Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 43(3): 443-455, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36727521

RESUMO

BACKGROUND: Calciprotein particles (CPPs) are associated with the development of vascular calcifications in chronic kidney disease. The role of endothelial cells (ECs) in this process is unknown. Here, we investigated the interaction of CPPs and ECs, thereby focusing on endothelial nitric oxide metabolism and oxidative stress. METHODS: CPPs were generated in calcium- and phosphate-enriched medium. Human umbilical vein endothelial cells were exposed to different concentrations of CPPs (0-100 µg/mL) for 24 or 72 hours. Ex vivo porcine coronary artery rings were used to measure endothelial cell-dependent vascular smooth muscle cell relaxation after CPP exposure. Serum samples from an early chronic kidney disease cohort (n=245) were analyzed for calcification propensity (measure for CPP formation) and nitrate and nitrite levels (NOx). RESULTS: CPP exposure for 24 hours reduced eNOS (endothelial nitric oxide synthase) mRNA expression and decreased nitrite production, indicating reduced nitric oxide bioavailability. Also, 24-hour CPP exposure caused increased mitochondria-derived superoxide generation, together with nitrotyrosine protein residue formation. Long-term (72 hours) exposure of human umbilical vein endothelial cells to CPPs induced eNOS uncoupling and decreased eNOS protein expression, indicating further impairment of the nitric oxide pathway. The ex vivo porcine coronary artery model showed a significant reduction in endothelial-dependent vascular smooth muscle cell relaxation after CPP exposure. A negative association was observed between NOx levels and calcification propensity (r=-0.136; P=0.049) in sera of (early) chronic kidney disease patients. CONCLUSIONS: CPPs cause endothelial cell dysfunction by impairing nitric oxide metabolism and generating oxidative stress. Our findings provide new evidence for direct effects of CPPs on ECs and pathways involved.


Assuntos
Insuficiência Renal Crônica , Doenças Vasculares , Humanos , Animais , Suínos , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Endotélio/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Insuficiência Renal Crônica/metabolismo , Endotélio Vascular/metabolismo
2.
J Vasc Res ; 60(1): 1-11, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36882023

RESUMO

Endovascular revascularization is the preferred treatment for peripheral arterial disease. Restenosis often occurs as a response to procedure-induced arterial damage. Reducing vascular injury during endovascular revascularization may improve its success rate. This study developed and validated an ex vivo flow model using porcine iliac arteries, obtained from a local abattoir. Twenty arteries (of 10 pigs) were equally allocated to two groups: a mock-treated control group and an endovascular intervention group. Arteries of both groups were perfused with porcine blood for 9 min, including 3 min of balloon angioplasty in the intervention group. Vessel injury was assessed by calculating the presence of endothelial cell denudation, vasomotor function, and histopathological analysis. MR imaging displayed balloon positioning and inflation. Endothelial cell staining showed 76% of denudation after ballooning compared to 6% in the control group (p < 0.001). This was confirmed by histopathological analysis, showing a significantly reduced endothelial nuclei count after ballooning compared to the controls (median: 22 vs. 37 nuclei/mm, p = 0.022). In the intervention group, vasoconstriction and endothelium-dependent relaxation were significantly reduced (p < 0.05).We present an ex vivo flow model to test the effects of endovascular therapy on the vessel's wall morphology, endothelial denudation, and endothelial-dependent vasomotor function under physiological conditions. Additionally, it allows the future testing of human arterial tissue.


Assuntos
Angioplastia Coronária com Balão , Angioplastia com Balão , Humanos , Suínos , Animais , Angioplastia Coronária com Balão/métodos , Angioplastia com Balão/efeitos adversos , Imageamento por Ressonância Magnética , Vasoconstrição/fisiologia , Artéria Ilíaca/diagnóstico por imagem , Artéria Ilíaca/cirurgia
3.
Am J Physiol Renal Physiol ; 310(10): F1047-53, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26911850

RESUMO

In healthy rats, the physiological variation of baseline endothelial function of intrarenal arteries correlates with the severity of renal damage in response to a subsequent specific renal injury. However, whether such a variation in endothelial function may also condition or predict the variable response to angiotensin-converting enzyme-inhibiting treatment in these individuals has not been addressed before. To study this, 5/6 nephrectomy was performed to induce renal injury and chronic kidney disease in a group of healthy Wistar rats. At the time of nephrectomy, interlobar arteries were obtained from the extirpated right kidney and studied in vitro for endothelium-dependent relaxation to acetylcholine. Six weeks thereafter, treatment with lisinopril was started (n = 11) and continued for 9 wk. Proteinuria (metabolic cages) and systolic blood pressure (SBP; tail cuff) were evaluated weekly, and these were analyzed in relation to renal endothelial function at baseline. 5/6 Nephrectomy induced an increase in SBP and progressive proteinuria. Treatment with lisinopril reduced SBP and slowed proteinuria, albeit to a variable degree among individuals. The acetylcholine-induced renal artery dilation at baseline negatively correlated with lisinopril-induced reduction of proteinuria (r(2) = 0.648, P = 0.003) and with the decrease in SBP (r(2) = 0.592, P = 0.006). Our data suggest that angiotensin-converting enzyme-inhibitor attenuates the progression of renal damage the most in those individuals with decreased basal renal endothelial-mediated vasodilation.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Endotélio Vascular/efeitos dos fármacos , Lisinopril/uso terapêutico , Insuficiência Renal Crônica/tratamento farmacológico , Vasodilatação , Acetilcolina , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Técnicas In Vitro , Lisinopril/farmacologia , Nefrectomia , Proteinúria/tratamento farmacológico , Distribuição Aleatória , Ratos Wistar , Artéria Renal/efeitos dos fármacos
4.
Am J Physiol Heart Circ Physiol ; 311(6): H1459-H1469, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27769995

RESUMO

Vitamin D deficiency is one of the most common nutritional deficiencies worldwide. Maternal vitamin D deficiency is associated with increased susceptibility to hypertension in offspring, but the reasons for this remain unknown. The aim of this study was to determine if parental vitamin D deficiency leads to altered DNA methylation in offspring that may relate to hypertension. Male and female Sprague-Dawley rats were fed a standard or vitamin D-depleted diet. After 10 wk, nonsibling rats were mated. The conceived pups received standard chow. We observed an increased systolic and diastolic blood pressure in the offspring from depleted parents (F1-depl). Genome-wide methylation analyses in offspring identified hypermethylation of the promoter region of the Pannexin-1 (Panx1) gene in F1-depl rats. Panx1 encodes a hemichannel known to be involved in endothelial-dependent relaxation, and we demonstrated that in F1-depl rats the increase in blood pressure was associated with impaired endothelial relaxation of the large vessels, suggesting an underlying biological mechanism of increased blood pressure in children from parents with vitamin deficiency. Parental vitamin D deficiency is associated with epigenetic changes and increased blood pressure levels in offspring.


Assuntos
Pressão Sanguínea/genética , Conexinas/genética , Metilação de DNA , Hipertensão/genética , Proteínas do Tecido Nervoso/genética , Exposição Paterna , Complicações na Gravidez/genética , Efeitos Tardios da Exposição Pré-Natal/genética , Deficiência de Vitamina D/genética , Animais , Fator Natriurético Atrial/genética , Western Blotting , Endotélio Vascular/fisiopatologia , Epigênese Genética , Feminino , Hipertensão/fisiopatologia , Masculino , Exposição Materna , Hormônio Paratireóideo/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptor Tipo 1 de Angiotensina/genética , Receptores de Calcitriol/genética , Renina/genética , Vasodilatação/genética , Vitamina D/análogos & derivados , Vitamina D/metabolismo , Deficiência de Vitamina D/metabolismo
5.
Am J Physiol Renal Physiol ; 305(9): F1249-64, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23986522

RESUMO

Growth differentiation factor 15 (GDF15) is emerging as valuable biomarker in cardiovascular disease and diabetic kidney disease. Also, GDF15 represents an early response gene induced after tissue injury and studies performed in GDF15 knockout (KO) mice suggest that GDF15 plays a protective role after injury. In the current study, we investigated the role of GDF15 in the development of diabetic kidney damage in type 1 and type 2 models of diabetes. Renal damage was assessed in GDF15 KO mice and wild-type (WT) mice in streptozotocin type 1 and db/db type 2 diabetic models. Genetic deletion of GDF15 augmented tubular and interstitial damage in both models of diabetes, despite similar diabetic states in KO and WT mice. Increased tubular damage in KO animals was associated with increased glucosuria and polyuria in both type 1 and type 2 models of diabetes. In both models of diabetes, KO mice showed increased interstitial damage as indicated by increased α-smooth muscle actin staining and collagen type 1 expression. In contrast, glomerular damage was similarly elevated in diabetic KO and WT mice. In type 1 diabetes, GDF15 KO mice demonstrated increased expression of inflammatory markers. In type 2 diabetes, elevated levels of plasma creatinine indicated impaired kidney function in KO mice. GDF15 protects the renal interstitium and tubular compartment in experimental type 1 and 2 diabetes without affecting glomerular damage.


Assuntos
Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/etiologia , Fator 15 de Diferenciação de Crescimento/metabolismo , Animais , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Deleção de Genes , Fator 15 de Diferenciação de Crescimento/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Ratos Wistar , Regulação para Cima
6.
Am J Physiol Heart Circ Physiol ; 304(5): H709-18, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23262134

RESUMO

Growth differentiation factor 15 (GDF15) is an independent predictor of cardiovascular disease, and increased GDF15 levels have been associated with endothelial dysfunction in selected patients. We therefore investigated whether GDF15 modulates endothelial function in aortas of wild-type (WT) and GDF15 knockout (KO) mice. Vascular contractions to phenylephrine and relaxation to ACh were assessed in aortas obtained from healthy WT and GDF15 KO mice. The effects of GDF15 pretreatment and the involvement of ROS or caveolae were determined. Phenylephrine-induced contractions and ACh-mediated relaxations were similar in WT and GDF15 KO mice. Pretreatment with GDF15 inhibited contraction and relaxation in both groups. Inhibition of contraction by GDF15 was absent in denuded vessels or after blockade of nitric oxide (NO) synthase. Relaxation in WT mice was mediated mainly through NO and an unidentified endothelium-derived hyperpolarizin factor (EDHF), whereas GDF15 KO mice mainly used prostaglandins and EDHF. Pretreatment with GDF15 impaired relaxation in WT mice by decreasing NO; in GDF15 KO mice, this was mediated by decreased action of prostaglandins. Disruption of caveolae resulted in a similar inhibition of vascular responses as GDF15. ROS inhibition did not affect vascular function. In cultured endothelial cells, GDF15 pretreatment caused a dissociation between caveolin-1 and endothelial NO synthase. In conclusion, GDF15 impairs aortic contractile and relaxing function through an endothelium-dependent mechanism involving altered caveolar endothelial NO synthase signaling.


Assuntos
Aorta Torácica/fisiologia , Fator 15 de Diferenciação de Crescimento/genética , Fator 15 de Diferenciação de Crescimento/metabolismo , Transdução de Sinais/fisiologia , Vasodilatação/fisiologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Cavéolas/fisiologia , Caveolina 1/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Fator 15 de Diferenciação de Crescimento/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores para Leptina/genética , Transdução de Sinais/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
7.
J Pharmacol Exp Ther ; 345(3): 393-403, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23528611

RESUMO

Transactivation of epidermal growth factor receptor (EGFR) signaling by G protein-coupled receptors has been implicated in several cardiovascular (CV) conditions, including hypertension, heart failure, and cardiac and vascular hypertrophy. However, the therapeutic potential of EGFR inhibition in these conditions is currently unknown. The main objective of the present study was to investigate cardiac, vascular, and renal effects of EGFR inhibition by 4-[4-[[(1R)-1-phenylethyl]amino]-7H-pyrrolo[2,3-d]pyrimidin-6-yl]phenol (PKI-166) in the hypertensive chronic kidney disease model. Rats underwent 5/6 nephrectomy (5/6Nx) and were treated with PKI-166, lisinopril or vehicle from week 6 after disease induction until week 12. Sham animals received either PKI-166 or vehicle. Treatment with PKI-166 did not affect the development of the characteristic renal features in 5/6Nx, including proteinuria, diminished creatinine clearance, and increased glomerulosclerosis, whereas these were attenuated by lisinopril. Despite absence of effects on progressive renal damage, PKI-166 attenuated the progression of hypertension and maintained cardiac function (left ventricle end-diastolic pressure) to a similar extent as lisinopril. Also, PKI-166 attenuated the increase in phosphorylated EGFR in the heart as induced by 5/6Nx. Moreover, PKI-166 and lisinopril restored the impaired contraction of isolated thoracic aortic rings to phenylephrine and angiotensin II and impaired myogenic constriction of small mesenteric arteries in 5/6Nx rats. Blockade of the EGFR displays a CV benefit independent of limiting the progression of renal injury. Our findings extend the evidence on EGFR signaling as a target in CV disorders.


Assuntos
Cardiotônicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Hipertensão Renal/tratamento farmacológico , Rim/efeitos dos fármacos , Nefrectomia , Pirimidinas/farmacologia , Pirróis/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Hipertensão Renal/fisiopatologia , Imuno-Histoquímica , Lisinopril/farmacologia , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Tono Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Proteinúria/metabolismo , Ratos , Ratos Wistar , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptores Adrenérgicos alfa 1/efeitos dos fármacos
8.
EJNMMI Res ; 13(1): 106, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38079017

RESUMO

BACKGROUND: The apolipoprotein E-deficient (apoE-/-) mouse is a well-established model for studying atherosclerosis. However, its small size limits its use in longitudinal positron emission tomography (PET) imaging studies. Recently, the apoE-/- rat has emerged as an alternative. With this study, we investigate the feasibility of using apoE-/- rats as an in vivo model for longitudinal atherosclerotic PET/CT imaging. RESULTS: ApoE-/- rats showed significantly higher [18F]FDG uptake than controls in the aortic arch (+ 18.5%, p < 0.001) and abdominal aorta (+ 31.0%, p < 0.001) at weeks 12, 26, and 51. ApoE-/- rats exhibited hypercholesterolemia, as evidenced by plasma cholesterol levels that were up to tenfold higher, and total hepatic cholesterol levels that were up to threefold higher than the control rats at the end of the study. Fast protein liquid chromatography cholesterol profiling indicated very high levels of pro-atherogenic apoB-containing very low-density lipoprotein and low-density lipoprotein fractions in the apoE-/- rats. Atherosclerotic lesions cover 19.9% of the surface of the aortic arch (p = 0.0013), and there was a significantly higher subendothelial accumulation of ED1-positive macrophages in the abdominal aorta of the apoE-/- rats compared to control rats (Ctrl) (p = 0.01). No differences in neutral sterols were observed but higher levels of bile acids were found in the apoE-/- rats. CONCLUSION: These data demonstrate early signs of hypercholesterolemia, high levels of bile acids, the development of atherosclerotic lesions, and macrophage accumulation in apoE-/- rats. Therefore, this model shows promise for atherosclerosis imaging studies.

9.
Am J Physiol Renal Physiol ; 303(8): F1187-95, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22791345

RESUMO

Previously, it was shown that individuals with good baseline (a priori) endothelial function in isolated (in vitro) renal arteries developed less renal damage after 5/6 nephrectomy (5/6Nx; Gschwend S, Buikema H, Navis G, Henning RH, de Zeeuw D, van Dokkum RP. J Am Soc Nephrol 13: 2909-2915, 2002). In this study, we investigated whether preexisting glomerular vascular integrity predicts subsequent renal damage after 5/6Nx, using in vivo intravital microscopy and in vitro myogenic constriction of small renal arteries. Moreover, we aimed to elucidate the role of renal ANG II type 1 receptor (AT1R) expression in this model. Anesthetized rats underwent intravital microscopy to visualize constriction to ANG II of glomerular afferent and efferent arterioles, with continuous measurement of blood pressure, heart rate, and renal blood flow. Thereafter, 5/6Nx was performed, interlobar arteries were isolated from the extirpated kidney, and myogenic constriction was assessed in a perfused vessel setup. Blood pressure and proteinuria were assessed weekly for 12 wk, and focal glomerulosclerosis (FGS) was determined at the end of study. Relative expression AT1R in the kidney cortex obtained at 5/6Nx was determined by PCR. Infusion of ANG II induced significant constriction of both afferent and efferent glomerular arterioles, which strongly positively correlated with proteinuria and FGS at 12 wk after 5/6Nx. Furthermore, in vitro measured myogenic constriction of small renal arteries negatively correlated with proteinuria 12 wk after 5/6Nx. Moreover, in vivo vascular reactivity negatively correlated with in vitro reactivity. Additionally, relative expression of AT1R positively correlated with responses of glomerular arterioles and with markers of renal damage. Both in vivo afferent and efferent responses to ANG II and in vitro myogenic constriction of small renal arteries in the healthy rat predict the severity of renal damage induced by 5/6Nx. This vascular responsiveness is highly dependent on AT1R expression. Intraorgan vascular integrity may provide a useful tool to guide the prevention and treatment of renal end-organ damage.


Assuntos
Nefropatias/fisiopatologia , Glomérulos Renais/irrigação sanguínea , Rim/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Artéria Renal/fisiopatologia , Acetilcolina/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/fisiopatologia , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Fenilefrina/farmacologia , Proteinúria/fisiopatologia , Ratos , Ratos Wistar , Artéria Renal/efeitos dos fármacos , Circulação Renal/efeitos dos fármacos , Circulação Renal/fisiologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasoconstritores/farmacologia
10.
Biomater Adv ; 134: 112693, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35581088

RESUMO

One of the most common magnesium (Mg) applications in the biomedical field is in cardiovascular stents. Although Mg is an essential element for homeostasis, Mg is highly reactive, and locally high Mg concentrations can have toxic effects on the surrounding tissue. One strategy to circumvent the Mg toxicity is using coatings or surface modifications that prevent the leaching of excessive Mg ions. In the current study, commercially pure magnesium (c.p Mg) was modified through plasma electrolytic oxidation (PEO) to produce a protective coating primarily composed of Mg oxide (MgO) and Mg hydroxide (Mg(OH)2), which limits leaching of free Mg ions from the base material. As we intend to use this material to produce vascular stents, a biological evaluation of its performance is warranted. Primary human umbilical vein endothelial cells (HUVECs) and smooth muscle cells (SMCs) were the study object. The leaching of free Mg ions from the oxidized materials was investigated, as was its effect on local pH changes. We also investigated the influence of corrosion products, the effects of elevated free Mg concentrations and pH on the cellular behavior on the integrity of monolayers of HUVECs was studied in a static and dynamic model. Results showed that the harmful effect of Mg on cells due to changes in pH and a high concentration of Mg ions could decrease with the influence of flow diffusing corrosion products such as MgO, Mg(OH)2, and H2 among the system. Independently, Mg concentration and pH affected the cell activity of SMCs and HUVECs. Finally, to investigate the influence of leachables on vasomotor function, we exposed porcine aortic rings to PEO-modified Mg stents and assessed endothelial-dependent relaxation. Pure Mg reduced vasorelaxation from 100% in control samples to 30%. Oppositely, PEO-modified Mg did not affect the vasomotor function. Overall, we conclude from this study that the use of PEO coatings reduces the degradation rate of the material reducing the Mg release resulting in better cell viability and vessel function compared to the bare material.


Assuntos
Ligas , Magnésio , Ligas/farmacologia , Animais , Materiais Revestidos Biocompatíveis/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Magnésio/farmacologia , Hidróxido de Magnésio , Óxido de Magnésio , Suínos
11.
Arterioscler Thromb Vasc Biol ; 30(7): 1407-14, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20448207

RESUMO

OBJECTIVE: Endothelial cell senescence is an important contributor to vascular aging and is increased under diabetic conditions. Here we investigated whether the antidiabetic hormone glucagon-like peptide 1 (GLP-1) could prevent oxidative stress-induced cellular senescence in endothelial cells. METHODS AND RESULTS: In Zucker diabetic fatty rats, a significant 2-fold higher level of vascular senescence was observed compared with control lean rats. Dipeptidyl-peptidase 4 (DPP-4) inhibition significantly increased GLP-1 levels in these animals and reduced senescence almost to lean animal levels. In vitro studies with human umbilical vein endothelial cells showed that GLP-1 had a direct protective effect on oxidative stress (H(2)O(2))-induced senescence and was able to attenuate oxidative stress-induced DNA damage and cellular senescence. The GLP-1 analogue exendin-4 provided similar results, whereas exendin fragment 9-39, a GLP-1 receptor antagonist, abolished this effect. Intracellular signaling by the phosphoinositide 3-kinase (PI3K)/Akt survival pathway did not appear to be involved. Further analysis revealed that GLP-1 activates the cAMP response element-binding (CREB) transcription factor in a cAMP/protein kinase A (PKA)-dependent manner, and inhibition of the cAMP/PKA pathway abolished the GLP-1 protective effect. Expression analysis revealed that GLP-1 can induce the oxidative defense genes HO-1 and NQO1. CONCLUSIONS: Dipeptidyl-peptidase 4 inhibition protects against vascular senescence in a diabetic rat model. In vitro studies with human umbilical vein endothelial cells showed that reactive oxygen species-induced senescence was attenuated by GLP-1 in a receptor-dependent manner involving downstream PKA signaling and induction of antioxidant genes.


Assuntos
Senescência Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus/enzimologia , Células Endoteliais/enzimologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Adamantano/análogos & derivados , Adamantano/farmacologia , Animais , Células Cultivadas , Senescência Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dano ao DNA , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/patologia , Inibidores da Dipeptidil Peptidase IV/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Ativação Enzimática , Exenatida , Receptor do Peptídeo Semelhante ao Glucagon 1 , Heme Oxigenase-1/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Hipoglicemiantes/farmacologia , Masculino , NAD(P)H Desidrogenase (Quinona)/metabolismo , Nitrilas/farmacologia , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirrolidinas/farmacologia , Ratos , Ratos Zucker , Receptores de Glucagon/metabolismo , Transdução de Sinais , Peçonhas/farmacologia , Vildagliptina
12.
Am J Physiol Renal Physiol ; 298(3): F625-33, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20007352

RESUMO

It is unknown whether generalized vascular dysfunction precedes the development of kidney disease. Therefore, we studied myogenic constriction and endothelium-mediated dilatory responses in two inbred Fawn-Hooded (FH) rat strains, one of which spontaneously develops hypertension, proteinuria, and glomerulosclerosis (FHH), whereas the other (FHL) does not. Small renal, mesenteric resistance arteries and thoracic aorta isolated from FH rats before (7 wk old) and after the development of mild proteinuria (12 wks old) were mounted in perfused and isometric set-ups, respectively. Myogenic response, endothelium-dependent relaxation, and the contribution of endothelium-mediated dilatory compounds were studied using their respective inhibitors. Myogenic reactivity was assessed constructing pressure-diameter curves in the presence and absence of calcium. At the age of 7 wk, renal arteries isolated from kidneys of FHH rats developed significantly lower myogenic tone compared with FHL, most likely because of excessive cyclo-oxygenase 1-mediated production of constrictive prostaglandins. Consequently, young FHH demonstrated reduced maximal myogenic tone (22 +/- 4.8 vs. 10.8 +/- 2.0%, P = 0.03) and the peak myogenic index (-6.9 +/- 4.8 vs. 0.6 +/- 0.8%/mmHg, P = 0.07 for FHL vs. FHH, respectively). Active myogenic curves obtained in mesenteric arteries isolated from 7-wk-old rats did not differ between either strain, demonstrating a similar level of systemic myogenic tone in FHL and FHH rats. Therefore, before any renal end-organ damage is present, myogenic response seems selectively impaired in renal vasculature of FHH rats. Aortic reactivity did not differ between FHL and FHH at the time points studied. The present study shows that vascular dysfunction in both small renal and systemic arteries precedes renal end-organ damage in a spontaneous model of hypertension-associated renal damage. These early vascular changes might be potentially involved in the increased susceptibility of FHH rats to renal injury.


Assuntos
Hipertensão/fisiopatologia , Nefropatias/etiologia , Rim/irrigação sanguínea , Artéria Renal/fisiopatologia , Circulação Renal , Vasoconstrição , Vasodilatação , Fatores Etários , Envelhecimento , Animais , Aorta Torácica/fisiopatologia , Fatores Biológicos/metabolismo , Pressão Sanguínea , Ciclo-Oxigenase 1/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Hipertensão/complicações , Hipertensão/metabolismo , Hipertensão/patologia , Rim/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Nefropatias/fisiopatologia , Masculino , Proteínas de Membrana/metabolismo , Óxido Nítrico/metabolismo , Prostaglandinas/metabolismo , Proteinúria/etiologia , Proteinúria/patologia , Proteinúria/fisiopatologia , Ratos , Ratos Endogâmicos , Artéria Renal/efeitos dos fármacos , Artéria Renal/metabolismo , Circulação Renal/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
13.
Curr Opin Pharmacol ; 9(2): 132-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19157981

RESUMO

Nephrosis refers to a condition resulting from proteinuric kidney disease, leading to irreversible renal parenchymal damage and end-stage renal disease when left untreated. Furthermore, nephrosis appears to be a communicable disease carrying risks and complications to other organs such as the heart. Key pathophysiolgical processes involved in initiating and progressing renal damage in nephrosis and its complications may include altered glomerular hemodynamics after initial renal damage and loss of nephrons, nephrotoxicity of increased renal protein traffic enforcing intrinsic 'common pathway' mechanisms of renal scarring, and generalized endothelial dysfunction proceeding CV disease. The reader is first provided a basic overview on key mechanisms, targets and therapies in nephrosis while referred to some excellent updates hereon for more detailed information. The broader purpose of this short review, however, is to highlight caveolae/caveolins and caveolar function as central modulators in all the above key processes of nephrosis. Caveolae - little caves in the plasma membrane that are particularly abundant in endothelial cells, amongst others - are now known to be involved not only in endothelial transcytosis (e.g. of albumin) but also in cholesterol homeostasis (LDL-transport) and, importantly, in signal transduction such as insulin signalling and nitric oxide signalling in endothelial function and regulation of vasomotor tone, as well as signalling by growth factor receptors - such as TGF-beta - which may participate in renal scarring. It is suggested that caveolae may represent crucial sites where possible new druggable targets in nephrosis may be found.


Assuntos
Cavéolas/efeitos dos fármacos , Cavéolas/metabolismo , Cavéolas/fisiologia , Caveolinas/metabolismo , Caveolinas/fisiologia , Nefrose/tratamento farmacológico , Albuminúria/fisiopatologia , Animais , Antioxidantes/uso terapêutico , Sistemas de Liberação de Medicamentos , Endotélio Vascular/fisiopatologia , Fibrose/metabolismo , Taxa de Filtração Glomerular/fisiologia , Humanos , Rim/irrigação sanguínea , Rim/metabolismo , Rim/fisiopatologia , Nefrose/fisiopatologia
14.
Mamm Genome ; 20(3): 170-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19255705

RESUMO

In humans, the insertion/deletion polymorphism in the angiotensin converting enzyme (ACE) gene accounts for half of the variance in plasma ACE activity. The deletion allele is associated with high plasma ACE activity, cardiovascular disease, and renal disease. In rat, a similar association is found between the B and L alleles of a microsatellite marker in the ACE gene. We identified the B/L variation in the Wistar outbred rat and bred two lines homozygous for the two alleles (WU-B and WU-L). ACE activity was measured in serum, heart, kidney, and aorta homogenates. Immunohistochemistry and ACE mRNA expression were performed in heart, kidney, and aortic tissue. Aortic rings were collected and stimulated with AngI, AngII, and AngI with Lisinopril to measure ACE functional activity by vasoconstrictor response. Serum, heart, and kidney ACE activity and kidney mRNA expression were two-fold higher in WU-B. Kidney staining showed a clear difference in tubular ACE expression, with more staining in WU-B. While in aorta ACE activity and mRNA expression was twofold higher in WU-L, functional conversion of AngI was higher in WU-B, indicating either a functional difference in AngI to AngII conversion between the two alleles due to different splicing or the presence of other factors involved in the conversion that are differentially expressed as the result of differences in the ACE alleles. The newly developed WU-B and WU-L lines show tissue-specific differences in ACE expression and activity. This provides an experimental tool to study the pathophysiologic consequences of differences in ACE alleles in renal and cardiovascular disease.


Assuntos
Doenças Cardiovasculares/enzimologia , Expressão Gênica , Nefropatias/enzimologia , Peptidil Dipeptidase A/genética , Alelos , Angiotensinas/metabolismo , Animais , Cruzamento , Doenças Cardiovasculares/genética , Humanos , Rim/enzimologia , Nefropatias/genética , Miocárdio/enzimologia , Especificidade de Órgãos , Peptidil Dipeptidase A/sangue , Peptidil Dipeptidase A/metabolismo , Ratos , Ratos Wistar
15.
Eur J Heart Fail ; 11(3): 246-55, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19147448

RESUMO

AIMS: We previously showed that enhanced myogenic constriction (MC) of peripheral resistance arteries involves active AT(1) receptors in chronic heart failure (CHF). Recent data suggest both transactivation of EGF receptors and caveolae-like microdomains to be implicated in the activity of AT(1) receptors. Thus, we assessed their roles in increased MC in mesenteric arteries of CHF rats. METHODS AND RESULTS: Male Wistar rats underwent myocardial infarction to induce CHF and were sacrificed after 12 weeks. The number of caveolae in smooth muscle cells (SMC) of mesenteric arteries of CHF rats was decreased by 43.6 +/- 4.0%, this was accompanied by increased MC, which was fully normalized to the level of sham by antagonists of the AT(1)-receptor (losartan) or EGF-receptor (AG1478). Acute disruption of caveolae in sham rats affected caveolae numbers and MC to a similar extent as CHF, however MC was only reversed by the antagonist of the EGF-receptor, but not by the AT(1)-receptor antagonist. Further, in sham rats, MC was increased by a sub-threshold concentration of angiotensin II and reversed by both AT(1)- as well as EGF-receptor inhibition. In contrast, increased MC by a sub-threshold concentration of EGF was only reversed by EGF receptor inhibition. CONCLUSION: These findings provide the first evidence that decreased SMC caveolae numbers are involved in enhanced MC in small mesenteric arteries, by affecting AT(1)- and EGF-receptor function. This suggests a novel mechanism involved in increased peripheral resistance in CHF.


Assuntos
Cavéolas/ultraestrutura , Receptores ErbB/metabolismo , Insuficiência Cardíaca/fisiopatologia , Artérias Mesentéricas/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Receptor Tipo 1 de Angiotensina/metabolismo , Vasoconstrição/fisiologia , Animais , Contagem de Células , Modelos Animais de Doenças , Imunofluorescência , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Imuno-Histoquímica , Masculino , Artérias Mesentéricas/metabolismo , Artérias Mesentéricas/ultraestrutura , Microscopia Eletrônica , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/ultraestrutura , Contração Miocárdica/fisiologia , Ratos , Ratos Wistar
16.
Am J Nephrol ; 30(3): 209-17, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19420905

RESUMO

BACKGROUND: Proteinuria-associated endothelial dysfunction (ED) is assumed to play a main role in the cardiovascular morbidity in proteinuric patients. However, the connection between proteinuria and systemic endothelial function is not clear yet. Therefore, we studied aortic endothelial function in Munich Wistar Fromter (MWF) and fawn-hooded hypertensive (FHH) inbred rat strains with genetic proteinuria to determine the specific impact of proteinuria on the development of ED. METHODS: Proteinuria, cardiac function, systemic blood pressure, plasma lipid profiles, aortic endothelial function, plasma levels of cyclo-oxygenase products and dimethylarginines were investigated in 26-week-old inbred rat strains with (MWF and FHH) and without [Lewis (LEW) rats] proteinuric renal disease. RESULTS: The endothelium-dependent relaxation was significantly reduced in MWF (p < 0.05 vs. LEW or FHH). The plasma thromboxane B(2), prostaglandin F(2alpha) and prostaglandin E(2)levels were higher in MWF (p < 0.05 vs. LEW or FHH), whereas the 6-keto-prostaglandin F(1alpha) level was comparable in all groups. The arginine/asymmetric dimethylarginine ratio was highest in MWF. CONCLUSIONS: This study differentiates common risk factors for ED in renal disease. Despite clear-cut proteinuria, FHH rats were devoid of changes in aortic endothelial function, indicating that some other deleterious factors must accompany proteinuria in order for ED to ensue. Further exploration of this model may serve to dissect mechanistical pathways and guide the development of protective strategies in the vascular damage of renal disease.


Assuntos
Endotélio Vascular/fisiopatologia , Proteinúria/complicações , Proteinúria/fisiopatologia , Animais , Masculino , Ratos , Ratos Endogâmicos Lew , Ratos Endogâmicos
17.
Anesthesiology ; 111(3): 600-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19672168

RESUMO

BACKGROUND: Hemorrhagic shock is associated with changes in vascular responsiveness that may lead to organ dysfunction and, ultimately, multiple organ dysfunction syndrome. Volatile anesthetics interfere with vasoresponsiveness, which may contribute to organ hypoperfusion. In this study, the authors examined the influence of adjunct nitrous oxide on the vascular responsiveness after short-term hemorrhagic shock under isoflurane anesthesia. METHODS: Spontaneously breathing mice (n = 31, 27.6 +/- 0.31 g) were anesthetized with isoflurane (1.4%) or with isoflurane (1.4%) and adjunct nitrous oxide (66%). Both groups were divided into Sham, Shock, and Resuscitated groups. Vascular reactivity to phenylephrine and acetylcholine and expression of cyclooxygenases were studied in the aorta. RESULTS: In the isoflurane-anesthetized groups, the contractile response to phenylephrine was increased in the Shock as compared with the Sham and Resuscitated groups (Emax = 3.2 +/- 0.4, 1.2 +/- 0.4, and 2.5 +/- 0.5 mN, respectively). Adjunct nitrous oxide increased phenylephrine contraction to a similar level in all three groups. In the Sham isoflurane group, acetylcholine caused a biphasic response: An initial relaxation followed by a contractile response sensitive to cyclooxygenases inhibition by indomethacine. The contractile response was abrogated in the isoflurane-anesthetized groups that underwent shock. In all groups, adjunct nitrous oxide preserved the contractile phase. Shock induced a down-regulation of cyclooxygenases-1, which was normalized by adjunct nitrous oxide. CONCLUSION: Adjunct nitrous oxide attenuates shock-induced changes in vascular reactivity and cyclooxygenases expression of mice under isoflurane anesthesia. This implies that vascular reactive properties during anesthesia in hemorrhagic shock conditions may be influenced by the choice of anesthetics.


Assuntos
Anestesia por Inalação , Anestésicos Inalatórios/uso terapêutico , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiopatologia , Isoflurano , Óxido Nitroso/uso terapêutico , Choque Hemorrágico/tratamento farmacológico , Choque Hemorrágico/fisiopatologia , Acetilcolina/farmacologia , Animais , Gasometria , Pressão Sanguínea/fisiologia , Western Blotting , Ciclo-Oxigenase 1/biossíntese , Ciclo-Oxigenase 2/biossíntese , Camundongos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Doadores de Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasodilatadores/farmacologia
18.
Nephrol Dial Transplant ; 24(2): 413-20, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18728153

RESUMO

BACKGROUND: Susceptibility to renal injury varies among individuals. Previously, we found that individual endothelial function of healthy renal arteries in vitro predicted severity of renal damage after 5/6 nephrectomy. Here we hypothesized that individual differences in endothelial function in vitro and renal perfusion in vivo predict the severity of renal damage in a model of adriamycin-induced nephropathy. METHODS: In three separate studies, the following baseline parameters were measured in healthy male Wistar rats: (1) acetylcholine (ACh)-induced relaxation in small renal arteries in vitro (n = 16) and the contribution of prostaglandins, nitric oxide (NO) and endothelium-dependent hyperpolarizing factor (EDHF) to the relaxation; (2) glomerular filtration rate (GFR) and effective renal plasma flow (ERPF) in spontaneously voiding rats in vivo (n = 16) and (3) the acute effect of the NO-synthase inhibitor N(G)-nitro-l-arginine methyl ester (L-NAME, n = 12) on renal blood flow (RBF) as compared to vehicle infusion (n = 9). Following these measurements, adriamycin (1.75 mg/kg i.v.) was injected and subsequent renal damage after 6 weeks was related to the baseline parameters. RESULTS: Total ACh-induced (r = 0.51, P < 0.05) and EDHF-mediated relaxation (r = 0.68, P < 0.05), as well as ERPF (r = 0.66, P < 0.01), positively correlated with the severity of proteinuria 6 weeks after injection. In contrast, pronounced baseline NO-mediated dilation was associated with lower proteinuria (r = 0.71, P < 0.01). Nevertheless, an acute L-NAME infusion, strongly reducing RBF by 22 +/- 8%, during adriamycin administration provided protection against the development of proteinuria. CONCLUSIONS: Individual animals with pronounced baseline endothelial dilatory ability measured in vitro and high ERPF in vivo are vulnerable to renal damage after the adriamycin injection. Acute inhibition of NO during adriamycin administration, resulting in a decrease of RBF, protects against renal injury, probably by limiting the delivery of the drug to the kidney. Therefore, interindividual variability in renal haemodynamics might be crucially involved in susceptibility to nephrotoxic renal damage.


Assuntos
Doxorrubicina/toxicidade , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Circulação Renal , Acetilcolina/farmacologia , Animais , Suscetibilidade a Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Rim/lesões , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/fisiologia , Nitroprussiato/farmacologia , Ratos , Ratos Wistar , Circulação Renal/efeitos dos fármacos , Fluxo Plasmático Renal Efetivo/efeitos dos fármacos , ômega-N-Metilarginina/farmacologia
19.
Nephrol Dial Transplant ; 23(6): 1854-60, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18218687

RESUMO

BACKGROUND: Nephrosis-induced endothelial dysfunction is assumed to play a main role in cardiovascular morbidity. Adriamycin-induced proteinuria is a well-established rat model for nephrotic syndrome. However, induction of nephrosis by intravenous adriamycin administration might exert direct adriamycin cardiovasculotoxicity that could obscure or modify nephrosis-induced vascular dysfunction. The present study, therefore, investigated in vitro vascular function in the isolated thoracic aorta and isolated perfused hearts of rats with adriamycin nephrosis, as compared to non-nephrotic adriamycin exposed rats. METHODS: Adult rats received a single slow intravenous injection of either adriamycin (1.5 mg/kg, adriamycin nephrotic rats) or saline (healthy controls). In a third group of rats, the cardiovascular system, but not the kidneys, were exposed to adriamycin by transient clipping of renal arteries during adriamycin injection (adriamycin control rats). RESULTS: Exposure of the kidneys to adriamycin induced severe proteinuria with corresponding systemic nephrosis, as apparent from hypercholesterolaemia. Adriamycin exposure of the vascular bed led to marked blunting of the aortic response to the endothelium-dependent vasodilator, acetylcholine (ACh), both in non-nephrotic and nephrotic rats. The nephrotic state reduced the bradykinin-induced increase in coronary flow and enhanced the aortic constrictor response to angiotensin II associated with reduced basal aortic NO-activity, as shown by the comparison between adriamycin nephrotic rats and healthy and adriamycin controls. CONCLUSIONS: Vascular adriamycin exposure and nephrosis affect vascular function in a distinct and qualitatively different fashion in adriamycin-induced nephrotic syndrome. The differential effects of nephrosis and vascular adriamycin exposure have to be accounted for in the interpretation of vascular studies in adriamycin nephrosis.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Doxorrubicina/efeitos adversos , Endotélio Vascular/efeitos dos fármacos , Síndrome Nefrótica/induzido quimicamente , Animais , Determinação da Pressão Arterial , Sistema Cardiovascular/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Doxorrubicina/farmacologia , Masculino , Síndrome Nefrótica/patologia , Proteinúria/induzido quimicamente , Proteinúria/fisiopatologia , Ratos , Ratos Wistar , Valores de Referência , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Sanguíneo Regional/fisiologia , Sensibilidade e Especificidade
20.
Eur J Pharmacol ; 585(2-3): 256-60, 2008 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-18423600

RESUMO

Discovery in the early 1990s of caveolin-1, the structural protein responsible for maintaining the ohm shape of caveolae, greatly enhanced investigations to elucidate the role of these little caves in the plasma membrane. Perhaps one of the most important realizations concerning caveolae and caveolin is that these elements play an important functional role in the modulation of cell signal transduction pathways, including those involved in endothelial nitric oxide synthase (eNOS) function. Their role was confirmed by studies with caveolin-1 knockout mice which lack caveolae and display abnormal endothelial function responses. One limitation of these knockout models, however, is that absence of the caveolin protein not only results in the lack of caveolae as a structure but also in the lack of interaction/modulation of enzymes/molecules (e.g. eNOS) to which caveolin binds (whether in- or outside caveolae). In contrast to caveolin knockout models, recent experimental findings suggest that in certain cardiovascular diseases caveolin may dissociate from caveolae to the cytosol, hence decreasing the number of caveolae without a change in the total amount of caveolin. Therefore, as the importance of defining the role of caveolins both in caveolae and in cellular regions is being highlighted, it seems also important at the same time to further define the role of caveolae per se being present in the plasma membrane as a structural entity. The objective of this review is to make an explorative tour on the role of caveolae in vascular endothelial function based on existing literature together with some preliminary experimental findings. Evidence and arguments are put forward that alterations in endothelial caveolae do occur in cardiovascular disease and may contribute to the observed endothelial dysfunction in these conditions.


Assuntos
Doenças Cardiovasculares/fisiopatologia , Cavéolas/fisiologia , Endotélio Vascular/fisiopatologia , Animais , Fatores Biológicos/metabolismo , Cálcio/metabolismo , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/patologia , Cavéolas/efeitos dos fármacos , Caveolina 1/genética , Caveolina 1/metabolismo , Colesterol/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Humanos , Camundongos , Camundongos Knockout , Óxido Nítrico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA