Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Nat Med ; 4(5): 575-80, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9585231

RESUMO

The cyclin-dependent kinase inhibitor p27Kip1 controls cell proliferation in response to normal mitogenic stimuli. We show here that p27Kip1 also safeguards against excessive cell proliferation in specific pathophysiologic settings. We used experimental glomerulonephritis as a paradigm for immune mediated inflammation and ureteral obstruction as a model for non-immune mediated inflammation. Renal function was substantially decreased in nephritic p27-/- mice compared with control mice, and this was associated with increased glomerular cell proliferation, apoptosis and matrix protein accumulation. Tubular epithelial cell proliferation and apoptosis was also increased in p27-/- mice following ureteral obstruction. p27Kip1 may have a general role in protecting cells and tissues from inflammatory injury.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Glomerulonefrite/fisiopatologia , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Supressoras de Tumor , Obstrução Ureteral/fisiopatologia , Animais , Apoptose , Divisão Celular , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Rim/imunologia , Testes de Função Renal , Glomérulos Renais/patologia , Túbulos Renais/patologia , Camundongos , Camundongos Mutantes , Proteínas Associadas aos Microtúbulos/genética , Proteínas Serina-Treonina Quinases/metabolismo
2.
Kidney Int ; 74(1): 22-36, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18418356

RESUMO

Proteinuria is the hallmark of diabetic kidney disease (DKD) and is an independent risk factor for both renal disease progression, and cardiovascular disease. Although the characteristic pathological changes in DKD include thickening of the glomerular basement membrane and mesangial expansion, these changes per se do not readily explain how patients develop proteinuria. Recent advances in podocyte and glomerular endothelial cell biology have shifted our focus to also include these cells of the glomerular filtration barrier in the development of proteinuria in DKD. This review describes the pathophysiological mechanisms at a cellular level which explain why patients with DKD develop proteinuria.


Assuntos
Nefropatias Diabéticas/complicações , Proteinúria/etiologia , Nefropatias Diabéticas/patologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Taxa de Filtração Glomerular , Humanos , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Proteinúria/patologia
3.
J Clin Invest ; 100(10): 2512-20, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9366565

RESUMO

Glomerular injury is characterized by mesangial cell (MC) proliferation and matrix formation. We sought to determine if reducing the activity of cyclin-dependent kinase 2 (CDK2) with the purine analogue, Roscovitine, decreased MC proliferation in vitro and in vivo. Roscovitine (25 microM) inhibited FCS-induced proliferation (P < 0.0001) in cultured MC. Rats with experimental mesangial proliferative glomerulonephritis (Thy1 model) were divided into two groups. A prevention group received daily intraperitoneal injections of Roscovitine in DMSO (2.8 mg/kg) starting at day 1. A treatment group received daily Roscovitine starting at day 3, when MC proliferation was established. Control Thy1 rats received DMSO alone. MC proliferation (PCNA +/OX7 + double immunostaining) was reduced by > 50% at days 5 and 10 in the Roscovitine prevention group, and at day 5 in the treatment group (P < 0.0001). Early administration of Roscovitine reduced immunostaining for collagen type IV, laminin, and fibronectin at days 5 and 10 (r = 0.984; P < 0.001), which was associated with improved renal function (urinary protein/creatinine, blood urea nitrogen, P < 0.05). We conclude that reducing the activity of CDK2 with Roscovitine in experimental glomerulonephritis decreases cell proliferation and matrix production, resulting in improved renal function, and may be a useful therapeutic intervention in disease characterized by proliferation.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclo Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/fisiologia , Mesângio Glomerular/patologia , Glomerulonefrite Membranoproliferativa/patologia , Glomerulonefrite Membranoproliferativa/fisiopatologia , Proteínas Serina-Treonina Quinases/fisiologia , Purinas/farmacologia , Animais , Ciclo Celular/fisiologia , Núcleo Celular/ultraestrutura , Células Cultivadas , Colágeno/biossíntese , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Fibronectinas/biossíntese , Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/fisiopatologia , Testes de Função Renal , Laminina/biossíntese , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ratos , Ratos Wistar , Roscovitina
4.
J Clin Invest ; 98(2): 262-70, 1996 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8755633

RESUMO

Excessive mesangial cell (MC) proliferation is a hallmark of many glomerulopathies. In our recent study on cultured rat MC (Matousovic, K., J.P. Grande, C.C.S. Chini, E.N. Chini, and T.P. Dousa. 1995. J. Clin. Invest. 96:401-410) we found that inhibition of isozyme cyclic-3',5'-nucleotide phosphodiesterase (PDE) type III (PDE-III) suppressed MC mitogenesis by activating cAMP-dependent protein kinase (PKA) and by decreasing activity of mitogen-activated protein kinase (MAPK). We also found that inhibition of another PDE isozyme, PDE-IV, suppresses superoxide generation in glomeruli (Chini, C.C.S., E.N. Chini, J.M. Williams, K. Matousovic, and T.P. Dousa. 1994. Kidney Int. 46:28-36). We thus explored whether administration in vivo of the selective PDE-III antagonist, lixazinone (LX), together with the specific PDE-IV antagonist, rolipram (RP), can attenuate development of mesangioproliferative glomerulonephritis (MSGN) induced in rats by anti-rat thymocyte serum (ATS). Unlike the vehicle-treated MSGN rats, rats with MSGN treated with LX and RP did not develop proteinuria and maintained normal renal function when examined 5 d after injection of ATS. In PAS-stained kidneys from PDE-antagonists-treated MSGN-rats the morphology of glomeruli showed a reduction in cellularity compared with control rats with ATS. Compared with MSGN rats receiving vehicle, the MSGN rats receiving PDE-antagonists had less glomerular cell proliferation (PCNA delta -65%), a significantly lesser macrophage infiltration (delta -36% ED-1) and a significant reduction of alpha-smooth muscle actin expression by activated MC; in contrast, immunostaining for platelet antigens and laminin were not different. The beneficial effect of PDE inhibitors was not due to a moderate decrease (approximately -20%) in systolic blood pressure (SBP); as a similar decrease in SBP due to administration of hydralazine, a drug devoid of PDE inhibitory effect, did not reduce severity of MSGN in ATS-injected rats. We conclude that antagonists of PDE-III and PDE-IV administered in submicromolar concentrations in vivo to ATS-injected rats can decrease the activation and proliferation of MC, inhibit the macrophage accumulation, and prevent proteinuria in the acute phase of MSGN. We propose that PDE isozyme inhibitors act to block (negative "crosstalk") the mitogen-stimulated intracellular signaling pathway which controls MC proliferation due to activating of the cAMP-PKA pathway. These results suggest that antagonists of PDE-111 and IV may have a suppressive effect in acute phases or relapses of glomerulopathies associated with MC proliferations.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Glomerulonefrite Membranoproliferativa/prevenção & controle , Inibidores de Fosfodiesterase/farmacologia , Pirrolidinonas/farmacologia , Quinazolinas/farmacologia , Actinas/biossíntese , Animais , Pressão Sanguínea/efeitos dos fármacos , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Glomerulonefrite Membranoproliferativa/patologia , Glomerulonefrite Membranoproliferativa/fisiopatologia , Hidralazina/farmacologia , Isoenzimas/antagonistas & inibidores , Testes de Função Renal , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Laminina/análise , Masculino , Proteinúria , Ratos , Ratos Wistar , Rolipram , Superóxidos/metabolismo
5.
J Clin Invest ; 100(4): 786-94, 1997 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9259576

RESUMO

We investigated the origin of the glomerular mesangial cell, a smooth muscle-like cell that provides structural support in the glomerulus. Injection of anti-Thy 1 antibody that binds the Thy 1 antigen on rat mesangial cells eliminated (> 95%) the mesangial population at 20-28 h, while Thy 1-positive cells in the juxtaglomerular apparatus (JGA) were sequestered from the circulation and survived. Single pulse labeling with [3H]thymidine at 36 h labeled Thy 1-positive cells in the JGA and hilus. Serial biopsies demonstrated the progressive migration (5-15 micron/d) and proliferation of these mesangial reserve cells until the entire glomerulus was repopulated. The regenerating mesangial population expressed contractile and migratory proteins preferentially at the leading edge of the migratory front. Single as well as multiple pulse labeling with [3H]thymidine confirmed that the entire mesangial cell repopulation originated from only a few mesangial reserve cells. These reserve cells resided in the extraglomerular mesangium in the JGA and were not renin-secreting cells, macrophages, smooth muscle cells, or endothelial cells. These studies document mesangial cell migration in the anti-Thy 1 model of mesangial proliferative glomerulonephritis and provide evidence for a new role for the juxtaglomerular apparatus in the maintenance of the mesangial cell population.


Assuntos
Mesângio Glomerular/patologia , Glomerulonefrite/patologia , Sistema Justaglomerular/patologia , Glomérulos Renais/patologia , Animais , Antígenos de Superfície/metabolismo , Movimento Celular , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Mesângio Glomerular/metabolismo , Imuno-Histoquímica , Isoanticorpos/administração & dosagem , Sistema Justaglomerular/metabolismo , Glomérulos Renais/metabolismo , Masculino , Ratos , Ratos Wistar , Fatores de Tempo
6.
J Clin Invest ; 103(5): 597-604, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10074476

RESUMO

Proliferation and apoptosis are increased in many types of inflammatory diseases. A role for the cyclin kinase inhibitor p27(Kip1) (p27) in limiting proliferation has been shown. In this study, we show that p27(-/-) mesangial cells and fibroblasts have strikingly elevated rates of apoptosis, not proliferation, when deprived of growth factors. Apoptosis was rescued by restoration of p27 expression. Cyclin A-cyclin-dependent kinase 2 (CDK2) activity, but not cyclin E-CDK2 activity, was increased in serum-starved p27(-/-) cells, and decreasing CDK2 activity, either pharmacologically (Roscovitine) or by a dominant-negative mutant, inhibited apoptosis. Our results show that a new biological function for the CDK inhibitor p27 is protection of cells from apoptosis by constraining CDK2 activity. These results suggest that CDK inhibitors are necessary for coordinating the cell cycle and cell-death programs so that cell viability is maintained during exit from the cell cycle.


Assuntos
Apoptose , Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Mesângio Glomerular/metabolismo , Mesângio Glomerular/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Supressoras de Tumor , Animais , Apoptose/genética , Ciclina E/genética , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Regulação Enzimológica da Expressão Gênica , Genes Supressores de Tumor , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Oncogene ; 21(11): 1750-8, 2002 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-11896606

RESUMO

Apoptosis is closely linked to proliferation. In this study we showed that inducing apoptosis in mouse mesangial cells with ultraviolet (UV) irradiation was associated with increased cyclin A-cyclin dependent kinase (CDK) 2 activity. Inhibiting CDK2 activity with Roscovitine or dominant negative mutant reduced apoptosis. Because apoptosis typically begins in the cytoplasm, we tested the hypothesis that the subcellular localization of CDK2 determines the proliferative or apoptotic fate of the cell. Our results showed that cyclin A-CDK2 was nuclear in proliferating cells. However, inducing apoptosis in proliferating cells with UV irradiation was associated with a decrease in nuclear cyclin A and CDK2 protein levels. This coincided with an increase in protein and kinase activity for cyclin A-CDK2 in the cytoplasm. Translocation of cyclin A-CDK2 also occurred in p53-/- mesangial cells. Finally, we showed that caspase-3 activity was significantly reduced by inhibiting CDK2 activity with Roscovitine. In summary, our results show that apoptosis is associated with an increase in cytoplasmic cyclin A-CDK2 activity, which is p53 independent and upstream of caspase-3. We propose that the subcellular localization of CDK2 determines the proliferative or apoptotic fate of the cell.


Assuntos
Apoptose , Quinases relacionadas a CDC2 e CDC28 , Quinases Ciclina-Dependentes/fisiologia , Mesângio Glomerular/citologia , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Transporte Biológico , Caspase 3 , Caspases/fisiologia , Divisão Celular , Células Cultivadas , Ciclina A/fisiologia , Ciclina E/fisiologia , Quinase 2 Dependente de Ciclina , Quinases Ciclina-Dependentes/análise , Citoplasma/enzimologia , Mesângio Glomerular/enzimologia , Mesângio Glomerular/ultraestrutura , Camundongos , Membrana Nuclear/enzimologia , Proteínas Serina-Treonina Quinases/análise , Proteína Supressora de Tumor p53/fisiologia
8.
Curr Mol Med ; 13(1): 13-23, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23176147

RESUMO

Podocyte loss plays a key role in the progression of glomerular disorders towards glomerulosclerosis and chronic kidney disease. Podocytes form unique cytoplasmic extensions, foot processes, which attach to the outer surface of the glomerular basement membrane and interdigitate with neighboring podocytes to form the slit diaphragm. Maintaining these sophisticated structural elements requires an intricate actin cytoskeleton. Genetic, mechanic, and immunologic or toxic forms of podocyte injury can cause podocyte loss, which causes glomerular filtration barrier dysfunction, leading to proteinuria. Cell migration and cell division are two processes that require a rearrangement of the actin cytoskeleton; this rearrangement would disrupt the podocyte foot processes, therefore, podocytes have a limited capacity to divide or migrate. Indeed, all cells need to rearrange their actin cytoskeleton to assemble a correct mitotic spindle and to complete mitosis. Podocytes, even when being forced to bypass cell cycle checkpoints to initiate DNA synthesis and chromosome segregation, cannot complete cytokinesis efficiently and thus usually generate aneuploid podocytes. Such aneuploid podocytes rapidly detach and die, a process referred to as mitotic catastrophe. Thus, detached or dead podocytes cannot be adequately replaced by the proliferation of adjacent podocytes. However, even glomerular disorders with severe podocyte injury can undergo regression and remission, suggesting alternative mechanisms to compensate for podocyte loss, such as podocyte hypertrophy or podocyte regeneration from resident renal progenitor cells. Together, mitosis of the terminally differentiated podocyte rather accelerates podocyte loss and therefore glomerulosclerosis. Finding ways to enhance podocyte regeneration from other sources remains a challenge goal to improve the treatment of chronic kidney disease in the future.


Assuntos
Citoesqueleto/metabolismo , Mitose , Podócitos/patologia , Podócitos/fisiologia , Actinas/genética , Actinas/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Humanos , Nefropatias/patologia , Glomérulos Renais/citologia , Podócitos/citologia , Células-Tronco/metabolismo
9.
Kidney Int ; 72(1): 26-36, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17457377

RESUMO

Human genetic and in vivo animal studies have helped to define the critical importance of podocytes for kidney function in health and disease. However, as in any other research area, by default these approaches do not allow for mechanistic studies. Such mechanistic studies require the availability of cells grown ex vivo (i.e., in culture) with the ability to directly study mechanistic events and control the environment such that specific hypotheses can be tested. A seminal breakthrough came about a decade ago with the documentation of differentiation in culture of primary rat and human podocytes and the subsequent development of conditionally immortalized differentiated podocyte cell lines that allow deciphering the decisive steps of differentiation and function of 'in vivo' podocytes. Although this paper is not intended to provide a comprehensive review of podocyte biology, nor their role in proteinuric renal diseases or progressive glomerulosclerosis, it will focus specifically on several aspects of podocytes in culture. In particular, we will discuss the scientific and research rationale and need for cultured podocytes, how podocyte cell-culture evolved, and how cultured podocytes are currently being used to uncover novel functions of podocytes that can then be validated in vivo in animal or human studies. In addition, we provide a detailed description of how to properly culture and characterize podocytes to avoid potential pitfalls.


Assuntos
Técnicas de Cultura de Células/tendências , Podócitos/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Nefropatias/patologia , Nefropatias/fisiopatologia , Camundongos , Fenótipo , Podócitos/fisiologia , Ratos
10.
Kidney Int ; 72(4): 489-98, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17554257

RESUMO

Detachment or apoptosis of podocytes leads to proteinuria and glomerulosclerosis. There are no current interventions for diabetic or non-diabetic glomerular diseases specifically preventing podocyte apoptosis. Binding of erythropoiesis stimulating proteins (ESPs) to receptors on non-hematopoietic cells has been shown to have anti-apoptotic effects in vitro, in vivo, and in preliminary human studies. Recently, erythropoietin receptors were identified on podocytes; therefore, we tested effects of darbepoetin alfa in preventing podocyte apoptosis. Cultured immortalized mouse podocytes were treated with low-dose ultraviolet-C (uv-C) irradiation to induce apoptosis in the absence or presence of darbepoetin alfa. Apoptosis was quantified by Hoechst staining and by caspase 3 cleavage assessed by Western blots. Pretreatment with darbepoetin alfa significantly reduced podocyte apoptosis with this effect involving intact Janus family protein kinase-2 (JAK2) and AKT signaling pathways. Additionally, darbepoetin alfa was found protective against transforming growth factor-beta1 but not puromycin aminonucleoside induced apoptosis. Mice with anti-glomerular antibody induced glomerulonephritis had significantly less proteinuria, glomerulosclerosis, and podocyte apoptosis when treated with darbepoetin alfa. Our studies show that treatment of progressive renal diseases characterized by podocyte apoptosis with ESPs may be beneficial in slowing progression of chronic kidney disease.


Assuntos
Apoptose/efeitos dos fármacos , Eritropoetina/análogos & derivados , Glomerulonefrite/prevenção & controle , Podócitos/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Anticorpos , Apoptose/efeitos da radiação , Autoanticorpos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Darbepoetina alfa , Modelos Animais de Doenças , Progressão da Doença , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Glomerulonefrite/complicações , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Glomerulonefrite/patologia , Glomerulosclerose Segmentar e Focal/etiologia , Glomerulosclerose Segmentar e Focal/patologia , Glomerulosclerose Segmentar e Focal/prevenção & controle , Janus Quinase 2/metabolismo , Camundongos , Podócitos/metabolismo , Podócitos/patologia , Podócitos/efeitos da radiação , Substâncias Protetoras/uso terapêutico , Proteinúria/etiologia , Proteinúria/patologia , Proteinúria/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/metabolismo , Puromicina Aminonucleosídeo/farmacologia , Receptores da Eritropoetina/efeitos dos fármacos , Receptores da Eritropoetina/metabolismo , Transdução de Sinais/efeitos da radiação , Fator de Crescimento Transformador beta1/metabolismo , Raios Ultravioleta
11.
Kidney Int ; 72(4): 455-63, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17457371

RESUMO

Podocyte injury is a significant contributor to proteinuria and glomerulosclerosis. Recent studies have shown a renoprotective effect of erythropoietin (EPO) during ischemic kidney disease. In this study, we examine mechanisms by which a long acting recombinant EPO analog, darbepoetin, may confer renoprotection in the puromycin aminonucleoside-induced model of nephrotic syndrome. Darbepoetin decreased the proteinuria of rats treated with puromycin. This protective effect was correlated with the immunohistochemical disappearance of the podocyte injury markers desmin and the immune costimulator molecule B7.1 with the reappearance of nephrin expression in the slit diaphragm. Podocyte foot process retraction and effacement along with actin filament rearrangement, determined by electron microscopy, were all reversed by darbepoetin treatment. The protective effects were confirmed in puromycin-induced nephrotic rats that had been hemodiluted to normal hematocrit levels. Furthermore, puromycin treatment of rat podocytes in culture caused actin cytoskeletal reorganization along with deranged nephrin distribution. All these effects in vitro were reversed by darbepoetin. Our study demonstrates that darbepoetin treatment ameliorates podocyte injury and decreases proteinuria by a direct effect on podocytes. This may be accomplished by maintenance of the actin cytoskeleton and nephrin expression.


Assuntos
Citoesqueleto/efeitos dos fármacos , Eritropoetina/análogos & derivados , Proteínas de Membrana/metabolismo , Síndrome Nefrótica/prevenção & controle , Podócitos/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Proteinúria/prevenção & controle , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Antígeno B7-1/metabolismo , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/patologia , Darbepoetina alfa , Desmina/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Marcação In Situ das Extremidades Cortadas , Masculino , Síndrome Nefrótica/induzido quimicamente , Síndrome Nefrótica/complicações , Síndrome Nefrótica/metabolismo , Síndrome Nefrótica/patologia , Podócitos/metabolismo , Podócitos/ultraestrutura , Substâncias Protetoras/uso terapêutico , Proteinúria/etiologia , Proteinúria/metabolismo , Proteinúria/patologia , Puromicina Aminonucleosídeo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores Proteína Tirosina Quinases/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Fatores de Tempo
12.
Kidney Int ; 69(12): 2131-47, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16688120

RESUMO

The terminally differentiated podocyte, also called glomerular visceral epithelial cell, are highly specialized cells. They function as a critical size and charge barrier to prevent proteinuria. Podocytes are injured in diabetic and non-diabetic renal diseases. The clinical signature of podocyte injury is proteinuria, with or without loss of renal function owing to glomerulosclerosis. There is an exciting and expanding literature showing that hereditary, congenital, or acquired abnormalities in the molecular anatomy of podocytes leads to proteinuria, and at times, glomerulosclerosis. The change in podocyte shape, called effacement, is not simply a passive process following injury, but is owing to a complex interplay of proteins that comprise the molecular anatomy of the different protein domains of podocytes. These will be discussed in this review. Recent studies have also highlighted that a reduction in podocyte number directly causes proteinuria and glomerulosclerosis. This is owing to several factors, including the relative inability for these cells to proliferate, detachment, and apoptosis. The mechanisms of these events are being elucidated, and are discussed in this review. It is the hope that by delineating the events following injury to podocytes, therapies might be developed to reduce the burden of proteinuric renal diseases.


Assuntos
Nefropatias Diabéticas/fisiopatologia , Glomerulosclerose Segmentar e Focal/fisiopatologia , Podócitos/patologia , Podócitos/fisiologia , Proteinúria/patologia , Corticosteroides/farmacologia , Corticosteroides/uso terapêutico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Apoptose/fisiologia , Adesão Celular/fisiologia , Contagem de Células , Ciclo Celular/fisiologia , Proliferação de Células , Citoesqueleto/patologia , Citoesqueleto/fisiologia , Citoesqueleto/ultraestrutura , Dano ao DNA , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/patologia , Endotélio/patologia , Endotélio/fisiopatologia , Membrana Basal Glomerular/patologia , Membrana Basal Glomerular/fisiopatologia , Membrana Basal Glomerular/ultraestrutura , Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Hipertrofia/patologia , Hipertrofia/fisiopatologia , Podócitos/efeitos dos fármacos , Proteinúria/tratamento farmacológico , Proteinúria/fisiopatologia , Tretinoína/farmacologia , Tretinoína/uso terapêutico
13.
Kidney Int ; 70(11): 1962-73, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17035936

RESUMO

A decline in podocyte number correlates with progression to glomerulosclerosis. A mechanism underlying reduced podocyte number is the podocyte's relative inability to proliferate in response to injury. Injury by the podocyte toxin puromycin aminonucleoside (PA) is mediated via reactive oxygen species (ROS). The precise role of ROS in the pathogenesis of PA-induced glomerulosclerosis remains to be determined. We sought to examine whether PA-induced ROS caused podocyte DNA damage, possibly accounting for the podocyte's inability to proliferate in response to PA. In vitro, podocytes were exposed to PA, with or without the radical scavenger 1,3-dimethyl-2-thiourea (DMTU). In vivo, male Sprague-Dawley rats were divided into experimental groups (n = 6/group/time point): PA, PA with DMTU, and control, killed at days 1.5, 3, or 7. DNA damage was measured by DNA precipitation, apurinic/apyrimidinic site, Comet, and 8-hydroxydeoxyguanosine assays. Cell cycle checkpoint protein upregulation (by immunostaining and Western blotting), histopathology, and biochemical parameters were examined. DNA damage was increased in cultured podocytes that received PA, but not PA with DMTU. PA exposure activated specific cell cycle checkpoint proteins, with attenuation by DMTU. DNA repair enzymes were activated, providing evidence for attempted DNA repair. The PA-treated animals developed worse proteinuria and histopathologic disease and exhibited more DNA damage than the DMTU pretreated group. No significant apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling staining. A mechanism underlying the lack of podocyte proliferation following PA-induced injury in vitro and in vivo may be ROS-mediated DNA damage, with upregulation of specific cell cycle checkpoints leading to cell cycle arrest.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Dano ao DNA , Podócitos/efeitos dos fármacos , Puromicina Aminonucleosídeo/farmacologia , Animais , Apoptose , Proteínas de Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Enzimas Reparadoras do DNA/efeitos dos fármacos , Nefropatias/induzido quimicamente , Nefropatias/patologia , Masculino , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Proteinúria , Ratos , Ratos Sprague-Dawley
14.
Kidney Int ; 69(1): 99-104, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16374429

RESUMO

In previous work, we have demonstrated that Ras GTPases regulate proliferation in a range of human renal cells. The present work compares human and mouse mesangial cell (HMC and MMC) responses to specific knockdown of Ras genes with antisense oligonucleotides (AS-oligos), and examines the role of the p21 (cip1) and p27 (kip1) cyclin-dependent kinase inhibitors in these responses in mouse cells. HMC and MMC were lipofectin transfected with ras-targeted AS-oligo at 200-400 nM for 18 h followed by growth of cells in 20% serum for 18-72 h. Cell proliferation was assessed with an MTS assay and bromodeoxyuridine (BrdU) uptake. Apoptosis was quantified using nuclear stain with Hoechst 33342 dye. In MMC, Ha-ras AS-oligo caused an increase in apoptosis from <2% to 10-15% of cells after 18 h in serum (P<0.01). Control, Ki-ras and N-ras AS-oligos had minimal effects on apoptosis. BrdU uptake studies showed that BrdU+ve MMC were increased by 20-40% (P<0.05) after Ha-ras AS-oligo at 24 h; other ras AS-oligos were inactive. HMC number was reduced by 40-80% (P<0.01) at 48-72 h by both Ha-ras and Ki-ras AS-oligos. These actions were associated with reductions in BrdU+ve cells. In HMC, the ras AS-oligos did not induce apoptosis. p21(-,-) MMC showed exaggerated apoptotic responses to Ha-Ras AS-oligo. In mouse cells, Ha-Ras expression appears necessary to prevent apoptotic cell death; Ras expression does not appear necessary for cells to progress through the cell cycle. In human cells, Ras does not appear necessary to prevent apoptosis but Ha-Ras and Ki-Ras appear to be required for cell cycle progression.


Assuntos
Apoptose , Mesângio Glomerular/citologia , Proteínas ras/fisiologia , Animais , Proliferação de Células , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/análise , Inibidor de Quinase Dependente de Ciclina p27/análise , DNA/biossíntese , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-akt/análise , Proteínas Proto-Oncogênicas c-akt/fisiologia
15.
Kidney Int ; 56(4): 1208-15, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10610411

RESUMO

Evidence is accumulating that directly responsible for the rate of progression of glomerular disease are specific positive (cyclins and cyclin-dependent kinases) and negative (cyclin-kinase inhibitors) cell cycle regulatory proteins. The challenge for nephrologists is to determine which ones are expressed in renal disease and their precise role in glomerular cell proliferation, hypertrophy and differentiation. Ultimately the goal is to find ever more appropriate therapeutic strategies to arrest or prevent progressive renal disease.


Assuntos
Proteínas de Ciclo Celular/genética , Ciclo Celular/fisiologia , Mesângio Glomerular/patologia , Glomerulonefrite/patologia , Regulação Enzimológica da Expressão Gênica , Mesângio Glomerular/enzimologia , Glomerulonefrite/metabolismo , Humanos
16.
Kidney Int ; 47(3): 782-8, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7752577

RESUMO

Experimental type 1 diabetes mellitus is characterized by an early increase in kidney weight and glomerular volume, but changes in gene expression accompanying diabetic renal growth have not been elucidated. The early response genes, c-fos, c-jun, and c-myc encode proteins that regulate gene transcription, thus influencing the cellular responses to a stimulus. Accordingly, we studied c-fos, c-jun, and c-myc expression in glomeruli during the rapid phase of glomerular hypertrophy that follows the onset of hyperglycemia in diabetic rats. Total RNA was extracted by the method of Chomczynski from isolated glomeruli of streptozotocin (60 mg/kg i.v.) induced diabetic rats 24, 48, and 96 hours, and 1 week after the onset of hyperglycemia (blood glucose > 15 mmol/liter). A second group of rats, studied after streptozotocin administration, received twice daily insulin to maintain normoglycemia. A group of age-matched normal rats served as the control group. Northern blot analysis was performed with cDNA probes for c-fos, c-jun, and c-myc, and GAPDH. mRNA levels for c-fos increased fourfold 24 hours after the onset of hyperglycemia, but returned to baseline by 48 hours. mRNA levels for c-jun increased threefold 24 hours after the onset of hyperglycemia, in diabetic glomeruli, and the increase was sustained for one week. Intensive insulin treatment normalized blood glucose levels and abrogated the increases in c-fos and c-jun expression. There was no discernable increase in c-myc mRNA levels in the diabetic glomeruli.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Diabetes Mellitus Experimental/patologia , Glomérulos Renais/patologia , Proto-Oncogenes/fisiologia , Animais , Glicemia/metabolismo , Northern Blotting , Diabetes Mellitus Experimental/metabolismo , Expressão Gênica , Hipertrofia/genética , Hipertrofia/metabolismo , Técnicas In Vitro , Glomérulos Renais/metabolismo , Masculino , Tamanho do Órgão , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
17.
Am J Physiol Renal Physiol ; 278(4): F515-29, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10751212

RESUMO

The response to glomerular and tubulointerstitial cell injury in most forms of renal disease includes changes in cell number (proliferation and apoptosis) and cell size (hypertrophy). These events typically precede and may be responsible for the accumulation of extracellular matrix proteins that leads to a decrease in renal function. There is increasing evidence showing that positive (cyclins and cyclin-dependent kinases) and negative (cyclin-dependent kinase inhibitors) cell cycle regulatory proteins have a critical role in regulating these fundamental cellular responses to immune and nonimmune forms of injury. Data now show that altering specific cell cycle proteins affects renal cell proliferation and improves renal function. Equally exciting is the expanding body of literature showing novel biological roles for cell cycle proteins in the regulation of cell hypertrophy and apoptosis. With increasing understanding of the role for cell cycle regulatory proteins in renal disease comes the hope for potential therapeutic interventions.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Nefropatias/fisiopatologia , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Humanos , Hipertrofia , Rim/patologia , Nefropatias/patologia , Túbulos Renais/patologia
18.
Exp Nephrol ; 7(3): 207-11, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10352360

RESUMO

The growth response of resident glomerular cells is determined by the underlying disease. Thus glomerular cells can proliferate, fail to proliferate, hypertrophy or apoptose. Cell growth is controlled by cell cycle regulatory proteins, and cell proliferation requires that cyclin-dependent kinases (CDK) be activated by partner cyclins. Inhibiting CDK2 reduces mesangial cell proliferation. Mesangial cell proliferation also requires that levels of specific cyclin kinase inhibitors (CKI) decrease. In contrast, the visceral glomerular epithelial cells' inability to proliferate may be due to increased levels of CKI. Moreover it is becoming increasingly clear that mesangial cell hypertrophy in diabetes requires increased CKI expression. Finally, apoptosis, which is often linked to proliferation, may also be due to the increased activity of CDK2. Thus, identifying specific cell cycle regulatory proteins following injury may provide future targets for therapy in glomerular disease.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Nefropatias/fisiopatologia , Animais , Apoptose , Divisão Celular , Quinases Ciclina-Dependentes/fisiologia , Ciclinas/fisiologia , Humanos , Hipertrofia , Nefropatias/patologia , Glomérulos Renais/crescimento & desenvolvimento , Glomérulos Renais/patologia
19.
Miner Electrolyte Metab ; 24(2-3): 168-73, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9525701

RESUMO

Transforming growth factor-beta (TGF-beta) is an important cytokine in glomerular disease. Its major role may be to mediate extracellular matrix deposition, by both increasing the synthesis of matrix components and by reducing their degradation. Strong evidence supports the functional role for TGF-beta in mesangial matrix expansion. However, TGF-beta may also have other important functions in the glomerulus, including the regulation of cell proliferation, hypertrophy, and survival (apoptosis), as well as modulation of the local and systemic immune response.


Assuntos
Nefropatias , Glomérulos Renais , Fator de Crescimento Transformador beta/fisiologia , Animais , Divisão Celular , Sobrevivência Celular , Matriz Extracelular/patologia , Proteínas da Matriz Extracelular/metabolismo , Mesângio Glomerular/patologia , Humanos , Nefropatias/etiologia , Nefropatias/imunologia , Nefropatias/patologia , Glomérulos Renais/imunologia , Glomérulos Renais/patologia
20.
Exp Nephrol ; 7(2): 160-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10213869

RESUMO

Normal podocyte function requires attachment to the underlying glomerular basement membrane. Alteration or disruption of podocyte attachment occurs in many forms of glomerular injury, leading to the development of proteinuria and eventually progressive glomerulosclerosis. The inability of podocytes to proliferate and thereby recover denuded glomerular basement membrane areas may be central to the pathogenesis of certain forms of glomerular diseases.


Assuntos
Membrana Basal/fisiologia , Células Epiteliais/fisiologia , Glomerulonefrite/fisiopatologia , Nefropatias/fisiopatologia , Glomérulos Renais/fisiologia , Animais , Adesão Celular , Epitélio/fisiopatologia , Humanos , Glomérulos Renais/fisiopatologia , Proteinúria/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA