Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Int J Mol Sci ; 24(22)2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-38003732

RESUMO

Peritubular capillary rarefaction is a recurrent aspect of progressive nephropathies. We previously found that peritubular capillary density was reduced in BTBR ob/ob mice with type 2 diabetic nephropathy. In this model, we searched for abnormalities in the ultrastructure of peritubular capillaries, with a specific focus on the endothelial glycocalyx, and evaluated the impact of treatment with an angiotensin-converting enzyme inhibitor (ACEi). Mice were intracardially perfused with lanthanum to visualise the glycocalyx. Transmission electron microscopy analysis revealed endothelial cell abnormalities and basement membrane thickening in the peritubular capillaries of BTBR ob/ob mice compared to wild-type mice. Remodelling and focal loss of glycocalyx was observed in lanthanum-stained diabetic kidneys, associated with a reduction in glycocalyx components, including sialic acids, as detected through specific lectins. ACEi treatment preserved the endothelial glycocalyx and attenuated the ultrastructural abnormalities of peritubular capillaries. In diabetic mice, peritubular capillary damage was associated with an enhanced tubular expression of heparanase, which degrades heparan sulfate residues of the glycocalyx. Heparanase was also detected in renal interstitial macrophages that expressed tumor necrosis factor-α. All these abnormalities were mitigated by ACEi. Our findings suggest that, in experimental diabetic nephropathy, preserving the endothelial glycocalyx is important in order to protect peritubular capillaries from damage and loss.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Camundongos , Animais , Nefropatias Diabéticas/metabolismo , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/metabolismo , Diabetes Mellitus Experimental/metabolismo , Capilares/patologia , Glicocálix/metabolismo , Lantânio , Rim/patologia , Camundongos Endogâmicos
2.
Int J Mol Sci ; 23(15)2022 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-35955472

RESUMO

Sirtuin 3 (SIRT3) is the primary mitochondrial deacetylase that controls the antioxidant pathway and energy metabolism. We previously found that renal Sirt3 expression and activity were reduced in mice with type 2 diabetic nephropathy associated with oxidative stress and mitochondrial abnormalities and that a specific SIRT3 activator improved renal damage. SIRT3 is modulated by diet, and to assess whether Sirt3 deficiency aggravates mitochondrial damage and accelerates kidney disease in response to nutrient overloads, wild-type (WT) and Sirt3-/- mice were fed a high-fat-diet (HFD) or standard diet for 8 months. Sirt3-/- mice on HFD exhibited earlier and more severe albuminuria compared to WT mice, accompanied by podocyte dysfunction and glomerular capillary rarefaction. Mesangial matrix expansion, tubular vacuolization and inflammation, associated with enhanced lipid accumulation, were more evident in Sirt3-/- mice. After HFD, kidneys from Sirt3-/- mice showed more oxidative stress than WT mice, mitochondria ultrastructural damage in tubular cells, and a reduction in mitochondrial mass and energy production. Our data demonstrate that Sirt3 deficiency renders mice more prone to developing oxidative stress and mitochondrial abnormalities in response to HFD, resulting in more severe kidney diseases, and this suggests that mitochondria protection may be a method to prevent HFD-induced renal injury.


Assuntos
Nefropatias Diabéticas , Sirtuína 3/metabolismo , Animais , Antioxidantes/metabolismo , Dieta Hiperlipídica , Camundongos , Camundongos Knockout , Estresse Oxidativo , Sirtuína 3/genética
3.
Diabetologia ; 60(6): 1114-1125, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28364255

RESUMO

AIMS/HYPOTHESIS: Renal fibrosis is a common complication of diabetic nephropathy and is a major cause of end-stage renal disease. Despite the suggested link between renal fibrosis and microRNA (miRNA) dysregulation in diabetic nephropathy, the identification of the specific miRNAs involved is still incomplete. The aim of this study was to investigate miRNA profiles in the diabetic kidney and to identify potential downstream targets implicated in renal fibrosis. METHODS: miRNA expression profiling was investigated in the kidneys of 8-month-old Zucker diabetic fatty (ZDF) rats during overt nephropathy. Localisation of the most upregulated miRNA was established by in situ hybridisation. The candidate miRNA target was identified by in silico analysis and its expression documented in the diabetic kidney associated with fibrotic markers. Cultured tubule cells served to assess which of the profibrogenic stimuli acted as a trigger for the overexpressed miRNA, and to investigate underlying epigenetic mechanisms. RESULTS: In ZDF rats, miR-184 showed the strongest differential upregulation compared with lean rats (18-fold). Tubular localisation of miR-184 was associated with reduced expression of lipid phosphate phosphatase 3 (LPP3) and collagen accumulation. Transfection of NRK-52E cells with miR-184 mimic reduced LPP3, promoting a profibrotic phenotype. Albumin was a major trigger of miR-184 expression. Anti-miR-184 counteracted albumin-induced LPP3 downregulation and overexpression of plasminogen activator inhibitor-1. In ZDF rats, ACE-inhibitor treatment limited albuminuria and reduced miR-184, with tubular LPP3 preservation and tubulointerstitial fibrosis amelioration. Albumin-induced miR-184 expression in tubule cells was epigenetically regulated through DNA demethylation and histone lysine acetylation and was accompanied by binding of NF-κB p65 subunit to miR-184 promoter. CONCLUSIONS/INTERPRETATION: These results suggest that miR-184 may act as a downstream effector of albuminuria through LPP3 to promote tubulointerstitial fibrosis, and offer the rationale to investigate whether targeting miR-184 in association with albuminuria-lowering drugs may be a new strategy to achieve fully anti-fibrotic effects in diabetic nephropathy.


Assuntos
Albuminúria/metabolismo , Nefropatias Diabéticas/metabolismo , Fibrose/metabolismo , Nefropatias/metabolismo , MicroRNAs/metabolismo , Albuminúria/genética , Animais , Imunoprecipitação da Cromatina , Biologia Computacional , Nefropatias Diabéticas/genética , Fibrose/genética , Imuno-Histoquímica , Hibridização In Situ , Nefropatias/genética , Masculino , MicroRNAs/genética , NF-kappa B/metabolismo , Fosfatidato Fosfatase/genética , Fosfatidato Fosfatase/metabolismo , Regiões Promotoras Genéticas/genética , Ratos , Ratos Zucker , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Am Soc Nephrol ; 27(3): 699-705, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26116358

RESUMO

Chronic renal insufficiency inexorably progresses in patients, such as it does after partial renal ablation in rats. However, the progression of renal diseases can be delayed by angiotensin II blockers that stabilize renal function or increase GFR, even in advanced phases of the disease. Regression of glomerulosclerosis can be induced by angiotensin II antagonism, but the effect of these treatments on the entire vascular tree is unclear. Here, using microcomputed tomography and scanning electron microscopy, we compared the size and extension of kidney blood vessels in untreated Wistar rats with those in untreated and angiotensin II antagonist-treated Munich Wistar Frömter (MWF) rats that spontaneously develop kidney disease with age. The kidney vasculature underwent progressive rarefaction in untreated MWF rats, substantially affecting intermediate and small vessels. Microarray analysis showed increased Tgf-ß and endothelin-1 gene expression with age. Notably, 10-week inhibition of the renin-angiotensin system regenerated kidney vasculature and normalized Tgf-ß and endothelin-1 gene expression in aged MWF rats. These changes were associated with reduced apoptosis, increased endothelial cell proliferation, and restoration of Nrf2 expression, suggesting mechanisms by which angiotensin II antagonism mediates regeneration of capillary segments. These results have important implications in the clinical setting of chronic renal insufficiency.


Assuntos
Antagonistas de Receptores de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Capilares/fisiologia , Glomérulos Renais/irrigação sanguínea , Neovascularização Fisiológica/efeitos dos fármacos , Insuficiência Renal Crônica/tratamento farmacológico , Actinas/metabolismo , Animais , Apoptose , Capilares/metabolismo , Capilares/ultraestrutura , Proliferação de Células , Células Endoteliais/fisiologia , Endotelina-1/genética , Expressão Gênica , Microscopia Eletrônica de Varredura , Fator 2 Relacionado a NF-E2/metabolismo , Ratos , Ratos Wistar , Sistema Renina-Angiotensina/efeitos dos fármacos , Fator de Crescimento Transformador beta/genética , Microtomografia por Raio-X
5.
Am J Pathol ; 183(1): 119-30, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23707238

RESUMO

In nondiabetic rat models of renal disease, angiotensin II (Ang II) perpetuates podocyte injury and promotes progression to end-stage kidney disease. Herein, we wanted to explore the role of Ang II in diabetic nephropathy by a translational approach spanning from in vitro to in vivo rat and human studies, and to dissect the intracellular pathways involved. In isolated perfused rat kidneys and in cultured human podocytes, Ang II down-regulated nephrin expression via Notch1 activation and nuclear translocation of Snail. Hairy enhancer of split-1 was a Notch1-downstream gene effector that activated Snail in cultured podocytes. In vitro changes of the Snail/nephrin axis were similar to those in renal biopsy specimens of Zucker diabetic fatty rats and patients with advanced diabetic nephropathy, and were normalized by pharmacological inhibition of the renin-angiotensin system. Collectively, the present studies provide evidence that Ang II plays a relevant role in perpetuating glomerular injury in experimental and human diabetic nephropathy via persistent activation of Notch1 and Snail signaling in podocytes, eventually resulting in down-regulation of nephrin expression, the integrity of which is crucial for the glomerular filtration barrier.


Assuntos
Angiotensina II/metabolismo , Nefropatias Diabéticas/metabolismo , Proteínas de Membrana/metabolismo , Receptor Notch1/metabolismo , Fatores de Transcrição/metabolismo , Idoso , Animais , Estudos de Casos e Controles , Células Cultivadas , Diabetes Mellitus Tipo 2/complicações , Regulação para Baixo , Feminino , Humanos , Imuno-Histoquímica , Rim/metabolismo , Rim/patologia , Modelos Lineares , Masculino , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição da Família Snail
6.
J Am Soc Nephrol ; 23(9): 1496-505, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22822076

RESUMO

The contribution of microRNA (miRNA) to the pathogenesis of renal fibrosis is not well understood. Here, we investigated whether miRNA modulates the fibrotic process in Munich Wistar Fromter (MWF) rats, which develop spontaneous progressive nephropathy. We analyzed the expression profile of miRNA in microdissected glomeruli and found that miR-324-3p was the most upregulated. In situ hybridization localized miR-324-3p to glomerular podocytes, parietal cells of Bowman's capsule, and most abundantly, cortical tubules. A predicted target of miR-324-3p is prolyl endopeptidase (Prep), a serine peptidase involved in the metabolism of angiotensins and the synthesis of the antifibrotic peptide N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP). In cultured tubular cells, transient transfection with a miR-324-3p mimic reduced Prep protein and activity, validating Prep as a target of this miRNA. In MWF rats, upregulation of miR-324-3p associated with markedly reduced expression of Prep in both glomeruli and tubules, low urine Ac-SDKP, and increased deposition of collagen. ACE inhibition downregulated glomerular and tubular miR-324-3p, promoted renal Prep expression, increased plasma and urine Ac-SDKP, and attenuated renal fibrosis. In summary, these results suggest that dysregulation of the miR-324-3p/Prep pathway contributes to the development of fibrosis in progressive nephropathy. The renoprotective effects of ACE inhibitors may result, in part, from modulation of this pathway, suggesting that it may hold other potential therapeutic targets.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Rim/metabolismo , Rim/patologia , MicroRNAs/metabolismo , Peptidil Dipeptidase A/efeitos dos fármacos , Peptidil Dipeptidase A/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Endopeptidases/metabolismo , Fibrose , Técnicas In Vitro , Nefropatias/metabolismo , Nefropatias/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Masculino , Ratos , Ratos Wistar , Regulação para Cima
7.
Nephron ; 145(4): 428-444, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33910203

RESUMO

BACKGROUND/AIM: Necrotizing crescentic glomerulonephritis (GN) associated with anti-neutrophil cytoplasmic antibodies (ANCA) against myeloperoxidase (MPO) is a devastating disease that quickly progresses to kidney failure. Current therapies are broadly immunosuppressive and associated with adverse effects. We wanted to set up a model that could be suitable for testing narrowly targeted therapies. METHODS: The model was constructed in male Wistar Kyoto rats through injections of human MPO (hMPO) and pertussis toxin, followed by a sub-nephritogenic dose of sheep anti-rat glomerular basement membrane (GBM) serum to boost the disease. Rats were monitored for 35 days. Rats given hMPO alone, saline, or human serum albumin with or without anti-GBM serum were also studied. RESULTS: Rats receiving hMPO developed circulating anti-hMPO and anti-rat MPO antibodies. Challenging hMPO-immunized rats with the anti-GBM serum led to more glomerular neutrophil infiltration and MPO release, and severe haematuria, heavy proteinuria, and higher blood urea nitrogen than hMPO alone. Pauci-immune GN developed with crescents, affecting 25% of glomeruli. The majority of crescents were fibrocellular. Necrotizing lesions and Bowman capsule ruptures were detected. Cells double positive for claudin-1 (a marker of parietal epithelial cells [PECs]) and neural cell adhesion molecule (NCAM; progenitor PECs) were present in crescents. Double staining for NCAM and Ki-67 established proliferative status of progenitor PECs. Podocyte damage was associated with endothelial and GBM changes by electron microscopy. Monocyte/macrophages and CD4+ and CD8+ T cells accumulated in glomeruli and the surrounding area and in the tubulointerstitium. Lung haemorrhage also manifested. CONCLUSION: This model reflects histological lesions of human ANCA-associated rapidly progressive GN and may be useful for investigating new therapies.


Assuntos
Anticorpos Anticitoplasma de Neutrófilos/imunologia , Glomerulonefrite/imunologia , Peroxidase/imunologia , Animais , Nitrogênio da Ureia Sanguínea , Cápsula Glomerular/patologia , Células Epiteliais/patologia , Membrana Basal Glomerular/imunologia , Hematúria/etiologia , Humanos , Glomérulos Renais/patologia , Masculino , Infiltração de Neutrófilos , Toxina Pertussis/farmacologia , Proteinúria/etiologia , Ratos , Ratos Endogâmicos WKY
8.
Am J Pathol ; 174(3): 797-807, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19164508

RESUMO

Angiotensin-converting enzyme (ACE) inhibition induces glomerular repair in the Munich Wistar Frömter (MWF) rat, a model of spontaneous glomerular injury. In this study, we investigated whether this effect is related to changes in glomerular cell number, particularly of podocytes, which are progressively lost with age. MWF rats with advanced nephropathy were studied at both 40 weeks and after 20 weeks of observation either with or without treatment with the ACE inhibitor lisinopril. Forty-week-old Wistar rats were used as controls. In untreated MWF rats, proteinuria, hypertension, glomerulosclerosis, and renal function worsened, while lisinopril induced regression of both functional and structural changes. Despite glomerular hypercellularity in untreated MWF rats, the number of endothelial cells per glomerulus did not change, and podocyte number even decreased. ACE inhibition halted the progressive increase in glomerular cell number and enhanced endothelial cell volume density. Surprisingly, lisinopril not only halted age-related podocyte loss but also increased the number of glomerular podocytes above baseline, which was associated with an increased number of proliferating Wilms tumor 1-positive cells, loss of cyclin-dependent kinase inhibitor p27 expression, and increased number of parietal podocytes. These data indicate that ACE inhibition restructures glomerular capillary, primarily by restoring the podocyte population in this model of glomerular injury. Increased parietal podocyte number in lisinopril-treated MWF rats suggests that the remodeling of Bowman's capsule epithelial cells contributes to this effect.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/toxicidade , Glomérulos Renais/patologia , Lisinopril/uso terapêutico , Podócitos/citologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Divisão Celular , Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Glomerulosclerose Segmentar e Focal/prevenção & controle , Hipertensão/tratamento farmacológico , Hipertensão/prevenção & controle , Testes de Função Renal , Glomérulos Renais/efeitos dos fármacos , Masculino , Proteinúria/tratamento farmacológico , Proteinúria/prevenção & controle , Ratos , Ratos Endogâmicos WF , Ratos Wistar
9.
J Am Soc Nephrol ; 20(1): 123-30, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19092126

RESUMO

The role of dendritic cells (DC) that accumulate in the renal parenchyma of non-immune-mediated proteinuric nephropathies is not well understood. Under certain circumstances, DC capture immunologically ignored antigens, including self-antigens, and present them within MHC class I, initiating an autoimmune response. We studied whether DC could generate antigenic peptides from the self-protein albumin. Exposure of rat proximal tubular cells to autologous albumin resulted in its proteolytic cleavage to form an N-terminal 24-amino acid peptide (ALB1-24). This peptide was further processed by the DC proteasome into antigenic peptides that had binding motifs for MHC class I and were capable of activating syngeneic CD8+ T cells. In vivo, the rat five-sixths nephrectomy model allowed the localization and activation of renal DC. Accumulation of DC in the renal parenchyma peaked 1 wk after surgery and decreased at 4 wk, concomitant with their appearance in the renal draining lymph nodes. DC from renal lymph nodes, loaded with ALB1-24, activated syngeneic CD8+ T cells in primary culture. The response of CD8+ T cells of five-sixths nephrectomized rats was amplified with secondary stimulation. In contrast, DC from renal lymph nodes of five-sixths nephrectomized rats treated with the proteasomal inhibitor bortezomib lost their capacity to stimulate CD8+ T cells in primary and secondary cultures. These data suggest that albumin can be a source of potentially antigenic peptides upon renal injury and that renal DC play a role in processing self-proteins through a proteasome-dependent pathway.


Assuntos
Albuminas/metabolismo , Apresentação de Antígeno , Células Dendríticas/fisiologia , Rim/imunologia , Complexo de Endopeptidases do Proteassoma/fisiologia , Animais , Antígeno CD11c/análise , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Tolerância Imunológica , Túbulos Renais Proximais/metabolismo , Inibidores de Proteassoma , Proteinúria/imunologia , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
10.
Front Pharmacol ; 11: 586892, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33519447

RESUMO

Diabetic nephropathy (DN) is one of the major microvascular complications of diabetes mellitus and the leading cause of end-stage kidney disease. The standard treatments for diabetic patients are glucose and blood pressure control, lipid lowering, and renin-angiotensin system blockade; however, these therapeutic approaches can provide only partial renoprotection if started late in the course of the disease. One major limitation in developing efficient therapies for DN is the complex pathobiology of the diabetic kidney, which undergoes a set of profound structural, metabolic and functional changes. Despite these difficulties, experimental models of diabetes have revealed promising therapeutic targets by identifying pathways that modulate key functions of podocytes and glomerular endothelial cells. In this review we will describe recent advances in the field, analyze key molecular pathways that contribute to the pathogenesis of the disease, and discuss how they could be modulated to prevent or reverse DN.

11.
Kidney Int ; 74(9): 1112-4, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18854846

RESUMO

Chen et al. show that candesartan (Cand) displays an antioxidant action independent of angiotensin type 1 receptor (AT1R) blockade that translates into a superior renoprotection of chronic renal inflammation. The peculiarity of Cand in combining AT1R-blocking activity and an antioxidant action could have important theoretical implications for therapy.


Assuntos
Benzimidazóis/farmacologia , Nefropatias/prevenção & controle , Tetrazóis/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Antioxidantes/farmacologia , Compostos de Bifenilo , Humanos , Inflamação/prevenção & controle , Nefropatias/patologia , Substâncias Protetoras/farmacologia
12.
Antioxid Redox Signal ; 25(4): 185-99, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-26972664

RESUMO

SIGNIFICANCE: Mitochondrial integrity is instrumental in protecting against damage associated with aging and a variety of chronic disease conditions. Mitochondrial silent information regulator 3 (Sirt3) plays pivotal roles in maintaining mitochondrial homeostasis by regulating different aspects of the organelle processes. RECENT ADVANCES: Mitochondria are highly dynamic organelles that constantly fuse and divide to maintain normal cell function, and perturbation in mitochondrial dynamics is responsible for mitochondrial dysfunction. Improved knowledge of mitochondrial physiology has disclosed the pleiotropic role of Sirt3 in mitochondria and shows how alterations in protein expression and/or activity may have an important impact on aging-associated organ dysfunction. CRITICAL ISSUES: This review describes updated experimental evidence on the role of mitochondrial dysfunction during aging and renal diseases and highlights the emerging role of Sirt3 as a crucial regulator of mitochondrial dynamics. FUTURE DIRECTIONS: Strategies that activate Sirt3 may offer attractive therapies to achieve healthy longevity and preserve functional integrity of multiple organs. Antioxid. Redox Signal. 25, 185-199.


Assuntos
Envelhecimento/genética , Suscetibilidade a Doenças , Longevidade/genética , Dinâmica Mitocondrial/genética , Sirtuína 3/genética , Envelhecimento/metabolismo , Animais , Metabolismo Energético/genética , Regulação da Expressão Gênica , Humanos , Nefropatias/genética , Nefropatias/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Especificidade de Órgãos/genética , Biogênese de Organelas , Oxirredução , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Sirtuína 3/metabolismo
13.
Thromb Haemost ; 93(3): 443-52, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15735793

RESUMO

Endothelial injury is the central factor in the events leading to thrombotic microangiopathy (TMA); however, the mechanisms involved are not fully understood. Here we investigate the role of neutrophils (PMNs) and of complement activation in inducing microvascular damage and loss of thromboresistance in TMA associated with ADAMTS-13 deficiency. PMNs isolated during the acute phase of the disease released excessive amounts of reactive-oxygen species (ROS), N-derived oxidants and proteinases and induced damage and thromboresistance loss in human microvascular endothelial cell line (HMEC-1) ex vivo. Endothelial cytotoxicity and thromboresistance loss was also induced by TMA serum. Complement-derived products were responsible for the above effects: in fact, TMA serum caused C3 and Membrane Attack Complex (MAC) deposition on HMEC-1 and its cytotoxic effect was abolished by complement inhibition. TMA serum caused surface expression of P-selectin on HMEC-1 which may promote PMN adhesion and resulted in increased PMN cytotoxicity, indicating that complement may have a role in PMN activation. In addition, TMA serum stimulated control PMNs to release ROS and proteinases, and to cause endothelial cell cytotoxicity. All of the above effects were abrogated by complement inactivation. These data document for the first time that complement-initiated PMN activation and endothelial injury may have a crucial role in microvascular thrombosis of TMA associated with ADAMTS-13 deficiency.


Assuntos
Proteínas ADAM/deficiência , Ativação do Complemento/fisiologia , Síndrome Hemolítico-Urêmica/etiologia , Púrpura Trombocitopênica Trombótica/etiologia , Proteína ADAMTS13 , Linhagem Celular , Endotélio Vascular/patologia , Humanos , Ativação de Neutrófilo/imunologia , Oxidantes/sangue , Peptídeo Hidrolases/sangue , Espécies Reativas de Oxigênio/sangue
14.
PLoS One ; 10(5): e0127172, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25993470

RESUMO

BACKGROUND: Angiotensin II promotes insulin resistance. The mechanism underlying this abnormality, however, is still poorly defined. In a different setting, skeletal muscle metabolism and insulin signaling are regulated by Sirtuin3. OBJECTIVE: Here, we investigate whether angiotensin II-induced insulin resistance in skeletal muscle is associated with Sirtuin3 dysregulation and whether pharmacological manipulation of Sirtuin3 confers protection. STUDY DESIGN: Parental and GLUT4-myc L6 rat skeletal muscle cells exposed to angiotensin II are used as in vitro models of insulin resistance. GLUT4 translocation, glucose uptake, intracellular molecular signals such as mitochondrial reactive oxygen species, Sirtuin3 protein expression and activity, along with its downstream targets and upstream regulators, are analyzed both in the absence and presence of acetyl-L-carnitine. The role of Sirtuin3 in GLUT4 translocation and intracellular molecular signaling is also studied in Sirtuin3-silenced as well as over-expressing cells. RESULTS: Angiotensin II promotes insulin resistance in skeletal muscle cells via mitochondrial oxidative stress, resulting in a two-fold increase in superoxide generation. In this context, reactive oxygen species open the mitochondrial permeability transition pore and significantly lower Sirtuin3 levels and activity impairing the cell antioxidant defense. Angiotensin II-induced Sirtuin3 dysfunction leads to the impairment of AMP-activated protein kinase/nicotinamide phosphoribosyltransferase signaling. Acetyl-L-carnitine, by lowering angiotensin II-induced mitochondrial superoxide formation, prevents Sirtuin3 dysfunction. This phenomenon implies the restoration of manganese superoxide dismutase antioxidant activity and AMP-activated protein kinase activation. Acetyl-L-carnitine protection is abrogated by specific Sirtuin3 siRNA. CONCLUSIONS: Our data demonstrate that angiotensin II-induced insulin resistance fosters mitochondrial superoxide generation, in turn leading to Sirtuin3 dysfunction. The present results also highlight Sirtuin3 as a therapeutic target for the insulin-sensitizing effects of acetyl-L-carnitine.


Assuntos
Angiotensina II/farmacologia , Resistência à Insulina , Músculo Esquelético/fisiologia , Sirtuína 3/fisiologia , Acetilcarnitina/farmacologia , Animais , Linhagem Celular , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Músculo Esquelético/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo
15.
Am J Kidney Dis ; 40(4): 783-93, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12324914

RESUMO

BACKGROUND: Activation of polymorphonuclear neutrophils (PMNs) and monocytes has been described during hemodialysis (HD), which results in the release of reactive oxygen species and cytokines. Acetate-free biofiltration (AFB) has been shown to cause less monocyte activation and cytokine release than bicarbonate HD (BHD). No data are available to date on the effect of AFB on PMN activation. METHODS: We studied ex vivo superoxide anion release by PMNs isolated from nine patients treated in random order with AFB or BHD (three sessions each). Plasma interleukin-1beta (IL-1beta) levels and the nitric oxide (NO) synthetic pathway also were evaluated. A polyacrylonitrile (AN69; Hospal, Bologna, Italy) dialyzer was used for both treatments. Fourteen healthy volunteers were used as controls. Blood samples were drawn predialysis and 5 and 15 minutes after starting dialysis to obtain plasma and PMNs. RESULTS: Neither ex vivo superoxide anion release nor blood PMN count was affected by AFB. Conversely, a peak in superoxide anion production associated with a decrease in PMN count was observed at 5 minutes during BHD. Results of superoxide anion production by control PMNs exposed in vitro to AFB or bicarbonate dialysis bath or Hank's balanced salt solution supplemented with bicarbonate or acetate indicated that BHD-induced PMN activation could be attributed to the amount of bicarbonate present in the dialysis bath. IL-1beta plasma levels did not change during dialysis with AFB and were numerically higher at 5 and 15 minutes with respect to predialysis values during BHD. Uremic plasma obtained during either AFB or BHD induced greater NO synthesis by human umbilical vein endothelial cells than control plasma. CONCLUSION: AFB, unlike BHD, does not cause PMN and monocyte activation, which could have a positive impact on dialysis-associated cardiovascular disease of dialyzed patients.


Assuntos
Bicarbonatos/farmacologia , Hemodiafiltração/métodos , Ativação de Neutrófilo , Diálise Renal/métodos , Idoso , Separação Celular , Células Cultivadas , Meios de Cultivo Condicionados/química , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Endotoxinas/análise , Feminino , Hemodiafiltração/efeitos adversos , Soluções para Hemodiálise/química , Humanos , Interleucina-1/sangue , Teste do Limulus , Masculino , Pessoa de Meia-Idade , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/metabolismo , Óxido Nítrico/metabolismo , Diálise Renal/instrumentação , Superóxidos/metabolismo , Veias Umbilicais/citologia , Uremia/sangue
16.
Kidney Int Suppl (2011) ; 4(1): 58-64, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26312151

RESUMO

Interstitial fibrosis represents the final common pathway of any form of progressive renal disease. The severity of tubular interstitial damage is highly correlated to the degree of decline of renal function, even better than the glomerular lesions do. Angiotensin II (Ang II), the main effector of the renin-angiotensin system, is a critical promoter of fibrogenesis. It represents a nexus among glomerular capillary hypertension, barrier dysfunction, and renal tubular injury caused by abnormally filtered proteins. Transforming growth factor (TGF)-ß1 and reactive oxygen species (ROS) are the key mediators of the pro-fibrotic effect of Ang II causing apoptosis and epithelial-to-mesenchymal transition of the renal tubular epithelium. Recent studies link fibrosis to changes of microRNA (miRNA) modulated by Ang II through TGF-ß1, unraveling that antifibrotic action of Ang II antagonism is attributable to epigenetic control of fibrosis-associated genes. Other mechanisms of Ang II-induced fibrosis include ROS-dependent activation of hypoxia-inducible factor-1. Finally, Ang II via angiotensin type 1 receptor regulates the activation and transdifferentiation of pericytes and fibrocytes into scar-forming myofibroblasts. Detachment and phenotypic changes of the former can lead to the loss of peritubular capillaries and also contribute to hypoxia-dependent fibrosis.

17.
Histol Histopathol ; 25(5): 655-68, 2010 05.
Artigo em Inglês | MEDLINE | ID: mdl-20238303

RESUMO

In the last few years great progress has been made in the search for the cellular and molecular mechanisms of chronic kidney disease and its progression to end-stage renal failure. The possibility of remission/regression of chronic nephropathy has become a reality for some patients on therapy based on renin-angiotensin system blockade - an example of how a public health concern can be successfully addressed by translational medicine. This review describes experimental and clinical investigations documenting the advances achieved in the management of chronic kidney diseases by targeting angiotensin II.


Assuntos
Insuficiência Renal Crônica/tratamento farmacológico , Sistema Renina-Angiotensina/efeitos dos fármacos , Angiotensina II/antagonistas & inibidores , Angiotensina II/fisiologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/fisiopatologia , Quimioterapia Combinada , Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Glomerulosclerose Segmentar e Focal/fisiopatologia , Humanos , Hipertensão Renal/tratamento farmacológico , Hipertensão Renal/fisiopatologia , Modelos Biológicos , Proteinúria/tratamento farmacológico , Proteinúria/fisiopatologia , Indução de Remissão , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia , Sistema Renina-Angiotensina/fisiologia
18.
Am J Pathol ; 169(6): 1965-75, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17148661

RESUMO

Shigatoxin (Stx) is the offending agent of post-diarrheal hemolytic uremic syndrome, characterized by glomerular ischemic changes preceding microvascular thrombosis. Because podocytes are highly sensitive to Stx cytotoxicity and represent a source of vasoactive molecules, we studied whether Stx-2 modulated the production of endothelin-1 (ET-1), taken as candidate mediator of podocyte dysfunction. Stx-2 enhanced ET-1 mRNA and protein expression via activation of nuclear factor kappaB (NF-kappaB) and activator protein-1 (Ap-1) to the extent that transfection with the dominant-negative mutant of IkappaB-kinase 2 or with Ap-1 decoy oligodeoxynucleotides reduced ET-1 mRNA levels. We propose a role for p38 and p42/44 mitogen-activated protein kinases (MAPKs) in mediating NF-kappaB-dependent gene transcription induced by Stx-2, based on data that Stx-2 phosphorylated p38 and p42/44 MAPKs and that MAPK inhibitors reduced transcription of NF-kappaB promoter/luciferase reporter gene construct induced by Stx-2. Stx-2 caused F-actin redistribution and intercellular gaps via production of ET-1 acting on ETA receptor, because cytoskeleton changes were prevented by ETA receptor blockade. Exogenous ET-1 induced cytoskeleton rearrangement and intercellular gaps via phosphatidylinositol-3 kinase and Rho-kinase pathway and increased protein permeability across the podocyte monolayer. These data suggest that the podocyte is a target of Stx, a novel stimulus for the synthesis of ET-1, which may control cytoskeleton remodeling and glomerular permeability in an autocrine fashion.


Assuntos
Actinas/metabolismo , Endotelina-1/metabolismo , Podócitos/efeitos dos fármacos , RNA Mensageiro/metabolismo , Toxina Shiga II/farmacologia , Animais , Antígenos Glicosídicos Associados a Tumores/metabolismo , Diferenciação Celular , Permeabilidade da Membrana Celular , Células Cultivadas , Citoesqueleto/metabolismo , Endotelina-1/biossíntese , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/fisiologia , Transdução de Sinais , Fator de Transcrição AP-1/fisiologia , Transcrição Gênica , Regulação para Cima
19.
Am J Pathol ; 168(1): 42-54, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16400008

RESUMO

Changes in podocyte number or density have been suggested to play an important role in renal disease progression. Here, we investigated the temporal relationship between glomerular podocyte number and development of proteinuria and glomerulosclerosis in the male Munich Wistar Fromter (MWF) rat. We also assessed whether changes in podocyte number affect podocyte function and focused specifically on the slit diaphragm-associated protein nephrin. Age-matched Wistar rats were used as controls. Estimation of podocyte number per glomerulus was determined by digital morphometry of WT1-positive cells. MWF rats developed moderate hypertension, massive proteinuria, and glomerulosclerosis with age. Glomerular hypertrophy was already observed at 10 weeks of age and progressively increased thereafter. By contrast, mean podocyte number per glomerulus was lower than normal in young animals and further decreased with time. As a consequence, the capillary tuft volume per podocyte was more than threefold increased in older rats. Electron microscopy showed important changes in podocyte structure of MWF rats, with expansion of podocyte bodies surrounding glomerular filtration membrane. Glomerular nephrin expression was markedly altered in MWF rats and inversely correlated with both podocyte loss and proteinuria. Our findings suggest that reduction in podocyte number is an important determinant of podocyte dysfunction and progressive impairment of the glomerular permselectivity that lead to the development of massive proteinuria and ultimately to renal scarring.


Assuntos
Glomerulosclerose Segmentar e Focal/fisiopatologia , Proteínas de Membrana/metabolismo , Podócitos/metabolismo , Podócitos/ultraestrutura , Animais , Western Blotting , Modelos Animais de Doenças , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Hipertensão/etiologia , Imuno-Histoquímica , Masculino , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Proteinúria/etiologia , RNA Mensageiro/análise , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Am J Pathol ; 168(4): 1073-85, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16565484

RESUMO

Ameliorating the function of the glomerular barrier to circulating proteins by blocking angiotensin II (Ang II) translates into less risk of progression toward end-stage renal failure in diabetic and nondiabetic nephropathies. However, the mechanisms underlying this barrier protection are not clear. Specialized contacts between adjacent podocytes are major candidate targets, and the actin cytoskeleton is emerging as a regulatory element. Here, we present data demonstrating that Ang II induced reorganization of F-actin fibers and redistribution of zonula occludens-1 (ZO-1) that is physically associated with actin in murine podocytes. These effects were paralleled by increased albumin permeability across podocyte monolayers. The F-actin stabilizer jasplakinolide prevented both ZO-1 redistribution and albumin leakage, suggesting that actin cytoskeleton rearrangement is instrumental to podocyte permselective dysfunction induced by Ang II. Changes in both F-actin and ZO-1 patterns were confirmed in glomeruli of rat isolated perfused kidneys on short infusion of Ang II, leading to increased protein excretion. Podocyte dysfunction was mediated by Ang II type 1 receptor and was partly dependent on Src kinase-phospholipase C activation. These data demonstrate that strategies aimed at stabilizing podocyte-podocyte contacts and targeting the relevant intracellular signal transduction are crucial to renoprotection.


Assuntos
Angiotensina II/fisiologia , Junções Intercelulares/fisiologia , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Podócitos/fisiologia , Actinas/fisiologia , Angiotensina II/farmacologia , Animais , Diferenciação Celular , Células Cultivadas , Citoesqueleto/fisiologia , Depsipeptídeos/farmacologia , Ativação Enzimática , Técnicas In Vitro , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Camundongos , Permeabilidade , Podócitos/metabolismo , Ratos , Receptor Tipo 1 de Angiotensina/fisiologia , Soroalbumina Bovina/metabolismo , Transdução de Sinais , Fosfolipases Tipo C/metabolismo , Proteína da Zônula de Oclusão-1 , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA