia , Proteinúria/epidemiologia , Proteinúria/fisiopatologia , Fatores de Risco , Especificidade da Espécie
9.
Am J Physiol Renal Physiol ; 304(5): F614-22, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23283998

RESUMO

Increased angiotensin II (ANG II) or adenosine can potentiate each other in the regulation of renal hemodynamics and tubular function. Diabetes is characterized by hyperfiltration, yet the roles of ANG II and adenosine receptors for controlling baseline renal blood flow (RBF) or tubular Na(+) handling in diabetes is presently unknown. Accordingly, the changes in their functions were investigated in control and 2-wk streptozotocin-diabetic rats after intrarenal infusion of the ANG II AT1 receptor antagonist candesartan, the adenosine A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), or their combination. Compared with controls, the baseline blood pressure, RBF, and renal vascular resistance (RVR) were similar in diabetics, whereas the glomerular filtration rate (GFR) and filtration fraction (FF) were increased. Candesartan, DPCPX, or the combination increased RBF and decreased RVR similarly in all groups. In controls, the GFR was increased by DPCPX, but in diabetics, it was decreased by candesartan. The FF was decreased by candesartan and DPCPX, independently. DPCPX caused the most pronounced increase in fractional Na(+) excretion in both controls and diabetics, whereas candesartan or the combination only affected fractional Li(+) excretion in diabetics. These results suggest that RBF, via a unifying mechanism, and tubular function are under strict tonic control of both ANG II and adenosine in both control and diabetic kidneys. Furthermore, increased vascular AT1 receptor activity is a contribution to diabetes-induced hyperfiltration independent of any effect of adenosine A1 receptors.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Rim/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Antagonistas do Receptor A1 de Adenosina/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Benzimidazóis/farmacologia , Compostos de Bifenilo , Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/fisiopatologia , Relação Dose-Resposta a Droga , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/irrigação sanguínea , Rim/metabolismo , Rim/fisiopatologia , Masculino , Ratos , Circulação Renal/efeitos dos fármacos , Tetrazóis/farmacologia , Resistência Vascular/efeitos dos fármacos , Xantinas/farmacologia
10.
Adv Exp Med Biol ; 765: 185-193, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22879032

RESUMO

Sustained hyperglycemia is closely associated with increased risk to develop nephropathy. We have previously reported alterations in the intrarenal oxygen metabolism already after the early onset of diabetes. Furthermore, formation of advanced glycation end-products (AGE) is postulated as a major contributor to diabetic nephropathy. We therefore investigated the possible relationship between altered oxygen metabolism and AGE in diabetic kidneys.Normoglycemic and streptozotocin-diabetic rats with and without chronic treatment with aminoguanidine (AGE inhibitor; 600 mg/kg bw/24 h in drinking water) or L-N(6)-(1-Iminoethyl)lysine (L-NIL, iNOS inhibitor, 1 mg/kg bw/24 h in drinking water) were studied 2 weeks after induction of diabetes. Glomerular filtration rate (GFR) was estimated by inulin clearance, oxygen tension (pO(2)) and interstitial pH by microelectrodes and regional renal blood flow (RBF) by laser-Doppler. Histological changes were evaluated on fixed tissue.Glomerular hyperfiltration was unaffected by aminoguanidine, whereas L-NIL normalized GFR in diabetic rats. pO(2) and interstitial pH, but not RBF, were lower in both kidney cortex and medulla compared to control rats, but was unaffected by both chronic treatments. Urinary protein excretion was higher in diabetic rats and unaffected by L-NIL, whereas aminoguanidine paradoxically increased this parameter. Damage scores were similar in all groups.In conclusion, diabetes-induced alterations in intrarenal oxygen metabolism are independent of the AGE pathway, and precede any morphological changes. These findings highlight the early stage of diabetes as being a metabolic disorder also in the kidney.


Assuntos
Complicações do Diabetes/etiologia , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/etiologia , Produtos Finais de Glicação Avançada/metabolismo , Hipóxia/patologia , Rim/patologia , Animais , Complicações do Diabetes/tratamento farmacológico , Complicações do Diabetes/metabolismo , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Inibidores Enzimáticos/uso terapêutico , Taxa de Filtração Glomerular , Guanidinas/uso terapêutico , Hipóxia/metabolismo , Rim/metabolismo , Testes de Função Renal , Masculino , Óxido Nítrico Sintase/antagonistas & inibidores , Ratos , Ratos Endogâmicos WF
11.
Adv Exp Med Biol ; 789: 9-14, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23852470

RESUMO

Intrarenal tissue hypoxia is an acknowledged common pathway to end-stage renal disease in clinically common conditions associated with development of chronic kidney disease, such as diabetes and hypertension. In diabetic kidneys, increased oxygen metabolism mediated by mitochondrial uncoupling results in decreased kidney oxygen tension (PO2) and contributes to the development of diabetic nephropathy. The present study investigated whether increased intrarenal oxygen metabolism per se can cause intrarenal tissue hypoxia and kidney damage, independently of confounding factors such as hyperglycemia and oxidative stress. Male Sprague-Dawley rats were untreated or treated with either triiodothyronine (T3, 10 g/kg bw/day, subcutaneously for 10 days) or the mitochondria uncoupler dinitrophenol (DNP, 30 mg/kg bw/day, oral gavage for 14 days), after which in vivo kidney function was evaluated in terms of glomerular filtration rate (GFR, inulin clearance), renal blood flow (RBF, Transonic, PAH clearance), cortical PO2 (Clark-type electrodes), kidney oxygen consumption (QO2), and proteinuria. Administration of both T3 and DNP increased kidney QO2 and decreased PO2 which resulted in proteinuria. However, GFR and RBF were unaltered by either treatment. The present study demonstrates that increased kidney metabolism per se can cause intrarenal tissue hypoxia which results in proteinuria. Increased kidney QO2 and concomitantly reduced PO2 may therefore be a mechanism for the development of chronic kidney disease and progression to end-stage renal disease.


Assuntos
Dinitrofenóis/farmacologia , Córtex Renal/efeitos dos fármacos , Córtex Renal/metabolismo , Nefropatias/metabolismo , Tri-Iodotironina/farmacologia , Animais , Hipóxia Celular/fisiologia , Córtex Renal/patologia , Nefropatias/patologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Oxigênio/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Ratos , Ratos Sprague-Dawley
12.
Am J Physiol Regul Integr Comp Physiol ; 303(1): R39-47, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22461175

RESUMO

Glomerular filtration rate (GFR) and renal blood flow (RBF) are normally kept constant via renal autoregulation. However, early diabetes results in increased GFR and the potential mechanisms are debated. Tubuloglomerular feedback (TGF) inactivation, with concomitantly increased RBF, is proposed but challenged by the finding of glomerular hyperfiltration in diabetic adenosine A(1) receptor-deficient mice, which lack TGF. Furthermore, we consistently find elevated GFR in diabetes with only minor changes in RBF. This may relate to the use of a lower streptozotocin dose, which produces a degree of hyperglycemia, which is manageable without supplemental suboptimal insulin administration, as has been used by other investigators. Therefore, we examined the relationship between RBF and GFR in diabetic rats with (diabetes + insulin) and without suboptimal insulin administration (untreated diabetes). As insulin can affect nitric oxide (NO) release, the role of NO was also investigated. GFR, RBF, and glomerular filtration pressures were measured. Dynamic RBF autoregulation was examined by transfer function analysis between arterial pressure and RBF. Both diabetic groups had increased GFR (+60-67%) and RBF (+20-23%) compared with controls. However, only the diabetes + insulin group displayed a correlation between GFR and RBF (R(2) = 0.81, P < 0.0001). Net filtration pressure was increased in untreated diabetes compared with both other groups. The difference between untreated and insulin-treated diabetic rats disappeared after administering N(ω)-nitro-l-arginine methyl ester to inhibit NO synthase and subsequent NO release. In conclusion, mechanisms causing diabetes-induced glomerular hyperfiltration are animal model-dependent. Supplemental insulin administration results in a RBF-dependent mechanism, whereas elevated GFR in untreated diabetes is mediated primarily by a tubular event. Insulin-induced NO release partially contributes to these differences.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Taxa de Filtração Glomerular/efeitos dos fármacos , Insulina/farmacologia , Rim/irrigação sanguínea , Fluxo Sanguíneo Regional/efeitos dos fármacos , Animais , Glicemia/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Diabetes Mellitus Experimental/induzido quimicamente , Modelos Animais de Doenças , Taxa de Filtração Glomerular/fisiologia , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/fisiologia , Estreptozocina/efeitos adversos
13.
Acta Physiol (Oxf) ; 233(1): e13668, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33900001

RESUMO

AIM: Hypoxia-inducible factors (HIFs) are O2 -sensitive transcription factors that regulate multiple biological processes which are essential for cellular adaptation to hypoxia. Small molecule inhibitors of HIF-prolyl hydroxylase domain (PHD) dioxygenases (HIF-PHIs) activate HIF-dependent transcriptional programs and have broad clinical potential. HIF-PHIs are currently in global late-stage clinical development for the treatment of anaemia associated with chronic kidney disease. Although the effects of hypoxia on renal haemodynamics and function have been studied in animal models and in humans living at high altitude, the effects of pharmacological HIF activation on renal haemodynamics, O2 metabolism and metabolic efficiency are not well understood. METHODS: Using a cross-sectional study design, we investigated renal haemodynamics, O2 metabolism, gene expression and NO production in healthy rats treated with different doses of HIF-PHIs roxadustat or molidustat compared to vehicle control. RESULTS: Systemic administration of roxadustat or molidustat resulted in a dose-dependent reduction in renovascular resistance (RVR). This was associated with increased glomerular filtration rate (GFR), urine flow and tubular sodium transport rate (TNa ). Although both total O2 delivery and TNa were increased, more O2 was extracted per transported sodium in rats treated with high-doses of HIF-PHIs, suggesting a reduction in metabolic efficiency. Changes in RVR and GFR were associated with increased nitric oxide (NO) generation and substantially suppressed by pharmacological inhibition of NO synthesis. CONCLUSIONS: Our data provide mechanistic insights into dose-dependent effects of short-term pharmacological HIF activation on renal haemodynamics, glomerular filtration and O2 metabolism and identify NO as a major mediator of these effects.


Assuntos
Fenômenos Biológicos , Insuficiência Renal Crônica , Animais , Estudos Transversais , Prolina Dioxigenases do Fator Induzível por Hipóxia , Óxido Nítrico , Prolil Hidroxilases , Ratos
14.
Am J Physiol Renal Physiol ; 298(2): F416-20, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19923416

RESUMO

Nitric oxide (NO) is a potent regulator of both vascular tone and cellular oxygen consumption (Q(O(2)). Diabetic kidneys have reduced NO availability and increased Q(O(2)). However, the exact nitric oxide synthase (NOS) isoform regulating Q(O(2)), hemodynamics, and excretory function in the diabetic kidney remains unclear. We therefore investigated the effects of both selective neuronal NOS (NOS1) inhibition and nonselective NOS inhibition. Oxygen utilization, electrolyte transport efficiency [tubular Na(+) transport (T(Na))/Q(O(2))], renal blood flow (RBF), glomerular filtration rate (GFR), and mean arterial pressure (MAP) were measured in vivo in control and streptozotocin-diabetic rats before and after administration of the selective NOS1 inhibitor S-methyl-L-thiocitrulline (SMTC) or the nonselective NOS inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME). Diabetic rats had higher baseline Q(O(2)) and GFR than control rats, although RBF was similar in the groups. SMTC and L-NAME increased Q(O(2)) and reduced T(Na)/Q(O(2)) only in the diabetic animals, whereas both inhibitors increased MAP and reduced RBF in both groups. GFR was reduced by L-NAME, but SMTC had no effect in either group. Carbachol increased RBF and decreased MAP in SMTC-treated rats, whereas it had no effect in L-NAME-treated rats, indicating that SMTC selectively inhibited NOS1. In conclusion, NO regulates RBF and GFR similarly in both control and diabetic rats. However, selective NOS1 inhibition increased Qo(2) and reduced T(Na)/Q(O(2)) in the diabetic rat kidney, indicating a pivotal role of NO produced by NOS1 in maintaining control of Q(O(2)) and tissue oxygenation in these kidneys.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Eletrólitos/metabolismo , Rim/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico/metabolismo , Consumo de Oxigênio , Animais , Transporte Biológico/efeitos dos fármacos , Pressão Sanguínea , Citrulina/análogos & derivados , Citrulina/farmacologia , Inibidores Enzimáticos/farmacologia , Taxa de Filtração Glomerular , Rim/irrigação sanguínea , Túbulos Renais/metabolismo , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Ratos , Ratos Endogâmicos WF , Fluxo Sanguíneo Regional , Sódio/metabolismo , Tioureia/análogos & derivados , Tioureia/farmacologia
15.
Am J Physiol Renal Physiol ; 299(2): F380-6, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20519374

RESUMO

In addition to causing uremic symptoms, uremic toxins accelerate the progression of renal failure. To elucidate the pathophysiology of uremic states, we investigated the effect of indoxyl sulfate (IS), a representative uremic toxin, on oxygen metabolism in tubular cells. We demonstrated an increase in oxygen consumption by IS in freshly isolated rat and human proximal tubules. Studies utilizing ouabain, the Na-K-ATPase inhibitor, and apocynin, the NADPH oxidase inhibitor, as well as the in vivo gene-silencing approach to knock down p22(phox) showed that the increase in tubular oxygen consumption by IS is dependent on Na-K-ATPase and oxidative stress. We investigated whether the enhanced oxygen consumption led to subsequent hypoxia of the kidney. An increase in serum IS concentrations in rats administered indole was associated with a decrease in renal oxygenation (8 h). The remnant kidney in rats developed hypoxia at 16 wk. Treatment of the rats with AST-120, an oral adsorbent that removes uremic toxins, reduced serum IS levels and improved oxygenation of the kidney. Amelioration of hypoxia in the remnant kidney was associated with better renal functions and less histological injury. Reduction of serum IS levels also led to a decrease in oxidative stress in the kidney. Our ex vivo and in vivo studies implicated that uremic states may deteriorate renal dysfunction via dysregulating oxygen metabolism in tubular cells. The abnormal oxygen metabolism in tubular cells by uremic toxins was, at least in part, mediated by oxidative stress.


Assuntos
Indicã/metabolismo , Falência Renal Crônica/metabolismo , Túbulos Renais Proximais/metabolismo , Estresse Oxidativo , Consumo de Oxigênio , Uremia/metabolismo , Acetofenonas/farmacologia , Animais , Carbono/farmacologia , Hipóxia Celular , Modelos Animais de Doenças , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Humanos , Indicã/sangue , Indóis , Falência Renal Crônica/etiologia , Falência Renal Crônica/fisiopatologia , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/fisiopatologia , Masculino , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , Nefrectomia , Ouabaína/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Óxidos/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , ATPase Trocadora de Sódio-Potássio/metabolismo , Fatores de Tempo , Uremia/etiologia , Uremia/fisiopatologia
16.
Diabetes Metab Res Rev ; 26(5): 349-56, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20583310

RESUMO

BACKGROUND: Sustained hyperglycaemia induces increased renal oxygen consumption resulting in reduced oxygen availability in the diabetic kidney. We investigated the roles of the nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase and the neuronal nitric oxide synthase (nNOS) for the increased oxygen consumption in streptozotocin-diabetic rats. METHODS: Oxygen consumption was measured in isolated proximal tubular cells (PTC) from streptozotocin-induced diabetic rats (n = 7-9 per group) with and without chronic treatment with apocynin, a NADPH-oxidase inhibitor, or S-methyl-L-thiocitrulline (SMTC), a selective nNOS inhibitor, or a combination of the two and the results were compared to normoglycaemic controls (n = 10). Oxidative stress was estimated from thiobarbituric acid reactive substances and protein expression measured by Western blot. RESULTS: Proximal tubular cells from untreated diabetic rats had increased oxygen consumption compared to controls (40.6 +/- 7.9 versus 10.9 +/- 2.0 nmol/mg protein/min). All treatments reduced the diabetes-induced increase in oxygen consumption (apocynin 10.5 +/- 1.7, SMTC 19.7 +/- 3.0 and apocynin + SMTC 21.6 +/- 3.6 nmol/mg protein/min). Neither apocynin nor SMTC had any effect on the oxygen consumption in cells pre-incubated with ouabain, an inhibitor of active electrolyte transport. Oxidative stress was elevated in the diabetic kidney and inhibited by all treatments. The increased oxygen consumption by diabetic proximal tubular cells correlated with increased protein expressions of p47(phox) and nNOS and the treatments prevented these increases. CONCLUSIONS: Diabetes induces oxidative stress, which increases oxygen consumption in proximal tubular cells. Inhibition of either NADPH-oxidase or nNOS prevented the increased oxygen consumption. The effect of blocking both these enzymes was less than additive suggesting overlapping pathways which warrant further studies.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Nefropatias Diabéticas/metabolismo , NADPH Oxidases/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Acetofenonas/farmacologia , Animais , Citrulina/análogos & derivados , Citrulina/farmacologia , Rim/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , NADPH Oxidases/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Ratos , Ratos Endogâmicos WF , Tioureia/análogos & derivados , Tioureia/farmacologia
17.
Biochim Biophys Acta ; 1777(7-8): 935-40, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18439413

RESUMO

We have previously reported increased O(2) consumption unrelated to active transport by tubular cells and up-regulated mitochondrial uncoupling protein (UCP)-2 expressions in diabetic kidneys. It is presently unknown if the increased UCP-2 levels in the diabetic kidney results in mitochondrial uncoupling and increased O(2) consumption, which we therefore investigated in this study. The presence of UCP-2 in proximal tubular cells was confirmed by immunohistochemistry and found to be increased (western blot) in homogenized tissue and isolated mitochondria from kidney cortex of diabetic rats. Isolated proximal tubular cells had increased total and ouabain-insensitive O(2) consumption compared to controls. Isolated mitochondria from diabetic animals displayed increased glutamate-stimulated O(2) consumption (in the absence of ADP and during inhibition of the ATP-synthase by oligomycin) compared to controls. Guanosine diphosphate, an UCP inhibitor, and bovine serum albumin which removes fatty acids that are essential for UCP-2 uncoupling activity, independently prevented the increased glutamate-stimulated O(2) consumption in mitochondria from diabetic animals. In conclusion, diabetic rats have increased mitochondrial UCP-2 expression in renal proximal tubular cells, which results in mitochondrial uncoupling and increased O(2) consumption. This mechanism may be protective against diabetes-induced oxidative stress, but will increase O(2) usage. The subsequently reduced O(2) availability may contribute to diabetes-induced progressive kidney damage.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Canais Iônicos/metabolismo , Túbulos Renais Proximais/fisiopatologia , Proteínas Mitocondriais/metabolismo , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Hiperglicemia/patologia , Hiperglicemia/fisiopatologia , Insulina/uso terapêutico , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Consumo de Oxigênio , Ratos , Ratos Endogâmicos WF , Proteína Desacopladora 2 , Regulação para Cima
18.
Am J Physiol Renal Physiol ; 297(5): F1265-72, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19741019

RESUMO

C-peptide reduces diabetes-induced glomerular hyperfiltration in diabetic patients and experimental animal models. However, the mechanisms mediating the beneficial effect of C-peptide remain unclear. We investigated whether altered renal afferent-efferent arteriole tonus or alterations in tubular Na+ transport (T(Na)) in response to C-peptide administration mediate the reduction of diabetes-induced glomerular hyperfiltration. Glomerular filtration rate, filtration fraction, total and cortical renal blood flow, total kidney O2 consumption (QO2), T(Na), fractional Na+ and Li+ excretions, and tubular free-flow and stop-flow pressures were measured in anesthetized adult male normoglycemic and streptozotocin-diabetic Sprague-Dawley rats. The specific effect of C-peptide on transport-dependent QO2 was investigated in vitro in freshly isolated proximal tubular cells. C-peptide reduced glomerular filtration rate (-24%), stop-flow pressure (-8%), and filtration fraction (-17%) exclusively in diabetic rats without altering renal blood flow. Diabetic rats had higher baseline T(Na) (+40%), which was reduced by C-peptide. Similarly, C-peptide increased fractional Na+ (+80%) and Li+ (+47%) excretions only in the diabetic rats. None of these parameters was affected by vehicle treatments in either group. Baseline QO2 was 37% higher in proximal tubular cells from diabetic rats than controls and was normalized by C-peptide. C-peptide had no effect on ouabain-pretreated diabetic cells from diabetic rats. C-peptide reduced diabetes-induced hyperfiltration via a net dilation of the efferent arteriole and inhibition of tubular Na+ reabsorption, both potent regulators of the glomerular net filtration pressure. These findings provide new mechanistic insight into the beneficial effects of C-peptide on diabetic kidney function.


Assuntos
Peptídeo C/farmacologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Taxa de Filtração Glomerular/efeitos dos fármacos , Túbulos Renais/metabolismo , Sódio/metabolismo , Vasodilatação/efeitos dos fármacos , Animais , Arteríolas/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Túbulos Renais/efeitos dos fármacos , Lítio/metabolismo , Masculino , Tono Muscular/efeitos dos fármacos , NG-Nitroarginina Metil Éster/farmacologia , Ouabaína/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos
19.
Adv Exp Med Biol ; 645: 213-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19227474

RESUMO

The renal medulla has been implicated as a key target for contrast media-induced nephropathy (CIN). Although the effects of contrast media (CM) on whole kidney blood flow are well characterized, the effect of CM on renal medullary blood flow has been controversial. It has been reported that an extremely high dose of a high osmolar CM (iothalamate; 2900 mg I/kg bw) injected rapidly increased the renal outer medullary blood flow (OMBF). However, more clinical relevant doses consistently result in a sustained decrease in medullary blood flow. Furthermore, simultaneous measurements using both laser-Doppler flowmetry and hydrogen washout yield similar results of a decrease in OMBF after CM administration. CM induced a transient 28% decrease in the laser-Doppler signal from the outer medulla, while the hydrogen washout rate in the same region was reduced by approximately 50%. Furthermore, CM administration consistently results in decreased medullary oxygen tension (PO2). The renal medulla works already during normal physiological conditions at the verge of hypoxia, and the majority of the studies published so far are in agreement with the hypothesis that CIN may have its origin in a further reduction in blood flow and/or oxygen availability of this region of the kidney.


Assuntos
Meios de Contraste/efeitos adversos , Iodo/efeitos adversos , Nefropatias/induzido quimicamente , Circulação Renal/efeitos dos fármacos , Adenosina/metabolismo , Animais , Nefropatias/metabolismo
20.
Adv Exp Med Biol ; 645: 199-204, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19227472

RESUMO

By applying invasive techniques for direct measurements of oxygen tension, we have reported decreased kidney oxygenation in experimental diabetes in rats. However, the non-invasive MRI technique utilizing the BOLD effect provides several advantages with the possibility to perform repetitive measurements in the same animals and in human subjects. In this study, we applied a modified single gradient echo micro-imaging sequence to detect the BOLD effect in kidneys of diabetic rats and compared the results to normoglycemic controls. All measurements were performed on inactin-anaesthetized adult male Wistar Furth rats. Diabetes was induced by streptozotocin (45 mg/kg) 14 days prior to MRI-analysis. Sixteen T2*-weighted image records (B0=1.5 T) were performed using radiofrequency spoiled gradient echo sequence with 2.6 ms step increments of TE (TE1=12 ms), while TR (75 ms) and bandwidth per pixel (71.4 Hz) were kept constant. T2* maps were computed by mono-exponential fitting of the pixel intensities. Relaxation rates R2* (1/T2*) in cortex and outer stripe of the outer medulla were similar in both groups (cortex for controls 22.3 +/- 0.4 vs. diabetics 23.1 +/- 1.8 Hz and outer stripe of outer medulla for controls 24.9 +/- 0.4 vs. diabetics 26.4 +/- 1.8 Hz; n=4 in both groups), whereas R2* was increased in the inner stripe of the outer medulla in diabetic rats (diabetics 26.1 +/- 2.4 vs. controls 18.8 +/- 1.4 Hz; n=4, P<0.05). This study demonstrates that experimental diabetes in rats induces decreased oxygenation of the renal outer medulla. Furthermore, the proposed T2*-weighted MR micro-imaging technique is suitable for detection of regional changes in kidney oxygenation in experimental animal models.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Rim/metabolismo , Oxigênio/metabolismo , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Rim/anatomia & histologia , Imageamento por Ressonância Magnética , Masculino , Tamanho do Órgão , Ratos , Ratos Wistar , Estreptozocina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA