Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
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
2.
Am J Physiol Renal Physiol ; 304(6): F808-19, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23136004

RESUMO

Bardoxolone methyl is an antioxidant inflammation modulator acting through induction of Keap1-Nrf2 pathway. Results from a recent phase IIb clinical trial reported that bardoxolone methyl was associated with improvement in the estimated glomerular filtration rate in patients with advanced chronic kidney disease and Type 2 diabetes. However, increases in albuminuria, serum transaminase, and frequency of adverse events were noted. We studied the effect of 3-mo treatment with RTA 405, a synthetic triterpenoid analog of bardoxolone methyl in Zucker diabetic fatty rats with overt Type 2 diabetes. Rats were treated from 3 mo of age with vehicle, RTA 405, ramipril, or RTA 405 plus ramipril. RTA 405 caused severe changes in food intake and diuresis with decline in body weight, worsening of dyslipidemia, and increase in blood pressure. Early elevation in serum transaminase was followed by liver injury. RTA 405 worsened proteinuria, glomerulosclerosis, and tubular damage. Ramipril was renoprotective, but when given with RTA 405 it was not able to limit its worsening effects. These data could be due to degradation products in the drug substance used, as disclosed by the company once the study was concluded. To overcome such a drawback, the company offered to test dh404, a variant of RTA 405, in Zucker diabetic fatty rats. The dh404 did not display beneficial effects on proteinuria, glomerulosclerosis, and interstitial inflammation. Rather, kidneys from three rats receiving dh404 showed the presence of a granulomatous and inflammatory process reminiscent of a pseudotumor. Altogether these data raise serious concerns on the use of bardoxolone analogs in Type 2 diabetic nephropathy.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Rim/efeitos dos fármacos , Ácido Oleanólico/análogos & derivados , Triterpenos/efeitos adversos , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Cromatografia Líquida , Nefropatias Diabéticas/patologia , Modelos Animais de Doenças , Diurese/efeitos dos fármacos , Ingestão de Líquidos/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Rim/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Espectrometria de Massas , Ácido Oleanólico/efeitos adversos , Ramipril/uso terapêutico , Ratos , Ratos Zucker , Circulação Renal/efeitos dos fármacos
3.
Am J Physiol Renal Physiol ; 303(9): F1370-81, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22952284

RESUMO

We previously reported that in a model of spontaneously progressive glomerular injury with early podocyte loss, abnormal migration, and proliferation of glomerular parietal epithelial progenitor cells contributed to the formation of synechiae and crescentic lesions. Here we first investigated whether a similar sequence of events could be extended to rats with adriamycin (ADR)-induced nephropathy. As a second aim, the regenerative potential of therapy with bone marrow-derived mesenchymal stem cells (MSCs) on glomerular resident cells was evaluated. In ADR-treated rats, decrease of WT1(+) podocyte number due to apoptosis was associated with reduced glomerular expression of nephrin and CD2AP. As a consequence of podocyte injury, glomerular adhesions of the capillary tuft to the Bowman's capsule were observed, followed by crescent-like lesions and glomerulosclerosis. Cellular components of synechiae were either NCAM(+) parietal progenitor cells or nestin(+) podocytes. In ADR rats, repeated injections of MSCs limited podocyte loss and apoptosis and partially preserved nephrin and CD2AP. MSCs attenuated the formation of glomerular podocyte-parietal epithelial cell bridges and normalized the distribution of NCAM(+) progenitor cells along the Bowman's capsule, thereby reducing glomerulosclerosis. Finding that MSCs increased glomerular VEGF expression and limited microvascular rarefaction may explain the prosurvival effect by stem cell therapy. MSCs also displayed anti-inflammatory activity. Coculture of MSCs with ADR-damaged podocytes showed a functional role of stem cell-derived VEGF on prosurvival pathways. These data suggest that MSCs by virtue of their tropism for damaged kidney and ability to provide a local prosurvival environment may represent a useful strategy to preserve podocyte viability and reduce glomerular inflammation and sclerosis.


Assuntos
Nefropatias/patologia , Nefropatias/fisiopatologia , Rim/fisiologia , Transplante de Células-Tronco Mesenquimais , Podócitos/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/fisiologia , Contagem de Células , Movimento Celular/fisiologia , Técnicas de Cocultura , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Doxorrubicina/efeitos adversos , Rim/patologia , Nefropatias/induzido quimicamente , Masculino , Proteínas de Membrana/metabolismo , Podócitos/patologia , Ratos , Ratos Endogâmicos Lew , Células-Tronco/patologia , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
N Engl J Med ; 361(4): 345-57, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19625716

RESUMO

BACKGROUND: The hemolytic-uremic syndrome consists of the triad of microangiopathic hemolytic anemia, thrombocytopenia, and renal failure. The common form of the syndrome is triggered by infection with Shiga toxin-producing bacteria and has a favorable outcome. The less common form of the syndrome, called atypical hemolytic-uremic syndrome, accounts for about 10% of cases, and patients with this form of the syndrome have a poor prognosis. Approximately half of the patients with atypical hemolytic-uremic syndrome have mutations in genes that regulate the complement system. Genetic factors in the remaining cases are unknown. We studied the role of thrombomodulin, an endothelial glycoprotein with anticoagulant, antiinflammatory, and cytoprotective properties, in atypical hemolytic-uremic syndrome. METHODS: We sequenced the entire thrombomodulin gene (THBD) in 152 patients with atypical hemolytic-uremic syndrome and in 380 controls. Using purified proteins and cell-expression systems, we investigated whether thrombomodulin regulates the complement system, and we characterized the mechanisms. We evaluated the effects of thrombomodulin missense mutations associated with atypical hemolytic-uremic syndrome on complement activation by expressing thrombomodulin variants in cultured cells. RESULTS: Of 152 patients with atypical hemolytic-uremic syndrome, 7 unrelated patients had six different heterozygous missense THBD mutations. In vitro, thrombomodulin binds to C3b and factor H (CFH) and negatively regulates complement by accelerating factor I-mediated inactivation of C3b in the presence of cofactors, CFH or C4b binding protein. By promoting activation of the plasma procarboxypeptidase B, thrombomodulin also accelerates the inactivation of anaphylatoxins C3a and C5a. Cultured cells expressing thrombomodulin variants associated with atypical hemolytic-uremic syndrome had diminished capacity to inactivate C3b and to activate procarboxypeptidase B and were thus less protected from activated complement. CONCLUSIONS: Mutations that impair the function of thrombomodulin occur in about 5% of patients with atypical hemolytic-uremic syndrome.


Assuntos
Ativação do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação de Sentido Incorreto , Trombomodulina/genética , Adolescente , Adulto , Criança , Complemento C3b , Fator I do Complemento , Via Alternativa do Complemento/fisiologia , Análise Mutacional de DNA , Síndrome Hemolítico-Urêmica/imunologia , Heterozigoto , Humanos , Pessoa de Meia-Idade , Linhagem , Polimorfismo de Nucleotídeo Único , Trombomodulina/metabolismo , Adulto Jovem
5.
Am J Physiol Renal Physiol ; 300(6): F1291-300, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21454255

RESUMO

Despite the central role of tubular plasma proteins that characterize progressive kidney diseases, protein concentrations along the nephron in pathological conditions have not been quantified so far. We combined experimental techniques and theoretical analysis to estimate glomerular and tubular levels of albumin in the experimental model of 5/6 nephrectomy (Nx) in the rat, with or without angiotensin-converting enzyme (ACE) inhibition. We measured glomerular permselectivity by clearance of fluorescent Ficoll and albumin and used theoretical analysis to estimate tubular albumin. As expected, 5/6 Nx induced an elevation of the fractional clearance of the largest Ficoll molecules (radii >56 Å, P < 0.05), increasing the importance of the shunt pathway of the glomerular membrane and the albumin excretion rate (119 ± 41 vs. 0.6 ± 0.2 mg/24 h, P < 0.01). ACE inhibition normalized glomerular permselectivity and urinary albumin (0.5 ± 0.3 mg/24 h). Theoretical analysis indicates that with 5/6 Nx, an increased albumin filtration overcomes proximal tubule reabsorption, with a massive increase in average albumin concentration along the tubule, reaching the highest value of >2,500 µg/ml at the end of the collecting duct. ACE inhibition improved glomerular permselectivity, limiting albumin filtration under proximal tubule reabsorption capacity, with low albumin concentration along the entire nephron, averaging <13 µg/ml at the end of the collecting duct. These results reinforce our understanding of the mechanisms of renal disease progression and the effects of angiotensin II antagonism. They also suggest that evaluation of tubular protein concentration levels could help to identify patients at risk of kidney disease progression and to improve clinical management.


Assuntos
Albuminas/metabolismo , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Nefropatias/metabolismo , Glomérulos Renais/efeitos dos fármacos , Albuminúria/metabolismo , Albuminúria/patologia , Análise de Variância , Animais , Taxa de Filtração Glomerular/efeitos dos fármacos , Nefropatias/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Nefrectomia , Proteinúria/metabolismo , Proteinúria/patologia , Ratos , Ratos Sprague-Dawley
6.
Am J Physiol Renal Physiol ; 301(5): F1114-23, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21816757

RESUMO

Diabetic nephropathy is associated with cardiovascular morbidity. Angiotensin-converting enzyme (ACE) inhibitors provide imperfect renoprotection in advanced type 2 diabetes, and cardiovascular risk remains elevated. Endothelin (ET)-1 has a role in renal and cardiac dysfunction in diabetes. Here, we assessed whether combination therapy with an ACE inhibitor and ET(A) receptor antagonist provided reno- and cardioprotection in rats with overt type 2 diabetes. Four groups of Zucker diabetic fatty (ZDF) rats were treated orally from 4 (when proteinuric) to 8 mo with vehicle, ramipril (1 mg/kg), sitaxsentan (60 mg/kg), and ramipril plus sitaxsentan. Lean rats served as controls. Combined therapy ameliorated proteinuria and glomerulosclerosis mostly as a result of the action of ramipril. Simultaneous blockade of ANG II and ET-1 pathways normalized renal monocyte chemoattractant protein-1 and interstitial inflammation. Cardiomyocyte loss, volume enlargement, and capillary rarefaction were prominent abnormalities of ZDF myocardium. Myocyte volume was reduced by ramipril and sitaxsentan, which also ameliorated heart capillary density. Drug combination restored myocardial structure and reestablished an adequate capillary network in the presence of increased cardiac expression of VEGF/VEGFR-1, and significant reduction of oxidative stress. In conclusion, in type 2 diabetes concomitant blockade of ANG II synthesis and ET-1 biological activity through an ET(A) receptor antagonist led to substantial albeit not complete renoprotection, almost due to the ACE inhibitor. The drug combination also showed cardioprotective properties, which however, were mainly dependent on the contribution of the ET(A) receptor antagonist through the action of VEGF.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Diabetes Mellitus Tipo 2/patologia , Antagonistas do Receptor de Endotelina A , Coração/efeitos dos fármacos , Rim/patologia , Miocárdio/patologia , Animais , Glicemia/metabolismo , Peso Corporal/fisiologia , Contagem de Células , Colágeno Tipo III/metabolismo , Diabetes Mellitus Tipo 2/genética , Endotelina-1/metabolismo , Hemodinâmica/efeitos dos fármacos , Imuno-Histoquímica , Rim/efeitos dos fármacos , Rim/metabolismo , Testes de Função Renal , Masculino , Células Musculares/efeitos dos fármacos , Miocárdio/metabolismo , Ratos , Ratos Zucker , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Sobrevida , Tirosina/análogos & derivados , Tirosina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Stem Cells ; 28(3): 513-22, 2010 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-20049901

RESUMO

In search for new sources of mesenchymal stem cells (MSCs) for renal repair in acute kidney injury (AKI), we investigated the potential of human cord blood (CB)-MSCs to cure mice with AKI. Infusion of CB-MSCs in immunodeficient mice with cisplatin-induced AKI ameliorated both renal function and tubular cell injury, and prolonged survival. Transplanted CB-MSCs localized in peritubular areas, limited capillary alterations and neutrophil infiltration. Apoptosis reduced and tubular cell proliferation increased by virtue of stem cell capacity to produce growth factors. The reno-protective effect of CB-MSCs was further confirmed by their ability to inhibit oxidative damage and to induce the prosurvival factor Akt in tubular cells. The evidence that CB-MSCs in vitro increased the production of growth factors and inhibited IL-1 beta and TNFalpha synthesis when cocultured with damaged proximal tubular cells indicates a regenerative and anti-inflammatory action of stem cell treatment. Altogether these results highlight the potential of human CB-MSCs as future cell therapy for testing in human AKI.


Assuntos
Diferenciação Celular/fisiologia , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Sobrevivência de Enxerto/fisiologia , Nefropatias/cirurgia , Rim/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Doença Aguda , Animais , Anti-Inflamatórios/metabolismo , Apoptose/fisiologia , Técnicas de Cultura de Células , Proliferação de Células , Técnicas de Cocultura , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/patologia , Rim/fisiopatologia , Nefropatias/fisiopatologia , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos SCID , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Am J Physiol Renal Physiol ; 299(5): F1203-11, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20719975

RESUMO

The capacity of renin-angiotensin system (RAS) inhibitors to delay progression of diabetic nephropathy depends on the time at which therapy is started. A multimodal intervention is required to afford renoprotection in overt diabetic nephropathy. Here we assessed the effects of maximal RAS inhibition by angiotensin-converting enzyme (ACE) inhibitor plus angiotensin II type 1 receptor blocker (ARB) in combination with statin in rats with overt diabetic nephropathy. Uninephrectomized rats made diabetic by streptozotocin were orally treated from 4 (when proteinuria and renal lesions had developed) to 8 mo with vehicle, lisinopril plus candesartan, lisinopril plus candesartan plus rosuvastatin, or rosuvastatin alone. Systolic blood pressure increased in diabetic rats and was significantly lowered by combined therapies. Dual RAS blockade significantly reduced proteinuria compared with vehicle. Addition of statin further lowered proteinuria to control levels. Glomerulosclerosis was ameliorated by RAS inhibitors or statin, and regression was achieved by the addition of statin. Loss of podocytes of diabetic rats was limited by ACE inhibitor plus ARB while normalized by the three drugs. Defective nephrin expression of diabetes was increased by dual RAS blockade or statin and restored by the triple therapy. Tubular damage, interstitial inflammation, and expression of the fibrotic markers transforming growth factor (TGF)-ß1 and phosphorylated Smad 2/3 in tubuli were significantly reduced by the triple regimen. These data suggest a strategy to target proteinuria to try to achieve regression of renal disease in diabetic patients who do not fully benefit from RAS inhibition alone.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Nefropatias Diabéticas/prevenção & controle , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Proteinúria/tratamento farmacológico , Animais , Pressão Sanguínea/efeitos dos fármacos , Colesterol/sangue , Ésteres do Colesterol/sangue , Nefropatias Diabéticas/urina , Fibrose , Imuno-Histoquímica , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Testes de Função Renal , Glomérulos Renais/patologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Proteínas de Membrana/biossíntese , Nefrite Intersticial/patologia , Podócitos/patologia , Proteinúria/etiologia , Ratos , Ratos Sprague-Dawley , Sistema Renina-Angiotensina/efeitos dos fármacos
9.
J Immunol ; 181(2): 1460-9, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18606701

RESUMO

Shiga toxins (Stx) are the virulence factors of enterohemorrhagic Escherichia coli O157:H7, a worldwide emerging diarrheal pathogen, which precipitates postdiarrheal hemolytic uremic syndrome, the leading cause of acute renal failure in children. In this study, we show that Stx2 triggered expression of fractalkine (FKN), a CX3C transmembrane chemokine, acting as both adhesion counterreceptor on endothelial cells and soluble chemoattractant. Stx2 caused in HUVEC expression of FKN mRNA and protein, which promoted leukocyte capture, ablated by Abs to either endothelial FKN or leukocyte CX3CR1 receptor. Exposure of human glomerular endothelial cells to Stx2 recapitulated its FKN-inducing activity and FKN-mediated leukocyte adhesion. Both processes required phosphorylation of Src-family protein tyrosine kinase and p38 MAPK in endothelial cells. Furthermore, they depended on nuclear import of NF-kappaB and other stress-responsive transcription factors. Inhibition of their nuclear import with the cell-penetrating SN50 peptide reduced FKN mRNA levels and FKN-mediated leukocyte capture by endothelial cells. Adenoviral overexpression of IkappaBalpha inhibited FKN mRNA up-regulation. The FKN-mediated responses to Stx2 were also dependent on AP-1. In mice, both virulence factors of Stx-producing E. coli, Stx and LPS, are required to elicit hemolytic uremic syndrome. In this study, FKN was detected within glomeruli of C57BL/6 mice injected with Stx2, and further increased after Stx2 plus LPS coadministration. This was associated with recruitment of CX3CR1-positive cells. Thus, in response to Stx2, FKN is induced playing an essential role in the promotion of leukocyte-endothelial cell interaction thereby potentially contributing to the renal microvascular dysfunction and thrombotic microangiopathy that underlie hemolytic uremic syndrome due to enterohemorrhagic E. coli O157:H7 infection.


Assuntos
Quimiocina CX3CL1/metabolismo , Células Endoteliais/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Glomérulos Renais/imunologia , Leucócitos/imunologia , Receptores de Quimiocinas/metabolismo , Toxina Shiga II/imunologia , Animais , Receptor 1 de Quimiocina CX3C , Adesão Celular , Células Cultivadas , Quimiocina CX3CL1/imunologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Escherichia coli O157/imunologia , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/citologia , Glomérulos Renais/metabolismo , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/imunologia , NF-kappa B/metabolismo , Receptores de Quimiocinas/imunologia , Toxina Shiga II/metabolismo , Transdução de Sinais , Fator de Transcrição AP-1/imunologia , Fator de Transcrição AP-1/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Pediatr Nephrol ; 25(11): 2231-40, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20424866

RESUMO

Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli O157:H7 has become a global threat to public health, as a primary cause of a worldwide spread of hemorrhagic colitis complicated by diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure that mainly affects early childhood. Endothelial dysfunction has been recognized as the trigger event in the development of microangiopathic processes. Endothelial cells, mainly those located in the renal microvasculature, are primary targets of the toxic effects of Stx1 and 2. Stxs bound to their specific globotriaosylceramide (Gb3Cer) receptor on the cell surface trigger a cascade of signaling events, involving NF-κB activation, that induce expression of genes encoding for adhesion molecules and chemokines, and culminate in the adhesion of leukocytes to endothelial cells, thereby increasing the endothelial susceptibility to leukocyte-mediated injury. Activated endothelial cells in response to Stxs lose the normal thromboresistance phenotype and become thrombogenic, initiating microvascular thrombus formation. Evidence is emerging that complement activation in response to Stxs favors platelet thrombus formation on endothelial cells, which may play a role in amplifying the inflammation-thrombosis circuit in Stx-associated HUS.


Assuntos
Endotélio Vascular/fisiopatologia , Síndrome Hemolítico-Urêmica/metabolismo , Toxina Shiga/metabolismo , Injúria Renal Aguda/metabolismo , Capilares/metabolismo , Quimiocinas/metabolismo , Pré-Escolar , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Escherichia coli O157/metabolismo , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/fisiopatologia , Humanos , Leucócitos/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Toxina Shiga/farmacologia , Triexosilceramidas/metabolismo
11.
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
12.
Am J Physiol Renal Physiol ; 297(5): F1448-56, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19675181

RESUMO

In the present study, we evaluated the effect of simultaneously blocking angiotensin II synthesis and endothelin (ET)-1 activity as a multimodal intervention to implement renoprotection in overt diabetic nephropathy. Mechanisms underlying combined therapy effectiveness were addressed by investigating podocyte structure and function and glomerular barrier size-selective properties. Uninephrectomized rats made diabetic by streptozotocin received orally placebo, lisinopril (12.5 mg/l), the ET(A) receptor antagonist avosentan (30 mg/kg), or their combination from 4 (when animals had proteinuria) to 8 mo. Proteinuria, renal damage, podocyte number, nephrin expression, and glomerular size selectivity by graded-size Ficoll molecule fractional clearance were assessed. Combined therapy normalized proteinuria, provided complete protection from tubulointerstitial damage, and induced regression of glomerular lesions, while only a partial renoprotection was achieved by each drug alone. Lisinopril plus avosentan restored to normal values the number of podocytes. Single therapies only limited podocyte depletion. Defective nephrin expression of diabetes was prevented by each drug. Altered glomerular size selectivity to large macromolecules of diabetic rats was remarkably improved by lisinopril and the combined treatment. Avosentan ameliorated peritubular capillary architecture and reduced interstitial inflammation and fibrosis. The ACE inhibitor and ET(A) receptor antagonist induced regression of glomerular lesions in overt diabetic nephropathy. Regression of renal disease was conceivably the result of the synergistic effect of the ACE inhibitor of preserving glomerular permselective properties and the ET(A) antagonist in improving tubulointerstitial changes. These findings provide mechanistic insights to explain the antiproteinuric effect of this combined therapy in diabetes.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Nefropatias Diabéticas/prevenção & controle , Lisinopril/uso terapêutico , Piridinas/uso terapêutico , Pirimidinas/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Capilares/patologia , Contagem de Células , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/mortalidade , Nefropatias Diabéticas/patologia , Quimioterapia Combinada , Antagonistas do Receptor de Endotelina A , Imuno-Histoquímica , Rim/patologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/patologia , Lipídeos/sangue , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/genética , Podócitos/efeitos dos fármacos , Podócitos/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Kidney Int ; 76(9): 960-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19625993

RESUMO

Blockade of the renin-angiotensin system (RAS), the standard treatment for chronic proteinuric nephropathy, slows but may not halt progression of the disease, particularly when therapy is started late. Because vasopressin may also play a role in the progression of renal disease, we measured the effect of a dual V(1a) and V(2) vasopressin receptor antagonist (RWJ-676070) alone or combined with angiotensin-converting enzyme inhibition or angiotensin II type 1 receptor blockade on proteinuria and renal disease progression during overt nephropathy. Twenty-one days after renal mass reduction, a time of established injury, rats were given vehicle, RWJ-676070, enalapril, losartan, RWJ-676070 plus enalapril, or losartan in drinking water for an additional 39 days. RWJ-676070 returned the blood pressure to pre-treatment levels, which were significantly lower than those in vehicle-treated rats. Enalapril, losartan, and the combined therapies reduced blood pressure to a greater extent. RWJ-676070 afforded a partial antiproteinuric effect, which was enhanced by the addition of enalapril or losartan. Renal functional impairment, and glomerular and tubular changes were partially ameliorated by RWJ-676070; parameters significantly improved with either enalapril or losartan alone and improved to a greater extent with the combined therapies. Our findings suggest that vasopressin receptor antagonists could be of additional therapeutic value in the treatment of chronic proteinuric nephropathy.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Antagonistas dos Receptores de Hormônios Antidiuréticos , Benzazepinas/farmacologia , Enalapril/farmacologia , Antagonistas de Hormônios/farmacologia , Nefropatias/tratamento farmacológico , Rim/efeitos dos fármacos , Losartan/farmacologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Compostos de Espiro/farmacologia , Animais , Benzazepinas/sangue , Benzazepinas/farmacocinética , Biomarcadores/sangue , Biomarcadores/urina , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Doença Crônica , Modelos Animais de Doenças , Progressão da Doença , Diurese/efeitos dos fármacos , Ingestão de Líquidos/efeitos dos fármacos , Quimioterapia Combinada , Ingestão de Alimentos/efeitos dos fármacos , Antagonistas de Hormônios/sangue , Antagonistas de Hormônios/farmacocinética , Rim/metabolismo , Rim/patologia , Rim/fisiopatologia , Nefropatias/metabolismo , Nefropatias/patologia , Nefropatias/fisiopatologia , Masculino , Nefrectomia , Proteinúria/tratamento farmacológico , Proteinúria/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Vasopressinas/metabolismo , Compostos de Espiro/sangue , Compostos de Espiro/farmacocinética , Fatores de Tempo
14.
Kidney Int ; 75(10): 1050-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19242507

RESUMO

Intrarenal complement activation plays an important role in the progression of chronic kidney disease. A key target of the activated complement cascade is the proximal tubule, a site where abnormally filtered plasma proteins and complement factors combine to promote injury. This study determined whether protein overloading of human proximal tubular cells (HK-2) in culture enhances complement activation by impairing complement regulation. Addition of albumin or transferrin to the cells incubated with diluted human serum as a source of complement caused increased apical C3 deposition. Soluble complement receptor-1 (an inhibitor of all 3 activation pathways) blocked complement deposition while the classical and lectin pathway inhibitor, magnesium chloride-EGTA, was, ineffective. Media containing albumin as well as complement had additive proinflammatory effects as shown by increased fractalkine and transforming growth factor-beta mRNA expression. This paralleled active C3 and C5b-9 generations, effects not shared by transferrin. Factor H, one of the main natural inhibitors of the alternative pathway, binds to heparan sulfate proteoglycans. Both the density of heparan sulfate and factor H binding were reduced with protein loading, thereby enhancing the albumin- and serum-dependent complement activation potential. Thus, protein overload reduces the ability of the tubule cell to bind factor H and counteract complement activation, effects instrumental to renal disease progression.


Assuntos
Fator H do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Túbulos Renais Proximais/patologia , Proteínas/farmacologia , Linhagem Celular , Ativação do Complemento , Progressão da Doença , Humanos , Nefropatias/etiologia , Nefropatias/patologia , Ligação Proteica , Albumina Sérica/farmacologia , Transferrina/farmacologia
15.
Stem Cells ; 26(8): 2075-82, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18499895

RESUMO

Transplantation of bone marrow mesenchymal stem cells (BM-MSC) or stromal cells from rodents has been identified as a strategy for renal repair in experimental models of acute kidney injury (AKI), a highly life-threatening clinical setting. The therapeutic potential of BM-MSC of human origin has not been reported so far. Here, we investigated whether human BM-MSC treatment could prevent AKI induced by cisplatin and prolong survival in an immunodeficient mouse model. Results showed that human BM-MSC infusion decreased proximal tubular epithelial cell injury and ameliorated the deficit in renal function, resulting in reduced recipient mortality. Infused BM-MSC became localized predominantly in peritubular areas and acted to reduce renal cell apoptosis and to increase proliferation. BM-MSC also induced protection against AKI-related peritubular capillary changes consisting of endothelial cell abnormalities, leukocyte infiltration, and low endothelial cell and lumen volume density as assessed by morphometric analysis. These findings indicate that human MSC of bone marrow origin hold potential to prolong survival in AKI and should be considered for testing in a clinical trial. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células da Medula Óssea/citologia , Rim/lesões , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Animais , Apoptose , Peso Corporal , Diferenciação Celular , Proliferação de Células , Cisplatino/farmacologia , Feminino , Humanos , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos SCID , Fenótipo
16.
J Am Soc Nephrol ; 19(6): 1158-67, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18354030

RESUMO

Intrarenal complement activation leads to chronic tubulointerstitial injury in animal models of proteinuric nephropathies, making this process a potential target for therapy. This study investigated whether a C3-mediated pathway promotes renal injury in the protein overload model and whether the abnormal exposure of proximal tubular cells to filtered complement could trigger the resulting inflammatory response. Mice with C3 deficiency were protected to a significant degree against the protein overload-induced interstitial inflammatory response and tissue damage, and they had less severe podocyte injury and less proteinuria. When the same injury was induced in wild-type (WT) mice, antiproteinuric treatment with the angiotensin-converting enzyme inhibitor lisinopril reduced the amount of plasma protein filtered, decreased the accumulation of C3 by proximal tubular cells, and protected against interstitial inflammation and damage. For determination of the injurious role of plasma-derived C3, as opposed to tubular cell-derived C3, C3-deficient kidneys were transplanted into WT mice. Protein overload led to the development of glomerular injury, accumulation of C3 in podocytes and proximal tubules, and tubulointerstitial changes. Conversely, when WT kidneys were transplanted into C3-deficient mice, protein overload led to a more mild disease and abnormal C3 deposition was not observed. These data suggest that the presence of C3 increases the glomerular filtration barrier's susceptibility to injury, ultrafiltered C3 contributes more to tubulointerstitial damage induced by protein overload than locally synthesized C3, and local C3 synthesis is irrelevant to the development of proteinuria. It is speculated that therapies targeting complement combined with interventions to minimize proteinuria would more effectively prevent the progression of renal disease.


Assuntos
Complemento C3/deficiência , Insuficiência Renal/etiologia , Animais , Complemento C3/fisiologia , Progressão da Doença , Taxa de Filtração Glomerular , Masculino , Camundongos
17.
J Am Soc Nephrol ; 18(11): 2921-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17942965

RESUMO

In mice with cisplatin-induced acute kidney injury, administration of bone marrow-derived mesenchymal stem cells (MSC) restores renal tubular structure and improves renal function, but the underlying mechanism is unclear. Here, we examined the process of kidney cell repair in co-culture experiments with MSC and cisplatin-injured proximal tubular epithelial cells (PTEC). Exposure of PTEC to cisplatin markedly reduced cell viability at 4 days, but co-culture with MSC provided a protective effect by promoting tubular cell proliferation. This effect was mediated by insulin-like growth factor-1 (IGF-1), highly expressed by MSC as mRNA and protein, since blocking the growth factor's function with a specific antibody attenuated cell proliferation of PTEC. Confirming this, knocking down IGF-1 expression in MSC by small interfering-RNA also resulted in a significant decrease in PTEC proliferation and increased apoptosis. Furthermore, in the murine model of cisplatin-induced kidney injury, administering IGF-1 gene-silenced MSC limited their protective effect on renal function and tubular structure. These findings indicate that MSC exert beneficial effects on tubular cell repair in acute kidney injury by producing the mitogenic and pro-survival factor IGF-1.


Assuntos
Células Epiteliais/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Nefropatias/terapia , Túbulos Renais Proximais/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Animais , Técnicas de Cultura de Células , Proliferação de Células , Sobrevivência Celular/fisiologia , Cisplatino , Técnicas de Cocultura , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
18.
Am J Nephrol ; 27(6): 630-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17851231

RESUMO

BACKGROUND: Evidence is accumulating that statins can reduce proteinuria and disease progression in chronic kidney disease. However, some safety concerns have been recently raised on the use of these agents, mainly due to transient episodes of proteinuria observed in patients receiving high doses of rosuvastatin. METHODS: We investigated in rats with renal mass ablation (RMR) whether rosuvastatin (5 or 20 mg/day) worsens proteinuria as compared to untreated RMR animals. Moreover, we also examined whether rosuvastatin-induced changes in proteinuria would be due to the effect of the drug on permselective properties of glomerular capillary barrier, measured by the fractional clearance of graded-size Ficoll molecules and/or by proximal tubular mechanisms, by assessing urinary excretion of beta(2)-microglobulin. RESULTS: RMR rats given rosuvastatin for 28 days showed a progressive increase in proteinuria, with values numerically but not significantly higher than those in RMR animals given the vehicle. In RMR rats, rosuvastatin did not significantly affect the fractional clearance of Ficoll as compared to vehicle-treated RMR animals. A significant correlation was found between urinary protein and beta(2)-microglobulin excretion in rats treated with rosuvastatin (r = 0.936, p < 0.001), but not in those given vehicle. Renal function, glomerular and tubulointerstitial injury were comparable in rosuvastatin-treated and untreated RMR rats at the end of the 28-day follow-up. CONCLUSION: In rats with RMR, rosuvastatin mildly enhances urinary protein excretion rate. This, however, was not the result of further changes in the size-permselective function of glomerular capillary barrier.


Assuntos
Capilares/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Fluorbenzenos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Glomérulos Renais/irrigação sanguínea , Pirimidinas/farmacologia , Sulfonamidas/farmacologia , Animais , Capilares/fisiopatologia , Modelos Animais de Doenças , Glomérulos Renais/efeitos dos fármacos , Proteinúria/induzido quimicamente , Proteinúria/fisiopatologia , Ratos , Ratos Sprague-Dawley , Rosuvastatina Cálcica
19.
Nephron Exp Nephrol ; 104(4): e158-68, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16902320

RESUMO

BACKGROUND: In diabetic rats with maximal activation of RAS induced by uninephrectomy, late treatment with anti-TGFbeta antibody limited renal injury only when combined with ACE inhibitor. We investigated whether in a two-kidney diabetic model the time at which treatment started predicted the response to TGFbeta antagonist. METHODS: 27 weeks after streptozotocin injection, animals had mild proteinuria and were randomized to receive irrelevant antibody, anti-TGFbeta antibody (1D11) or enalapril till 52 weeks (early treatment). The effect of agents alone or combined was also evaluated at the time of overt proteinuria (late treatment, 52-61 weeks). RESULTS: When given early, 1D11 displayed marked antihypertensive and antiproteinuric effects. Glomerulosclerosis was reduced to the extent that a remarkable percentage of glomeruli without sclerosis appeared after treatment. Podocyte number was normalized. Renoprotection of 1D11 was comparable to enalapril. Despite control of blood pressure, in late treatment single agents did not reduce proteinuria significantly. Glomerulosclerosis and podocyte loss were partially limited by 1D11 or enalapril, but full protection was achieved by combination. CONCLUSIONS: Renoprotective effect of TGFbeta antagonism crucially depends on the time at which treatment started. Effectiveness of early treatment with 1D11 would indicate that TGFbeta is a major mediator of damage in early diabetes. To tackle the renal damage in the phase of advanced disease, a combined treatment with ACE inhibitor is needed.


Assuntos
Anticorpos/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Nefropatias Diabéticas/prevenção & controle , Fator de Crescimento Transformador beta/imunologia , Albuminúria/urina , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Nefropatias Diabéticas/patologia , Enalapril/uso terapêutico , Glomérulos Renais/patologia , Masculino , Podócitos/efeitos dos fármacos , Proteinúria/urina , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
20.
Eur Cytokine Netw ; 14(4): 211-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14715412

RESUMO

TIR8, also known as single Ig IL-1R-related molecule (SIGIRR), is a member of the IL-1 receptor family. The present study was designed to investigate the expression and function of TIR8. TIR8 was mainly expressed in mouse and human epithelial tissues such as kidney, lung and gut. Resting and activated T and B lymphocytes and monocytes-macrophages expressed little or no TIR8, with the exception of the mouse GG2EE macrophage line. In the kidney, the organ with highest mRNA levels, TIR8 expression was confined to epithelial cells and, in situ, to tubular epithelium. A variety of signals failed to regulate TIR8 expression, but LPS reduced TIR8 mRNA transcripts. An NF-kB driven reporter system was used to investigate the function of TIR8. TIR8 did not activate NF-kB expression alone or in concert with IL-1R1. In contrast, TIR8 inhibited signaling from the IL-1R complex. Inhibition required the intracellular portion of TIR8 but the extracellular domain was dispensable for blocking activity. Thus, TIR8 is a unique member of the IL-1R family, with a distinct pattern of epithelial expression, including the kidney and mucosae, and an inhibitory function on IL-1 signaling.


Assuntos
Interleucina-1/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/fisiologia , Animais , Regulação para Baixo , Genes Reporter , Humanos , Inflamação/metabolismo , Rim/metabolismo , Camundongos , NF-kappa B/metabolismo , Especificidade de Órgãos , RNA Mensageiro/metabolismo , Receptores de Interleucina-1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA