Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Toxicol Pathol ; 51(4): 160-175, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37632371

RESUMO

Assessment of hypertensive tubulopathy for more than fifty animal models of hypertension in experimental pathology employs criteria that do not correspond to lesional descriptors for tubular lesions in clinical pathology. We provide a critical appraisal of experimental hypertension with the same approach used to estimate hypertensive renal tubulopathy in humans. Four models with different pathogenesis of hypertension were analyzed-chronic angiotensin (Ang) II-infused and renin-overexpressing (TTRhRen) mice, spontaneously hypertensive (SHR), and Goldblatt two-kidney one-clip (2K1C) rats. Mouse models, SHR, and the nonclipped kidney in 2K1C rats had no regular signs of hypertensive tubulopathy. Histopathology in animals was mild and limited to variations in the volume density of tubular lumen and epithelium, interstitial space, and interstitial collagen. Affected kidneys in animals demonstrated lesion values that are significantly different compared with healthy controls but correspond to mild damage if compared with hypertensive humans. The most substantial human-like hypertensive tubulopathy was detected in the clipped kidney of 2K1C rats. For the first time, our study demonstrated the regular presence of chronic progressive nephropathy (CPN) in relatively young mice and rats with induced hypertension. Because CPN may confound the assessment of rodent models of hypertension, proliferative markers should be used to verify nonhypertensive tubulopathy.


Assuntos
Hipertensão , Patologia Clínica , Humanos , Ratos , Camundongos , Animais , Ratos Endogâmicos SHR , Rim , Modelos Animais de Doenças
2.
PLoS One ; 17(2): e0264136, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35176122

RESUMO

Current research on hypertension utilizes more than fifty animal models that rely mainly on stable increases in systolic blood pressure. In experimental hypertension, grading or scoring of glomerulopathy in the majority of studies is based on a wide range of opinion-based histological changes that do not necessarily comply with lesional descriptors for glomerular injury that are well-established in clinical pathology. Here, we provide a critical appraisal of experimental hypertensive glomerulopathy with the same approach used to assess hypertensive glomerulopathy in humans. Four hypertensive models with varying pathogenesis were analyzed-chronic angiotensin II infused mice, mice expressing active human renin in the liver (TTRhRen), spontaneously hypertensive rats (SHR), and Goldblatt two-kidney one-clip rats (2K1C). Analysis of glomerulopathy utilized the same criteria applied in humans-hyalinosis, focal segmental glomerulosclerosis (FSGS), ischemic, hypertrophic and solidified glomeruli, or global glomerulosclerosis (GGS). Data from animal models were compared to human reference values. Kidneys in TTRhRen mice, SHR and the nonclipped kidneys in 2K1C rats had no sign of hyalinosis, FSGS or GGS. Glomerulopathy in these groups was limited to variations in mesangial and capillary compartment volumes, with mild increases in collagen deposition. Histopathology in angiotensin II infused mice corresponded to mesangioproliferative glomerulonephritis, but not hypertensive glomerulosclerosis. The number of nephrons was significantly reduced in TTRhRen mice and SHR, but did not correlate with severity of glomerulopathy. The most substantial human-like glomerulosclerotic lesions, including FSGS, ischemic obsolescent glomeruli and GGS, were found in the clipped kidneys of 2K1C rats. The comparison of affected kidneys to healthy control in animals produces lesion values that are numerically impressive but correspond to mild damage if compared to humans. Animal studies should be standardized by employing the criteria and classifications established in human pathology to make experimental and human data fully comparable for comprehensive analysis and model improvements.


Assuntos
Angiotensina II/toxicidade , Modelos Animais de Doenças , Glomerulosclerose Segmentar e Focal/patologia , Hipertensão Renal/patologia , Hipertensão/complicações , Nefrite/patologia , Nefroesclerose/patologia , Animais , Glomerulosclerose Segmentar e Focal/etiologia , Glomerulosclerose Segmentar e Focal/metabolismo , Humanos , Hipertensão/induzido quimicamente , Hipertensão Renal/etiologia , Hipertensão Renal/metabolismo , Masculino , Nefrite/etiologia , Nefrite/metabolismo , Nefroesclerose/etiologia , Nefroesclerose/metabolismo , Ratos , Ratos Endogâmicos SHR , Vasoconstritores/toxicidade
3.
Lab Invest ; 100(3): 414-425, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31527829

RESUMO

Prostaglandin E2 receptor EP1 (PGE2/EP1) promotes diabetic renal injury, and EP1 receptor deletion improves hyperfiltration, albuminuria, and fibrosis. The role of EP1 receptors in hypertensive kidney disease (HKD) remains controversial. We examined the contribution of EP1 receptors to HKD. EP1 null (EP1-/-) mice were bred with hypertensive TTRhRen mice (Htn) to evaluate kidney function and injury at 24 weeks. EP1 deletion had no effect on elevation of systolic blood pressure in Htn mice (HtnEP1-/-) but resulted in pronounced albuminuria and reduced FITC-inulin clearance, compared with Htn or wild-type (WT) mice. Ultrastructural injury to podocytes and glomerular endothelium was prominent in HtnEP1-/- mice; including widened subendothelial space, subendothelial lucent zones and focal lifting of endothelium from basement membrane, with focal subendothelial cell debris. Cortex COX2 mRNA was increased by EP1 deletion. Glomerular EP3 mRNA was reduced by EP1 deletion, and EP4 by Htn and EP1 deletion. In WT mice, PGE2 increased chloride reabsorption via EP1 in isolated perfused thick ascending limb (TAL), but PGE2 or EP1 deletion did not affect vasopressin-mediated chloride reabsorption. In WT and Htn mouse inner medullary collecting duct (IMCD), PGE2 inhibited vasopressin-water transport, but not in EP1-/- or HtnEP1-/- mice. Overall, EP1 mediated TAL and IMCD transport in response to PGE2 is unaltered in Htn, and EP1 is protective in HKD.


Assuntos
Hipertensão Renal , Podócitos , Receptores de Prostaglandina E Subtipo EP1 , Animais , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Deleção de Genes , Taxa de Filtração Glomerular/genética , Hipertensão Renal/metabolismo , Hipertensão Renal/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Camundongos , Camundongos Transgênicos , Podócitos/citologia , Podócitos/metabolismo , Podócitos/patologia , Receptores de Prostaglandina E Subtipo EP1/genética , Receptores de Prostaglandina E Subtipo EP1/metabolismo
4.
Clin Sci (Lond) ; 132(13): 1453-1470, 2018 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-29739827

RESUMO

Neuronal ubiquitin C-terminal hydrolase L1 (UCHL1) is a deubiquitinating enzyme that maintains intracellular ubiquitin pools and promotes axonal transport. Uchl1 deletion in mice leads to progressive axonal degeneration, affecting the dorsal root ganglion that harbors axons emanating to the kidney. Innervation is a crucial regulator of renal hemodynamics, though the contribution of neuronal UCHL1 to this is unclear. Immunofluorescence revealed significant neuronal UCHL1 expression in mouse kidney, including periglomerular axons. Glomerular filtration rate trended higher in 6-week-old Uchl1-/- mice, and by 12 weeks of age, these displayed significant glomerular hyperfiltration, coincident with the onset of neurodegeneration. Angiotensin converting enzyme inhibition had no effect on glomerular filtration rate of Uchl1-/- mice indicating that the renin-angiotensin system does not contribute to the observed hyperfiltration. DCE-MRI revealed increased cortical renal blood flow in Uchl1-/- mice, suggesting that hyperfiltration results from afferent arteriole dilation. Nonetheless, hyperglycemia, cyclooxygenase-2, and nitric oxide synthases were ruled out as sources of hyperfiltration in Uchl1-/- mice as glomerular filtration rate remained unchanged following insulin treatment, and cyclooxygenase-2 and nitric oxide synthase inhibition. Finally, renal nerve dysfunction in Uchl1-/- mice is suggested given increased renal nerve arborization, decreased urinary norepinephrine, and impaired vascular reactivity. Uchl1-deleted mice demonstrate glomerular hyperfiltration associated with renal neuronal dysfunction, suggesting that neuronal UCHL1 plays a crucial role in regulating renal hemodynamics.


Assuntos
Taxa de Filtração Glomerular/fisiologia , Doenças Neurodegenerativas/fisiopatologia , Ubiquitina Tiolesterase/fisiologia , Animais , Arteríolas/fisiopatologia , Ciclo-Oxigenase 2/metabolismo , Intolerância à Glucose/fisiopatologia , Rim/inervação , Rim/metabolismo , Camundongos Knockout , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Óxido Nítrico Sintase/metabolismo , Artéria Renal/fisiopatologia , Circulação Renal/fisiologia , Sistema Renina-Angiotensina/fisiologia , Ubiquitina Tiolesterase/deficiência , Ubiquitina Tiolesterase/metabolismo , Resistência Vascular/fisiologia
5.
J Extracell Vesicles ; 7(1): 1432206, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29435202

RESUMO

Tubulointerstitial fibrosis is a hallmark of advanced diabetic kidney disease that is linked to a decline in renal function, however the pathogenic mechanisms are poorly understood. Microparticles (MPs) are 100-1000 nm vesicles shed from injured cells that are implicated in intercellular signalling. Our lab recently observed the formation of MPs from podocytes and their release into urine of animal models of type 1 and 2 diabetes and in humans with type 1 diabetes. The purpose of the present study was to examine the role of podocyte MPs in tubular epithelial cell fibrotic responses. MPs were isolated from the media of differentiated, untreated human podocytes (hPODs) and administered to cultured human proximal tubule epithelial cells (PTECs). Treatment with podocyte MPs increased p38 and Smad3 phosphorylation and expression of the extracellular matrix (ECM) proteins fibronectin and collagen type IV. MP-induced responses were attenuated by co-treatment with the p38 inhibitor SB202190. A transforming growth factor beta (TGF-ß) receptor inhibitor (LY2109761) blocked MP-induced Smad3 phosphorylation and ECM protein expression but not p38 phosphorylation suggesting that these responses occurred downstream of p38. Finally, blockade of the class B scavenger receptor CD36 completely abrogated MP-mediated p38 phosphorylation, downstream Smad3 activation and fibronectin/collagen type IV induction. Taken together our results suggest that podocyte MPs interact with proximal tubule cells and induce pro-fibrotic responses. Such interactions may contribute to the development of tubular fibrosis in glomerular disease.

6.
Lab Invest ; 98(3): 360-370, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29251736

RESUMO

PGE2 regulates glomerular hemodynamics, renin secretion, and tubular transport. This study examined the contribution of PGE2 EP1 receptors to sodium and water homeostasis. Male EP1-/- mice were bred with hypertensive TTRhRen mice (Htn) to evaluate blood pressure and kidney function at 8 weeks of age in four groups: wildtype (WT), EP1-/-, Htn, HtnEP1-/-. Blood pressure and water balance were unaffected by EP1 deletion. COX1 and mPGE2 synthase were increased and COX2 was decreased in mice lacking EP1, with increases in EP3 and reductions in EP2 and EP4 mRNA throughout the nephron. Microdissected proximal tubule sglt1, NHE3, and AQP1 were increased in HtnEP1-/-, but sglt2 was increased in EP1-/- mice. Thick ascending limb NKCC2 was reduced in the cortex but increased in the medulla. Inner medullary collecting duct (IMCD) AQP1 and ENaC were increased, but AVP V2 receptors and urea transporter-1 were reduced in all mice compared to WT. In WT and Htn mice, PGE2 inhibited AVP-water transport and increased calcium in the IMCD, and inhibited sodium transport in cortical collecting ducts, but not in EP1-/- or HtnEP1-/- mice. Amiloride (ENaC) and hydrochlorothiazide (pendrin inhibitor) equally attenuated the effect of PGE2 on sodium transport. Taken together, the data suggest that EP1 regulates renal aquaporins and sodium transporters, attenuates AVP-water transport and inhibits sodium transport in the mouse collecting duct, which is mediated by both ENaC and pendrin-dependent pathways.


Assuntos
Dinoprostona/metabolismo , Hipertensão/metabolismo , Túbulos Renais Coletores/metabolismo , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Sódio/metabolismo , Animais , Aquaporinas/metabolismo , Pressão Sanguínea , Cálcio/metabolismo , Taxa de Filtração Glomerular , Masculino , Camundongos , Prostaglandina-E Sintases/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Trocador 3 de Sódio-Hidrogênio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
7.
Diabetologia ; 59(6): 1318-28, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26995650

RESUMO

AIMS/HYPOTHESIS: The first clinical manifestation of diabetes is polyuria. The prostaglandin E2 (PGE2) receptor EP3 antagonises arginine vasopressin (AVP)-mediated water reabsorption and its expression is increased in the diabetic kidney. The purpose of this work was to study the contribution of EP3 to diabetic polyuria and renal injury. METHODS: Male Ep 3 (-/-) (also known as Ptger3 (-/-)) mice were treated with streptozotocin (STZ) to generate a mouse model of diabetes and renal function was evaluated after 12 weeks. Isolated collecting ducts (CDs) were microperfused to study the contribution of EP3 to AVP-mediated fluid reabsorption. RESULTS: Ep 3 (-/-)-STZ mice exhibited attenuated polyuria and increased urine osmolality compared with wild-type STZ (WT-STZ) mice, suggesting enhanced water reabsorption. Compared with WT-STZ mice, Ep 3 (-/-)-STZ mice also had increased protein expression of aquaporin-1, aquaporin-2, and urea transporter A1, and reduced urinary AVP excretion, but increased medullary V2 receptors. In vitro microperfusion studies indicated that Ep 3 (-/-) and WT-STZ CDs responded to AVP stimulation similarly to those of wild-type mice, with a 60% increase in fluid reabsorption. In WT non-injected and WT-STZ mice, EP3 activation with sulprostone (PGE2 analogue) abrogated AVP-mediated water reabsorption; this effect was absent in mice lacking EP3. A major finding of this work is that Ep 3 (-/-)-STZ mice showed blunted renal cyclooxygenase-2 protein expression, reduced renal hypertrophy, reduced hyperfiltration and reduced albuminuria, as well as diminished tubular dilation and nuclear cysts. CONCLUSIONS/INTERPRETATION: Taken together, the data suggest that EP3 contributes to diabetic polyuria by inhibiting expression of aquaporins and that it promotes renal injury during diabetes. EP3 may prove to be a promising target for more selective management of diabetic kidney disease.


Assuntos
Rim/metabolismo , Poliúria/metabolismo , Receptores de Prostaglandina E Subtipo EP3/metabolismo , Receptores de Prostaglandina E/metabolismo , Estreptozocina/toxicidade , Água/metabolismo , Animais , Aquaporinas/genética , Aquaporinas/metabolismo , Arginina Vasopressina/metabolismo , Modelos Animais de Doenças , Masculino , Camundongos , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP3/genética
8.
J Am Soc Nephrol ; 27(3): 666-76, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26319242

RESUMO

An important measure of cardiovascular health is obtained by evaluating the global cardiovascular risk, which comprises a number of factors, including hypertension and type 2 diabetes, the leading causes of illness and death in the world, as well as the metabolic syndrome. Altered immunity, inflammation, and oxidative stress underlie many of the changes associated with cardiovascular disease, diabetes, and the metabolic syndrome, and recent efforts have begun to elucidate the contribution of PGE2 in these events. This review summarizes the role of PGE2 in kidney disease outcomes that accelerate cardiovascular disease, highlights the role of cyclooxygenase-2/microsomal PGE synthase 1/PGE2 signaling in hypertension and diabetes, and outlines the contribution of PGE2 to other aspects of the metabolic syndrome, particularly abdominal adiposity, dyslipidemia, and atherogenesis. A clearer understanding of the role of PGE2 could lead to new avenues to improve therapeutic options and disease management strategies.


Assuntos
Diabetes Mellitus/metabolismo , Dinoprostona/metabolismo , Hipertensão/metabolismo , Síndrome Metabólica/metabolismo , Insuficiência Renal Crônica/metabolismo , Gordura Abdominal , Aterosclerose/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dislipidemias/metabolismo , Humanos , Hipertensão/complicações , Oxirredutases Intramoleculares/metabolismo , Síndrome Metabólica/complicações , Prostaglandina-E Sintases , Insuficiência Renal Crônica/complicações , Transdução de Sinais
9.
Lab Invest ; 95(9): 1044-55, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26121313

RESUMO

Renal prostaglandin (PG) E2 regulates salt and water transport, and affects disease processes via EP1-4 receptors, but its role in the proximal tubule (PT) is unknown. Our study investigates the effects of PGE2 on mouse PT fluid reabsorption, and its role in growth, sodium transporter expression, fibrosis, and oxidative stress in a mouse PT cell line (MCT). To determine which PGE2 EP receptors are expressed in MCT, qPCR for EP1-4 was performed on cells stimulated for 24 h with PGE2 or transforming growth factor beta (TGFß), a known mediator of PT injury in kidney disease. EP1 and EP4 were detected in MCT, but EP2 and EP3 are not expressed. EP1 was increased by PGE2 and TGFß, but EP4 was unchanged. To confirm the involvement of EP1 and EP4, sulprostone (SLP, EP1/3 agonist), ONO8711 (EP1 antagonist), and EP1 and EP4 siRNA were used. We first show that PGE2, SLP, and TGFß reduced H(3)-thymidine and H(3)-leucine incorporation. The effects on cell-cycle regulators were examined by western blot. PGE2 increased p27 via EP1 and EP4, but TGFß increased p21; PGE2-induced p27 was attenuated by TGFß. PGE2 and SLP reduced cyclinE, while TGFß increased cyclinD1, an effect attenuated by PGE2 administration. Na-K-ATPase α1 (NaK) was increased by PGE2 via EP1 and EP4. TGFß had no effect on NaK. Additionally, PGE2 and TGFß increased fibronectin levels, reaching 12-fold upon co-stimulation. EP1 siRNA abrogated PGE2-fibronectin. PGE2 also increased ROS generation, and ONO-8711 blocked PGE2-ROS. Finally, PGE2 significantly increased fluid reabsorption by 31 and 46% in isolated perfused mouse PT from C57BL/6 and FVB mice, respectively, and this was attenuated in FVB-EP1 null mice. Altogether PGE2 acting on EP1 and EP4 receptors may prove to be important mediators of PT injury, and salt and water transport.


Assuntos
Dinoprostona/farmacologia , Túbulos Renais Proximais/fisiologia , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Reabsorção Renal/efeitos dos fármacos , Acridinas , Análise de Variância , Animais , Western Blotting , Compostos Bicíclicos com Pontes/farmacologia , Caproatos/farmacologia , Ciclina D1/metabolismo , Ciclina E/metabolismo , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Receptores de Prostaglandina E Subtipo EP1/agonistas , Receptores de Prostaglandina E Subtipo EP1/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia
10.
Am J Physiol Renal Physiol ; 307(3): F243-50, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24966087

RESUMO

Chronic kidney disease is a leading cause of morbidity and mortality in the world. A better understanding of disease mechanisms has been gained in recent years, but the current management strategies are ineffective at preventing disease progression. A widespread focus of research is placed on elucidating the specific processes implicated to find more effective therapeutic options. PGE2, acting on its four EP receptors, regulates many renal disease processes; thus EP receptors could prove to be important targets for kidney disease intervention strategies. This review summarizes the major pathogenic mechanisms contributing to initiation and progression of chronic kidney disease, emphasizing the role of hyperglycemia, hypertension, inflammation, and oxidative stress. We have long recognized the multifaceted role of PGs in both the initiation and progression of chronic kidney disease, yet studies are only now seriously contemplating specific EP receptors as targets for therapy. Given the plethora of renal complications attributed to PG involvement in the kidney, this review highlights these pathogenic events and emphasizes the PGE2 receptor targets as options available to complement current therapeutic strategies.


Assuntos
Progressão da Doença , Receptores de Prostaglandina E/fisiologia , Insuficiência Renal Crônica/tratamento farmacológico , Insuficiência Renal Crônica/fisiopatologia , Humanos , Hiperglicemia/fisiopatologia , Hipertensão/fisiopatologia , Inflamação/fisiopatologia , Estresse Oxidativo/fisiologia
11.
Am J Pathol ; 183(6): 1789-1802, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24113456

RESUMO

We hypothesized that the EP1 receptor promotes renal damage in diabetic nephropathy. We rendered EP1 (PTGER1, official symbol) knockout mice (EP1(-/-)) diabetic using the streptozotocin and OVE26 models. Albuminuria, mesangial matrix expansion, and glomerular hypertrophy were each blunted in EP1(-/-) streptozotocin and OVE26 cohorts compared with wild-type counterparts. Although diabetes-associated podocyte depletion was unaffected by EP1 deletion, EP1 antagonism with ONO-8711 in cultured podocytes decreased angiotensin II-mediated superoxide generation, suggesting that EP1-associated injury of remaining podocytes in vivo could contribute to filtration barrier dysfunction. Accordingly, EP1 deletion in OVE26 mice prevented nephrin mRNA expression down-regulation and ameliorated glomerular basement membrane thickening and foot process effacement. Moreover, EP1 deletion reduced diabetes-induced expression of fibrotic markers fibronectin and α-actin, whereas EP1 antagonism decreased fibronectin in cultured proximal tubule cells. Similarly, proximal tubule megalin expression was reduced by diabetes but was preserved in EP1(-/-) mice. Finally, the diabetes-associated increase in angiotensin II-mediated constriction of isolated mesenteric arteries was blunted in OVE26EP1(-/-) mice, demonstrating a role for EP1 receptors in the diabetic vasculature. These data suggest that EP1 activation contributes to diabetic nephropathy progression at several locations, including podocytes, proximal tubule, and the vasculature. The EP1 receptor facilitates the actions of angiotensin II, thereby suggesting that targeting of both the renin-angiotensin system and the EP1 receptor could be beneficial in diabetic nephropathy.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Deleção de Genes , Receptores de Prostaglandina E Subtipo EP1 , Actinas/biossíntese , Actinas/genética , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Compostos Bicíclicos com Pontes/farmacologia , Caproatos/farmacologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Fibronectinas/biossíntese , Fibronectinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Barreira de Filtração Glomerular/metabolismo , Barreira de Filtração Glomerular/patologia , Artérias Mesentéricas/metabolismo , Artérias Mesentéricas/patologia , Camundongos , Camundongos Knockout , Receptores de Prostaglandina E Subtipo EP1/genética , Receptores de Prostaglandina E Subtipo EP1/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/genética , Superóxidos/metabolismo , Vasoconstrição/efeitos dos fármacos
12.
J Am Soc Nephrol ; 24(10): 1512-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23970124

RESUMO

Reactive oxygen species (ROS) play an important role in normal cellular physiology. They regulate different biologic processes such as cell defense, hormone synthesis and signaling, activation of G protein-coupled receptors, and ion channels and kinases/phosphatases. ROS are also important regulators of transcription factors and gene expression. On the other hand, in pathologic conditions, a surplus of ROS in tissue results in oxidative stress with various injurious consequences such as inflammation and fibrosis. NADPH oxidases are one of the many sources of ROS in biologic systems, and there are seven isoforms (Nox1-5, Duox1, Duox2). Nox4 is the predominant form in the kidney, although Nox2 is also expressed. Nox4 has been implicated in the basal production of ROS in the kidney and in pathologic conditions such as diabetic nephropathy and CKD; upregulation of Nox4 may be important in renal oxidative stress and kidney injury. Although there is growing evidence indicating the involvement of NADPH oxidase in renal pathology, there is a paucity of information on the role of NADPH oxidase in the regulation of normal renal function. Here we provide an update on the role of NADPH oxidases and ROS in renal physiology and pathology.


Assuntos
Rim/enzimologia , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Humanos , Rim/metabolismo , Rim/patologia , Nefropatias/enzimologia , Nefropatias/metabolismo , Nefropatias/patologia
13.
Mol Cell Endocrinol ; 381(1-2): 210-9, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23877023

RESUMO

In rats and mice, the renal stanniocalcin-1 (STC-1) gene is expressed in most nephron segments, but is differentially induced in response to dehydration. In cortical segments STC-1 mRNA levels are upregulated by the hypertonicity of dehydration, while hypovolemia causes gene induction in the inner medulla (papilla). In both cases induction is mediated by arginine vasopressin (AVP) acting via the V2 receptor (V2R). The intent of STC-1 gene upregulation during dehydration has yet to be established. Therefore, to narrow down the range of possible actions, we mapped out the pathway by which V2R occupancy upregulates the gene. V2R occupancy activates two different renal pathways in response to dehydration. The first is antidiuretic in nature and is mediated by direct V2R occupancy. The second pathway is indirect and counter-regulates AVP-mediated antidiuresis. It involves COX-2 (cyclooxygenase-2) and the prostanoids, and is activated by the V2R-mediated rise in medullary interstitial osmolality. The resulting prostanoids counter-regulate AVP-mediated antidiuresis. They also upregulate renal cytoprotective mechanisms. The present studies employed models of COX inhibition and COX gene deletion to address the possible involvement of the COX pathway. The results showed that both general and specific inhibitors of COX-2 blocked STC-1 gene induction in response to dehydration. Gene induction in response to dehydration was also abolished in COX-2 null mice (cortex and papilla), but not in COX-1 null mice. STC-1 gene induction in response to V2R occupancy was also uniquely abolished in COX-2 nulls (both regions). These findings therefore collectively suggest that AVP-mediated elevations in STC-1 gene expression are wholly dependent on functional COX-2 activity. As such, a permissive role for STC-1 in AVP-mediated antidiuresis can be ruled out, and its range of possible actions has been narrowed down to AVP counter-regulation and renal cytoprotection.


Assuntos
Arginina Vasopressina/fisiologia , Ciclo-Oxigenase 2/fisiologia , Glicoproteínas/genética , Medula Renal/enzimologia , Ativação Transcricional , Animais , Desidratação/enzimologia , Desidratação/genética , Feminino , Glicoproteínas/metabolismo , Córtex Renal/enzimologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Néfrons/enzimologia , Ratos , Ratos Wistar , Receptores de Ocitocina/agonistas , Receptores de Ocitocina/metabolismo , Regulação para Cima
14.
Clin Sci (Lond) ; 124(11): 685-94, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23305077

RESUMO

The role of COXs/PGs (cyclo-oxygenases/prostaglandins) in diabetic kidneys remains unclear. NSAIDs (non-steroidal anti-inflammatory drugs) that inhibit COXs/PGs are known for their renal toxicity, and COX-2 inhibitors worsen cardiovascular outcomes in susceptible individuals. Given the renal controversies concerning COX-2 inhibitors, we compared the effect of chronic NSAIDs (non-selective, ibuprofen; COX-2-selective, celecoxib) on diabetic kidneys in OVE26 mice from 8 weeks of age. Systolic BPs (blood pressures) were increased by NSAIDs in diabetic mice at 20 weeks, but were unchanged at 32 weeks. Although NSAIDs further increased diabetic kidney/body weight ratios, they did not affect albuminuria. Mesangial matrix was increased 2-fold by celecoxib but not ibuprofen. Electron microscopy revealed that NSAIDs reduced GBM (glomerular basement membrane) thickness and slit pore diameters. Although diabetics had increased glomerular diameters and reduced foot process densities, these were unaltered by NSAIDs. Celecoxib does not exacerbate the diabetic state, but PG inhibition may contribute to disease progression by modifying the GBM, mesangial area and podocyte structure in OVE26 mice. Despite these findings, celecoxib remains safer than a similar dose of ibuprofen. The present study substantiates the need to more closely consider selective COX-2 inhibitors such as celecoxib as alternatives to non-selective NSAIDs for therapeutic management in a setting of chronic kidney disease.


Assuntos
Inibidores de Ciclo-Oxigenase 2/toxicidade , Inibidores de Ciclo-Oxigenase/toxicidade , Nefropatias Diabéticas/patologia , Membrana Basal Glomerular/efeitos dos fármacos , Mesângio Glomerular/efeitos dos fármacos , Ibuprofeno/toxicidade , Pirazóis/toxicidade , Sulfonamidas/toxicidade , Albuminas/metabolismo , Animais , Glicemia , Pressão Sanguínea/efeitos dos fármacos , Celecoxib , Creatinina/sangue , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Diabetes Mellitus Experimental/patologia , Membrana Basal Glomerular/patologia , Membrana Basal Glomerular/ultraestrutura , Mesângio Glomerular/patologia , Masculino , Camundongos , Camundongos Endogâmicos , Podócitos/efeitos dos fármacos , Podócitos/patologia , Prostaglandinas/metabolismo , Fator de Crescimento Transformador beta/metabolismo
15.
Am J Physiol Regul Integr Comp Physiol ; 303(7): R710-8, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22855276

RESUMO

Studies of experimental diabetes mellitus (DM) suggest that increased nitric oxide (NO) bioactivity contributes to renal hyperfiltration. However, the role of NO in mediating hyperfiltration has not been fully elucidated in humans. Our aim was to examine the effect of NO synthase inhibition on renal and peripheral vascular function in normotensive subjects with uncomplicated type 1 DM. Renal function and brachial artery flow-mediated vasodilatation (FMD) were measured before and after an intravenous infusion of the NO synthase inhibitor N(G)-nitro-l-arginine methyl ester (l-NMMA) in 21 healthy control and 37 type 1 DM patients. Measurements in DM participants were made under clamped euglycemic conditions. The effect of l-NMMA on circulating and urinary NO metabolites (NO(x)) and cGMP and on urinary prostanoids was also determined. Baseline characteristics were similar in the two groups. For analysis, the DM patients were divided into those with hyperfiltration (DM-H, n = 18) and normal glomerular filtration rate (GFR) levels (DM-N, n = 19). Baseline urine NO(x) and cGMP were highest in DM-H. l-NMMA led to a decline in GFR in DM-H (152 ± 16 to 140 ± 11 ml·min(-1)·1.73 m(-2)) but not DM-N or healthy control participants. The decline in effective renal plasma flow in response to l-NMMA (806 ± 112 to 539 ± 80 ml·min(-1)·1.73 m(-2)) in DM-H was also exaggerated compared with the other groups (repeated measures ANOVA, P < 0.05), along with declines in urinary NO(x) metabolites and cGMP. Baseline FMD was lowest in DM-H compared with the other groups and did not change in response to l-NMMA. l-NMMA reduced FMD and plasma markers of NO bioactivity in the healthy control and DM-N groups. In patients with uncomplicated type 1 DM, renal hyperfiltration is associated with increased NO bioactivity in the kidney and reduced NO bioactivity in the systemic circulation, suggesting a paradoxical state of high renal and low systemic vascular NO bioactivity.


Assuntos
Diabetes Mellitus Tipo 1/fisiopatologia , Endotélio Vascular/fisiologia , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/fisiologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico/antagonistas & inibidores , ômega-N-Metilarginina/farmacologia , Adulto , Artéria Braquial/efeitos dos fármacos , Artéria Braquial/fisiologia , AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Endotélio Vascular/efeitos dos fármacos , Feminino , Taxa de Filtração Glomerular/fisiologia , Humanos , Infusões Intravenosas , Rim/efeitos dos fármacos , Masculino , Óxido Nítrico/metabolismo , Circulação Renal/efeitos dos fármacos , Circulação Renal/fisiologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia , ômega-N-Metilarginina/administração & dosagem
16.
J Biomed Biotechnol ; 2012: 507057, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22500093

RESUMO

Peroxisome proliferator-activated receptor (PPARγ) has been shown to have a protective role in the nephron through its ability to inhibit a transforming growth factor- (TGF-ß) mediated fibrotic response. In contrast, PPARγ was also shown to induce a mesenchymal transformation in epithelial intestinal cells. A fibrotic response in the collecting duct has only recently been established; however, the entire collecting duct has not been fully examined. Inner medullary collecting duct cells (IMCD-K2) and mouse cortical collecting duct cells (M1), representing the cortical and medullary collecting duct, were exposed to 5-10 µM troglitazone for 24 hours. Troglitazone resulted in an elongated morphology, 60% decreases in E-cadherin and ß-catenin, a 35% decrease in α-catenin, and a 1.5-fold increase in fibronectin. These effects were not reversed with PPARγ antagonists or affected with PPARγ overexpression. Our results indicate that troglitazone induced a mesenchymal-like transformation in M1 and IMCD-K2 epithelial cells independently of PPARγ.


Assuntos
Cromanos/farmacologia , Citoesqueleto/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Actinas/metabolismo , Animais , Caderinas/metabolismo , Cateninas/metabolismo , Linhagem Celular Transformada , Forma Celular/efeitos dos fármacos , Citoesqueleto/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Camundongos , Microscopia de Fluorescência , PPAR gama/metabolismo , Troglitazona
17.
Biochem Biophys Res Commun ; 418(2): 372-7, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22266310

RESUMO

Cyclooxygenase-2 (COX-2) expression is increased by hypertonicity. Therefore we hypothesized that hypertonicity increased PGE(2) can modulate the sodium transporters (Na(+)/K(+)-ATPase: NKA, epithelial sodium channel: ENaC, and sodium hydrogen exchanger: NHE) in M1 cortical collecting duct (CCD) cells. We demonstrated by immunoblotting a 2-fold increase in NKA expression and activity following hypertonic treatment. α-ENaC was also increased, however sgk1, an ENaC activator, decreased in response to hypertonicity. Other CCD sodium transporters (ß-ENaC, NHE) were unchanged. Hypertonicity also increased PGE(2) but EP(4) receptor mRNA was unaltered. PGE(2) increased intracellular Na(+) and cAMP production in M1 cells, but PGE(2)-stimulated cAMP response was attenuated by hypertonicity. Overall, PGE(2) had no effect on sodium transporter levels. Since neither COX inhibition nor EP(4) siRNA altered the induction of NKA, we propose that sodium transporter regulation by hypertonicity is independent of PGE(2). Altogether, these data indicate that despite a concomitant increase in PGE(2) production and sodium transporter expression in hypertonicity, both pathways are acting independently of each other.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Animais , Linhagem Celular , Inibidores de Ciclo-Oxigenase 2/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos , RNA Interferente Pequeno/genética , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/genética , Solução Salina Hipertônica/farmacologia
18.
Diabetes Care ; 33(6): 1344-6, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20332349

RESUMO

OBJECTIVE: Our aim was to examine the effect of cyclooxygenase 2 (COX2) inhibition on endothelial function in subjects with type 1 diabetes analyzed on the basis of renal filtration status. RESEARCH DESIGN AND METHODS: Flow-mediated dilation (FMD) was determined in type 1 diabetic subjects and hyperfiltration (glomerular filtration rate >or=135 ml/min/1.73 m(2), n = 13) or normofiltration (glomerular filtration rate >or=135 ml/min/1.73 m(2), n = 11). Studies were performed before and after celecoxib (200 mg daily for 14 days) during euglycemia and hyperglycemia. RESULTS: Baseline parameters were similar in the two groups. Pretreatment, FMD was augmented in normofiltering versus hyperfiltering subjects during clamped euglycemia (10.2 +/- 5.3% vs. 5.9 +/- 2.3%, P = 0.003). COX2 inhibition suppressed FMD in normofiltering (10.2 +/- 5.3% to 5.8 +/- 3.4%, P = 0.006) versus hyperfiltering subjects (ANOVA interaction, P = 0.003). CONCLUSIONS: Systemic hemodynamic function, including the response to COX2 inhibition, is related to filtration status in diabetic subjects and may reflect general endothelial dysfunction.


Assuntos
Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/fisiopatologia , Rim/fisiopatologia , Pirazóis/uso terapêutico , Sulfonamidas/uso terapêutico , Adolescente , Adulto , Celecoxib , Inibidores de Ciclo-Oxigenase 2/farmacologia , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Adulto Jovem
19.
Am J Physiol Renal Physiol ; 298(5): F1105-12, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20164153

RESUMO

Peroxisome proliferator-activated receptor (PPAR)-γ is highly expressed in the collecting duct (CD), yet little is known about the effects of PPAR-γ ligands, thiazolidinediones (TZDs), on CD cell structure and function. M1 mouse cortical CD cells were treated with 5 µM troglitazone (TRO) and rosiglitazone (ROSI). First, growth was measured by [(3)H]thymidine and [(3)H]leucine incorporation, as well as analysis of cyclin D1 and the CDK inhibitor p27 by Western blot. [(3)H]thymidine incorporation was reduced by 56 and 24% by TRO and ROSI at 6 h, and [(3)H]leucine by 21 and 10%. A similar growth inhibition was also observed after 24 h for thymidine, but leucine was reduced by 48 and 24%, respectively. Likewise, cyclin D1 was diminished 60% by TRO, and p27 was elevated 1.6- and 1.7-fold in response to TRO and ROSI. Next, epithelial cell integrity was assessed by measuring different markers by Western blot analysis. While fibronectin and α-smooth muscle actin levels were unchanged, by 24 h E-cadherin was decreased by 50%, and ß-catenin levels were reduced 2- and 1.5-fold in response to TRO and ROSI, respectively. GW9662, a PPAR-γ antagonist, did not reverse any of the TZD responses in M1 cells. Of interest, phosho-p38 levels were also elevated 2-fold in response to TRO and 2.3-fold to ROSI, but MAPK inhibition by PD98059 or SB203580 caused an additive inhibition of cell growth and did not alter E-cadherin or ß-catenin in response to TZDs. Finally, apoptotic death was assessed by Western blot, but cleaved caspase-3 levels were unchanged from 15 min to 24 h in response to TZDs, and TRO did not affect cell viability or reactive oxygen species generation. Our data suggest that TZDs cause a disruption of M1 cell integrity that is preceded by an inhibition of cell growth. This response is independent of p38 or PPAR-γ activation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , PPAR gama/metabolismo , Tiazolidinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Caderinas/metabolismo , Cromanos/farmacologia , Túbulos Renais Coletores/citologia , Camundongos , Camundongos Transgênicos , Modelos Animais , Rosiglitazona , Tiazolidinedionas/farmacologia , Troglitazona , beta Catenina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Am J Nephrol ; 30(4): 346-53, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19609076

RESUMO

BACKGROUND/AIMS: The widespread use of non-steroidal anti-inflammatory drugs (NSAIDs) and the sizeable impact of diabetes on the development of end-stage renal disease substantiate the need to determine their effect on the evolution of diabetic nephropathy (DN). We hypothesized that chronic ibuprofen and NS-398 will differentially affect many aspects of DN in B6-Ins2(Akita) mice, including glomerular filtration rate (GFR), growth, and fibrosis. METHODS: B6-Ins2(Akita) and wild-type mice were separated into six groups. At 20 weeks of age, NSAIDs (1 mg/kg/day) were added to the drinking water daily. At 36 weeks indicators of renal function and DN were examined. RESULTS: Urinary PGE(2), PGEM, TXB(2), and 6-keto-PGF(1)alpha were increased in diabetics, and reduced by NSAIDs. Regional differences in cyclooxygenases were observed. Diabetics displayed hyperglycemia, albuminuria, and increased kidney/body weights, glomerular diameters, and FITC-inulin clearance. NSAIDs did not affect growth, but albuminuria and FITC-inulin clearance were reduced. Also, p27 and fibronectin were increased in diabetics and attenuated by ibuprofen. CONCLUSION: NSAIDs reduced diabetic change: GFR, albuminuria, p27, and fibronectin. The effects of ibuprofen are similar if not more beneficial than COX-2 inhibition by NS-398. This study has clinical relevance for diabetics prior to overt nephropathy. Future studies should reveal the effects of NSAIDs in a more severe disease environment.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Diabetes Mellitus Tipo 1/complicações , Nefropatias Diabéticas/tratamento farmacológico , Ibuprofeno/farmacologia , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Albuminúria/tratamento farmacológico , Albuminúria/metabolismo , Albuminúria/patologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Biomarcadores/metabolismo , Peso Corporal , Creatinina/urina , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Modelos Animais de Doenças , Fibronectinas/metabolismo , Rim/metabolismo , Rim/patologia , Falência Renal Crônica/tratamento farmacológico , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Tamanho do Órgão , Prostaglandinas/urina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA