Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 318(6): F1377-F1390, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32308020

RESUMO

Ste20-like kinase SLK is critical for embryonic development and may play an important role in wound healing, muscle homeostasis, cell migration, and tumor growth. Mice with podocyte-specific deletion of SLK show albuminuria and damage to podocytes as they age. The present study addressed the role of SLK in glomerular injury. We induced adriamycin nephrosis in 3- to 4-mo-old control and podocyte SLK knockout (KO) mice. Compared with control, SLK deletion exacerbated albuminuria and loss of podocytes, synaptopodin, and podocalyxin. Glomeruli of adriamycin-treated SLK KO mice showed diffuse increases in the matrix and sclerosis as well as collapse of the actin cytoskeleton. SLK can phosphorylate ezrin. The complex of phospho-ezrin, Na+/H+ exchanger regulatory factor 2, and podocalyxin in the apical domain of the podocyte is a key determinant of normal podocyte architecture. Deletion of SLK reduced glomerular ezrin and ezrin phosphorylation in adriamycin nephrosis. Also, deletion of SLK reduced the colocalization of ezrin and podocalyxin in the glomerulus. Cultured glomerular epithelial cells with KO of SLK showed reduced ezrin phosphorylation and podocalyxin expression as well as reduced F-actin. Thus, SLK deletion leads to podocyte injury as mice age and exacerbates injury in adriamycin nephrosis. The mechanism may at least in part involve ezrin phosphorylation as well as disruption of the cytoskeleton and podocyte apical membrane structure.


Assuntos
Citoesqueleto de Actina/enzimologia , Doxorrubicina , Glomerulosclerose Segmentar e Focal/enzimologia , Nefrose/enzimologia , Podócitos/enzimologia , Proteínas Serina-Treonina Quinases/deficiência , Citoesqueleto de Actina/patologia , Actinas/metabolismo , Albuminúria/induzido quimicamente , Albuminúria/enzimologia , Albuminúria/genética , Animais , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Camundongos Knockout , Proteínas dos Microfilamentos/metabolismo , Nefrose/induzido quimicamente , Nefrose/genética , Nefrose/patologia , Fosfoproteínas/metabolismo , Fosforilação , Podócitos/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo
2.
Lab Invest ; 100(7): 945-958, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32203149

RESUMO

Podocyte injury and endoplasmic reticulum (ER) stress have been implicated in the pathogenesis of various glomerular diseases. ERdj3 (DNAJB11) and mesencephalic astrocyte-derived neurotrophic factor (MANF) are ER chaperones lacking the KDEL motif, and may be secreted extracellularly. Since podocytes reside in the urinary space, we examined if podocyte injury is associated with secretion of KDEL-free ER chaperones from these cells into the urine, and if chaperones in the urine reflect ER stress in glomerulonephritis. In cultured podocytes, ER stress increased ERdj3 and MANF intracellularly and in culture medium, whereas GRP94 (KDEL chaperone) increased only intracellularly. ERdj3 and MANF secretion was blocked by the secretory trafficking inhibitor, brefeldin A. Urinary ERdj3 and MANF increased in rats injected with tunicamycin (in the absence of proteinuria). After induction of passive Heymann nephritis (PHN) and puromycin aminonucleoside nephrosis (PAN), there was an increase in glomerular ER stress, and appearance of ERdj3 and MANF in the urine, coinciding with the onset of proteinuria. Rats with PHN were treated with the chemical chaperone, 4-phenyl butyrate (PBA), starting at the time of disease induction, or after disease was established. In both protocols, 4-PBA reduced proteinuria and urinary ER chaperone secretion, compared with PHN rats treated with saline (control). In conclusion, urinary ERdj3 and MANF reflect glomerular ER stress. 4-PBA protected against complement-mediated podocyte injury and the therapeutic response could be monitored by urinary ERdj3 and MANF.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Glomerulonefrite/urina , Proteínas de Choque Térmico HSP40/urina , Fatores de Crescimento Neural/urina , Animais , Células Cultivadas , Glomerulonefrite/metabolismo , Glomerulonefrite/fisiopatologia , Proteínas de Choque Térmico HSP40/metabolismo , Glomérulos Renais/citologia , Glomérulos Renais/efeitos dos fármacos , Masculino , Camundongos , Fatores de Crescimento Neural/metabolismo , Ratos , Ratos Sprague-Dawley , Tunicamicina/farmacologia
3.
Kidney Int ; 96(4): 850-861, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31420194

RESUMO

The podocyte is a key component of the glomerular filtration barrier. Podocyte dysfunction is central to the underlying pathophysiology of many common glomerular diseases, including diabetic nephropathy, glomerulonephritis and genetic forms of nephrotic syndrome. Collectively, these conditions affect millions of people worldwide, and account for the majority of kidney diseases requiring dialysis and transplantation. The 12th International Podocyte Conference was held in Montreal, Canada from May 30 to June 2, 2018. The primary aim of this conference was to bring together nephrologists, clinician scientists, basic scientists and their trainees from all over the world to present their research and to establish networks with the common goal of developing new therapies for glomerular diseases based on the latest advances in podocyte biology. This review briefly highlights recent advances made in understanding podocyte structure and metabolism, experimental systems in which to study podocytes and glomerular disease, disease mediators, genetic and immune origins of glomerulopathies, and the development of novel therapeutic agents to protect podocyte and glomerular injury.


Assuntos
Nefropatias Diabéticas/terapia , Barreira de Filtração Glomerular/fisiopatologia , Glomerulonefrite/terapia , Síndrome Nefrótica/terapia , Podócitos/patologia , Animais , Canadá , Congressos como Assunto , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Glomerulonefrite/patologia , Glomerulonefrite/fisiopatologia , Humanos , Síndrome Nefrótica/patologia , Síndrome Nefrótica/fisiopatologia
4.
Am J Physiol Renal Physiol ; 315(1): F186-F198, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29187370

RESUMO

SLK is essential for embryonic development and may play a key role in wound healing, tumor growth, and metastasis. Expression and activation of SLK are increased in kidney development and during recovery from ischemic acute kidney injury. Overexpression of SLK in glomerular epithelial cells/podocytes in vivo induces injury and proteinuria. Conversely, reduced SLK expression leads to abnormalities in cell adhesion, spreading, and motility. Tight regulation of SLK expression thus may be critical for normal renal structure and function. We produced podocyte-specific SLK-knockout mice to address the functional role of SLK in podocytes. Mice with podocyte-specific deletion of SLK showed reduced glomerular SLK expression and activity compared with control. Podocyte-specific deletion of SLK resulted in albuminuria at 4-5 mo of age in male mice and 8-9 mo in female mice, which persisted for up to 13 mo. At 11-12 mo, knockout mice showed ultrastructural changes, including focal foot process effacement and microvillous transformation of podocyte plasma membranes. Mean foot process width was approximately twofold greater in knockout mice compared with control. Podocyte number was reduced by 35% in knockout mice compared with control, and expression of nephrin, synaptopodin, and podocalyxin was reduced in knockout mice by 20-30%. In summary, podocyte-specific deletion of SLK leads to albuminuria, loss of podocytes, and morphological evidence of podocyte injury. Thus, SLK is essential to the maintenance of podocyte integrity as mice age.


Assuntos
Albuminúria/enzimologia , Glomérulos Renais/enzimologia , Podócitos/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Fatores Etários , Albuminúria/genética , Albuminúria/fisiopatologia , Animais , Adesão Celular , Células Cultivadas , Colágeno/metabolismo , Feminino , Predisposição Genética para Doença , Glomérulos Renais/fisiopatologia , Glomérulos Renais/ultraestrutura , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fenótipo , Podócitos/ultraestrutura , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Ratos , Proteínas Repressoras/metabolismo , Fatores Sexuais , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , Proteínas WT1
5.
Am J Physiol Renal Physiol ; 315(4): F954-F966, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29873512

RESUMO

Mutations in α-actinin-4 (actinin-4) result in hereditary focal segmental glomerulosclerosis (FSGS) in humans. Actinin-4 mutants induce podocyte injury because of dysregulation of the cytoskeleton and proteotoxicity. Injury may be associated with endoplasmic reticulum (ER) stress and polyubiquitination of proteins. We assessed if the chemical chaperone 4-phenylbutyrate (4-PBA) can ameliorate the proteotoxicity of an actinin-4 mutant. Actinin-4 K255E, which causes FSGS in humans (K256E in the mouse), showed enhanced ubiquitination, accelerated degradation, aggregate formation, and enhanced association with filamentous (F)-actin in glomerular epithelial cells (GECs). The mutant disrupted ER function and stimulated autophagy. 4-PBA reduced actinin-4 K256E aggregation and its tight association with F-actin. Transgenic mice that express actinin-4 K256E in podocytes develop podocyte injury, proteinuria, and FSGS in association with glomerular ER stress. Treatment of these mice with 4-PBA in the drinking water over a 10-wk period significantly reduced albuminuria and ER stress. Another drug, celastrol, which enhanced expression of ER and cytosolic chaperones in GECs, tended to reduce actinin-4 aggregation but did not decrease the tight association of actinin-4 K256E with F-actin and did not reduce albuminuria in actinin-4 K256E transgenic mice. Thus, chemical chaperones, such as 4-PBA, may represent a novel therapeutic approach to certain hereditary glomerular diseases.


Assuntos
Actinina/genética , Glomérulos Renais/lesões , Mutação/genética , Proteostase/genética , Citoesqueleto de Actina/metabolismo , Animais , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Glomerulosclerose Segmentar e Focal/metabolismo , Glomérulos Renais/metabolismo , Camundongos Transgênicos , Podócitos/metabolismo , Proteinúria/metabolismo
6.
J Biol Chem ; 291(28): 14468-82, 2016 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-27226532

RESUMO

Glomerular visceral epithelial cells (podocytes) play a critical role in the maintenance of glomerular permselectivity. Podocyte injury, manifesting as proteinuria, is the cause of many glomerular diseases. We reported previously that calcium-independent phospholipase A2γ (iPLA2γ) is cytoprotective against complement-mediated glomerular epithelial cell injury. Studies in iPLA2γ KO mice have demonstrated an important role for iPLA2γ in mitochondrial lipid turnover, membrane structure, and metabolism. The aim of the present study was to employ iPLA2γ KO mice to better understand the role of iPLA2γ in normal glomerular and podocyte function as well as in glomerular injury. We show that deletion of iPLA2γ did not cause detectable albuminuria; however, it resulted in mitochondrial structural abnormalities and enhanced autophagy in podocytes as well as loss of podocytes in aging KO mice. Moreover, after induction of anti-glomerular basement membrane nephritis in young mice, iPLA2γ KO mice exhibited significantly increased levels of albuminuria, podocyte injury, and loss of podocytes compared with wild type. Thus, iPLA2γ has a protective functional role in the normal glomerulus and in glomerulonephritis. Understanding the role of iPLA2γ in glomerular pathophysiology provides opportunities for the development of novel therapeutic approaches to glomerular injury and proteinuria.


Assuntos
Glomerulonefrite/genética , Fosfolipases A2 do Grupo VI/genética , Glomérulos Renais/patologia , Podócitos/patologia , Envelhecimento , Animais , Autofagia , Células Cultivadas , Estresse do Retículo Endoplasmático , Glomerulonefrite/patologia , Glomérulos Renais/metabolismo , Proteínas de Membrana/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/patologia , Fosfolipases A2 Independentes de Cálcio/genética , Podócitos/metabolismo , Proteinúria/genética , Proteinúria/patologia
7.
Biochim Biophys Acta ; 1863(9): 2147-55, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27216364

RESUMO

Expression and activation of SLK increases during renal ischemia-reperfusion injury. When highly expressed, SLK signals via c-Jun N-terminal kinase and p38 to induce apoptosis, and it exacerbates apoptosis induced by ischemia-reperfusion injury. Overexpression of SLK in glomerular epithelial cells (GECs)/podocytes in vivo induces injury and proteinuria. In response to various stresses, cells enhance expression of chaperones or heat shock proteins (e.g. Hsp70), which are involved in the folding and maturation of newly synthesized proteins, and can refold denatured or misfolded proteins. We address the interaction of SLK with the heat shock factor 1 (HSF1)-Hsp70 pathway. Increased expression of SLK in GECs (following transfection) induced HSF1 transcriptional activity. Moreover, HSF1 transcriptional activity was increased by in vitro ischemia-reperfusion injury (chemical anoxia/recovery) and heat shock, and in both instances was amplified further by SLK overexpression. HSF1 binds to promoters of target genes, such as Hsp70 and induces their transcription. By analogy to HSF1, SLK stimulated Hsp70 expression. Hsp70 was also enhanced by anoxia/recovery and was further amplified by SLK overexpression. Induction of HSF1 and Hsp70 was dependent on the kinase activity of SLK, and was mediated via polo-like kinase-1. Transfection of constitutively active HSF1 enhanced Hsp70 expression and inhibited SLK-induced apoptosis. Conversely, the proapoptotic action of SLK was augmented by HSF1 shRNA, or the Hsp70 inhibitor, pifithrin-µ. In conclusion, increased expression/activity of SLK activates the HSF1-Hsp70 pathway. Hsp70 attenuates the primary proapoptotic effect of SLK. Modulation of chaperone expression may potentially be harnessed as cytoprotective therapy in renal cell injury.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Células COS , Proteínas de Ciclo Celular/metabolismo , Morte Celular , Hipóxia Celular , Chlorocebus aethiops , Células Epiteliais/metabolismo , Fatores de Transcrição de Choque Térmico , Resposta ao Choque Térmico , Glomérulos Renais/citologia , Mutação/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ativação Transcricional/genética , Quinase 1 Polo-Like
8.
Kidney Int ; 102(3): 669, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35988938
9.
J Biol Chem ; 290(5): 3009-20, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25492867

RESUMO

Injury of visceral glomerular epithelial cells (GECs) causes proteinuria in many glomerular diseases. We reported previously that calcium-independent phospholipase A2γ (iPLA2γ) is cytoprotective against complement-mediated GEC injury. Because iPLA2γ is localized at the endoplasmic reticulum (ER), this study addressed whether the cytoprotective effect of iPLA2γ involves the ER stress unfolded protein response (UPR). In cultured rat GECs, overexpression of the full-length iPLA2γ, but not a mutant iPLA2γ that fails to associate with the ER, augmented tunicamycin-induced activation of activating transcription factor-6 (ATF6) and induction of the ER chaperones, glucose-regulated protein 94 (GRP94) and glucose-regulated protein 78 (GRP78). Augmented responses were inhibited by the iPLA2γ inhibitor, (R)-bromoenol lactone, but not by the cyclooxygenase inhibitor, indomethacin. Tunicamycin-induced cytotoxicity was reduced in GECs expressing iPLA2γ, and the cytoprotection was reversed by dominant-negative ATF6. GECs from iPLA2γ knock-out mice showed blunted ATF6 activation and chaperone up-regulation in response to tunicamycin. Unlike ATF6, the two other UPR pathways, i.e. inositol-requiring enzyme 1α and protein kinase RNA-like ER kinase pathways, were not affected by iPLA2γ. Thus, in GECs, iPLA2γ amplified activation of the ATF6 pathway of the UPR, resulting in up-regulation of ER chaperones and cytoprotection. These effects were dependent on iPLA2γ catalytic activity and association with the ER but not on prostanoids. Modulating iPLA2γ activity may provide opportunities for pharmacological intervention in glomerular diseases associated with ER stress.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Fosfolipases A2 Independentes de Cálcio/metabolismo , Fator 6 Ativador da Transcrição/genética , Animais , Células COS , Chlorocebus aethiops , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/genética , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Fosfolipases A2 Independentes de Cálcio/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tunicamicina/farmacologia
10.
Biochim Biophys Acta ; 1853(10 Pt A): 2539-52, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26094769

RESUMO

Expression and activity of the Ste20-like kinase, SLK, are increased during kidney development and recovery from ischemia-reperfusion injury. SLK mediates apoptosis in various cells, and can regulate cell cycle progression and cytoskeletal remodeling. In cells, SLK is detected in a high molecular mass complex, suggesting that SLK is a dimer/oligomer, or is in tight association with other proteins. To better understand the regulation, localization and function of SLK, we sought to identify proteins in this high molecular mass complex. Analysis by mass spectroscopy identified the nucleoporin, translocated promoter region (Tpr), and the cytoskeletal protein, α-actinin-4, as potential SLK-interacting proteins. Using a protein complementation assay, we showed that the 350 amino acid C-terminal, coiled-coil domain of SLK was responsible for homodimerization, as well as interaction with Tpr and α-actinin-4. The association of SLK with Tpr and α-actinin-4, respectively, was confirmed by co-immunoprecipitation. Subsets of total cellular SLK colocalized with Tpr at the nuclear envelope, and α-actinin-4 in the cytoplasm. Expression of Tpr attenuated activation-specific autophosphorylation of SLK, and blocked SLK-induced apoptosis and AP-1 activity. In contrast to the effect of Tpr, autophosphorylation of SLK was not affected by α-actinin-4. Thus, SLK interacts with Tpr and α-actinin-4 in cells, and these protein-protein interactions may control the subcellular localization and the biological activity of SLK.


Assuntos
Actinina/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Actinina/genética , Animais , Células COS , Chlorocebus aethiops , Humanos , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Fosforilação/fisiologia , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico/fisiologia , Proteínas Proto-Oncogênicas/genética
11.
Am J Physiol Renal Physiol ; 311(5): F1035-F1046, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27630065

RESUMO

Kidney cell injury may be associated with protein misfolding and induction of endoplasmic reticulum (ER) stress. Examples include complement-induced glomerular epithelial cell (GEC)/podocyte injury in membranous nephropathy and ischemia-reperfusion injury. Renal cell injury can also result from mutations in integral proteins, which lead to their misfolding and accumulation. Certain nephrin missense mutants misfold, accumulate in the ER, and induce ER stress. We examined if enhancement of ubiquitin-proteasome system function may facilitate proteostasis and confer protection against injury. Ubiquitin-specific protease 14 (Usp14) is reported to retard proteasomal protein degradation. Thus inhibition of Usp14 may enhance degradation of misfolded proteins and attenuate cell injury. In GEC, the reporter proteins GFPu (a "misfolded" protein) and CD3δ (an ER-associated degradation substrate) undergo time-dependent proteasomal degradation. Complement did not affect degradation of CD3δ-yellow fluorescent protein (YFP), but accelerated degradation of GFPu, and the Usp14-directed inhibitor IU1 further accelerated this degradation. Conversely, overexpression of Usp14 reduced degradation of GFPu and CD3δ-YFP. In 293T cells, IU1 did not enhance degradation of disease-associated nephrin missense mutants I171N and S724C, whereas overexpression of Usp14 reduced degradation. IU1 was cytoprotective after injury induced by the ER stressor tunicamycin and in vitro ischemia-reperfusion, but did not affect complement-induced cytotoxicity. In conclusion, Usp14 controls proteasomal degradation of some misfolded proteins. In addition, a Usp14-directed inhibitor reduces cytotoxicity in the context of global protein misfolding during certain types of renal cell injury.


Assuntos
Células Epiteliais/metabolismo , Glomérulos Renais/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Ubiquitina Tiolesterase/metabolismo , Proteínas do Sistema Complemento/metabolismo , Células HEK293 , Humanos , Proteólise , Traumatismo por Reperfusão/metabolismo
12.
Am J Physiol Renal Physiol ; 310(10): F1148-56, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26719363

RESUMO

Focal segmental glomerular sclerosis (FSGS) is an irreversible renal pathology characterized by podocyte detachment from the glomerular basement membrane, hyalinosis, and sclerosis. Clinically, it manifests with proteinuria and progressive loss of glomerular filtration. Primary idiopathic FSGS can occur in isolation and frequently progresses to end-stage renal disease, requiring dialysis or kidney transplantation. In 30-50% of these patients, proteinuria and FSGS recur in the renal allograft, suggesting the presence of a podocyte-toxic factor(s) in the recipient's serum. Currently, there is no reliable way to quantify the serum activity or predict the subset of FSGS patients at risk for recurrence after transplantation. We describe a novel in vitro method that measures the podocyte-toxic activity of sera from FSGS patients using cultured human podocytes; we compare this with the effect of compounds such as adriamycin. Using immunofluorescence microscopy followed by computerized image-processing analysis, we show that incubation of human podocytes with adriamycin leads to a dose-dependent disassembly of focal adhesion complexes (FACs). We then demonstrate that sera from patients with posttransplant recurrent or idiopathic FSGS cause a similar FAC disturbance. In contrast, sera from nonrecurrent FSGS patients do not affect FACs. In some FSGS patients, toxic effects of serum can be prevented by blockade of the tumor necrosis factor-α pathway. We propose that this method may be useful as a diagnostic tool to identify FSGS patients with serum podocyte-toxic activity that presumably places them at increased risk for recurrence in the renal allograft.


Assuntos
Adesões Focais/efeitos dos fármacos , Glomerulosclerose Segmentar e Focal/sangue , Podócitos/efeitos dos fármacos , Toxinas Biológicas/sangue , Adolescente , Adulto , Células Cultivadas , Criança , Pré-Escolar , Doxorrubicina , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva , Reprodutibilidade dos Testes , Medição de Risco , Toxinas Biológicas/toxicidade , Fator de Necrose Tumoral alfa/metabolismo , Adulto Jovem
13.
J Am Soc Nephrol ; 26(3): 576-86, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25145929

RESUMO

The planar cell polarity (PCP) signaling pathway is crucial for tissue morphogenesis. Van Gogh-like protein 2 (Vangl2) is central in the PCP pathway; in mice, Vangl2 loss is embryonically lethal because of neural tube defects, and mutations in Vangl2 are associated with human neural tube defects. In the kidney, PCP signaling may be important for tubular morphogenesis and organization of glomerular epithelial cells (podocytes) along the glomerular basement membrane. Podocyte cell protrusions (foot processes) are critical for glomerular permselectivity; loss of foot process architecture results in proteinuria and FSGS. Previously, we showed a profound effect of PCP signaling on podocyte shape, actin rearrangement, cell motility, and nephrin endocytosis. To test our hypothesis that the PCP pathway is involved in glomerular development and function and circumvent lethality of the ubiquitous Vangl2 mutation in the Looptail mouse, we generated a mouse model with a podocyte-specific ablation of the Vangl2 gene. We report here that podocyte-specific deletion of Vangl2 leads to glomerular maturation defects in fetal kidneys. In adult mice, we detected significantly smaller glomeruli, but it did not affect glomerular permselectivity in aging animals. However, in the context of glomerular injury induced by injection of antiglomerular basement membrane antibody, deletion of Vangl2 resulted in exacerbation of injury and accelerated progression to chronic segmental and global glomerular sclerosis. Our results indicate that Vangl2 function in podocytes is important for glomerular development and protects against glomerular injury in adult animals.


Assuntos
Polaridade Celular , Glomérulos Renais/embriologia , Proteínas do Tecido Nervoso/metabolismo , Animais , Feminino , Glomérulos Renais/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfogênese , Podócitos/metabolismo
14.
J Biol Chem ; 288(6): 3871-85, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23258543

RESUMO

In experimental membranous nephropathy, complement C5b-9-induces glomerular epithelial cell (GEC) injury and proteinuria. The effects of C5b-9 are mediated via signaling pathways, including calcium-independent phospholipase A(2)γ (iPLA(2)γ), and mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38. The iPLA(2)γ pathway is cytoprotective. This study addresses the mechanisms of iPLA(2)γ activation. iPLA(2)γ activity was monitored by quantifying prostaglandin E(2) (PGE(2)) production. In GECs, iPLA(2)γ localized at the endoplasmic reticulum and mitochondria. Complement-mediated production of PGE(2) was amplified in GECs that overexpress iPLA(2)γ, compared with control cells, and was blocked by the iPLA(2)γ inhibitor bromoenol lactone in both iPLA(2)γ-overexpressing and control GECs. In GECs that overexpress iPLA(2)γ, complement-mediated PGE(2) production was reduced by inhibitors of MAP/ERK kinase 1 (MEK1) and p38 but not JNK. In COS-1 cells that overexpress iPLA(2)γ and cyclooxygenase-1, PGE(2) production was induced by co-expression of constitutively active MEK1 or MAPK-interacting kinase 1 (MNK1) as well as by stimulation with epidermal growth factor (EGF) + ionomycin. Complement- and EGF + ionomycin-stimulated iPLA(2)γ activity was attenuated by the S511A/S515A double mutation. Moreover, complement and EGF + ionomycin enhanced phosphorylation of Ser-511. Thus, complement-mediated activation of iPLA(2)γ is mediated via ERK and p38 pathways, and phosphorylation of Ser-511 and/or Ser-515 plays a key role in the catalytic activity and signaling of iPLA(2)γ. Defining the mechanisms by which complement activates iPLA(2)γ provides opportunities for development of novel therapeutic approaches to GEC injury and proteinuria.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Glomerulonefrite Membranosa/enzimologia , Fosfolipases A2 do Grupo VI/metabolismo , Glomérulos Renais/enzimologia , Substituição de Aminoácidos , Animais , Células COS , Ionóforos de Cálcio/farmacologia , Linhagem Celular , Chlorocebus aethiops , Complexo de Ataque à Membrana do Sistema Complemento/farmacologia , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Fator de Crescimento Epidérmico/farmacologia , Glomerulonefrite Membranosa/genética , Glomerulonefrite Membranosa/patologia , Fosfolipases A2 do Grupo VI/genética , Humanos , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ionomicina/farmacologia , Glomérulos Renais/lesões , Glomérulos Renais/patologia , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , Mutação de Sentido Incorreto , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteinúria/enzimologia , Proteinúria/genética , Ratos
15.
Am J Physiol Renal Physiol ; 307(5): F634-47, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25056348

RESUMO

The unfolded protein response and endoplasmic reticulum-associated degradation (ERAD) contribute to injury in renal glomerular diseases, including those mediated by complement C5b-9. In the present study, we address the role of protein-tyrosine phosphatase 1B (PTP1B) in complement-mediated glomerular injury and ERAD. In glomerular epithelial cells (GECs)/podocytes and PTP1B-deficient mouse embryonic fibroblasts exposed to complement, inhibition/deletion of PTP1B reduced ERAD, as monitored by the ERAD reporter CD3δ. Overexpression of PTP1B produced an effect similar to PTP1B deficiency on ERAD in complement-treated GECs. Complement-mediated cytotoxicity was reduced after PTP1B overexpression and tended to be reduced after PTP1B inhibition. PTP1B enhanced the induction of certain ERAD components via the inositol-requiring-1α branch of the unfolded protein response. PTP1B knockout mice with anti-glomerular basement membrane glomerulonephritis had decreased proteinuria and showed less podocyte loss and endoplasmic reticulum dysfunction compared with wild-type littermates. These results imply that endogenous levels of PTP1B are tightly regulated and that both overexpression and inhibition can affect ERAD. The cytoprotective effects of PTP1B deletion in cultured cells and in anti-glomerular basement membrane nephritis suggest that PTP1B may potentially be a therapeutic target in complement-mediated diseases.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Degradação Associada com o Retículo Endoplasmático/fisiologia , Glomerulonefrite/prevenção & controle , Glomerulonefrite/fisiopatologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/antagonistas & inibidores , Animais , Células Cultivadas , Proteínas do Sistema Complemento/farmacologia , Modelos Animais de Doenças , Degradação Associada com o Retículo Endoplasmático/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibroblastos/fisiologia , Glomerulonefrite/patologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Podócitos/efeitos dos fármacos , Podócitos/patologia , Podócitos/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Ratos , Resposta a Proteínas não Dobradas/fisiologia
16.
Am J Kidney Dis ; 63(3): 363-77, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24423780

RESUMO

The KDIGO (Kidney Disease: Improving Global Outcomes) clinical practice guideline for management of glomerulonephritis was recently released. The Canadian Society of Nephrology convened a working group to review the recommendations and comment on their relevancy and applicability to the Canadian context. A subgroup of adult nephrologists reviewed the guideline statements for management of glomerular disease in adults and agreed with most of the guideline statements developed by KDIGO. This commentary highlights areas for which there is lack of evidence and areas in need of translation of evidence into clinical practice. Areas of controversy or uncertainty, including the choice of second-line agents, are discussed in more detail. Existing practice variation also is addressed. The relevance of treatment recommendations to the Canadian practitioner is discussed.


Assuntos
Gerenciamento Clínico , Glomerulonefrite/terapia , Nefrologia , Guias de Prática Clínica como Assunto , Sociedades Médicas , Adulto , Canadá , Humanos
17.
Am J Kidney Dis ; 63(3): 354-62, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24423782

RESUMO

The KDIGO (Kidney Disease: Improving Global Outcomes) clinical practice guideline for management of glomerulonephritis was recently released. The Canadian Society of Nephrology convened a working group to review the recommendations and comment on their relevancy and applicability to the Canadian context. A subgroup of pediatric nephrologists reviewed the guideline statements for management of childhood nephrotic syndrome and agreed with most of the guideline statements developed by KDIGO. This commentary highlights areas in which there is lack of evidence and areas in need of translation of evidence into clinical practice. Areas of controversy or uncertainty, including the length of corticosteroid therapy for the initial presentation and relapses, definitions of steroid resistance, and choice of second-line agents, are discussed in more detail. Existing practice variation is also addressed.


Assuntos
Gerenciamento Clínico , Glomerulonefrite/tratamento farmacológico , Glucocorticoides/uso terapêutico , Nefrologia , Síndrome Nefrótica/tratamento farmacológico , Guias de Prática Clínica como Assunto , Sociedades Médicas , Canadá , Criança , Glomerulonefrite/complicações , Humanos , Síndrome Nefrótica/etiologia , Prognóstico
18.
Physiol Rep ; 12(1): e15897, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38163671

RESUMO

SLK controls the cytoskeleton, cell adhesion, and migration. Podocyte-specific deletion of SLK in mice leads to podocyte injury as mice age and exacerbates injury in experimental focal segment glomerulosclerosis (FSGS; adriamycin nephrosis). We hypothesized that adhesion proteins may be substrates of SLK. In adriamycin nephrosis, podocyte ultrastructural injury was exaggerated by SLK deletion. Analysis of a protein kinase phosphorylation site dataset showed that podocyte adhesion proteins-paxillin, vinculin, and talin-1 may be potential SLK substrates. In cultured podocytes, deletion of SLK increased adhesion to collagen. Analysis of paxillin, vinculin, and talin-1 showed that SLK deletion reduced focal adhesion complexes (FACs) containing these proteins mainly in adriamycin-induced injury; there was no change in FAC turnover (focal adhesion kinase Y397 phosphorylation). In podocytes, paxillin S250 showed basal phosphorylation that was slightly enhanced by SLK; however, SLK did not phosphorylate talin-1. In adriamycin nephrosis, SLK deletion did not alter glomerular expression/localization of talin-1 and vinculin, but increased focal adhesion kinase phosphorylation modestly. Therefore, SLK decreases podocyte adhesion, but FAC proteins in podocytes are not major substrates of SLK in health and disease.


Assuntos
Nefrose , Podócitos , Camundongos , Animais , Podócitos/metabolismo , Paxilina/metabolismo , Vinculina/metabolismo , Talina/genética , Talina/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Doxorrubicina/toxicidade , Proteínas Serina-Treonina Quinases/metabolismo
19.
Sci Rep ; 14(1): 11718, 2024 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-38778209

RESUMO

Protein misfolding in the endoplasmic reticulum (ER) of podocytes contributes to the pathogenesis of glomerular diseases. Protein misfolding activates the unfolded protein response (UPR), a compensatory signaling network. We address the role of the UPR and the UPR transducer, inositol-requiring enzyme 1α (IRE1α), in streptozotocin-induced diabetic nephropathy in mice. Diabetes caused progressive albuminuria in control mice that was exacerbated in podocyte-specific IRE1α knockout (KO) mice. Compared to diabetic controls, diabetic IRE1α KO mice showed reductions in podocyte number and synaptopodin. Glomerular ultrastructure was altered only in diabetic IRE1α KO mice; the major changes included widening of podocyte foot processes and glomerular basement membrane. Activation of the UPR and autophagy was evident in diabetic control, but not diabetic IRE1α KO mice. Analysis of human glomerular gene expression in the JuCKD-Glom database demonstrated induction of genes associated with the ER, UPR and autophagy in diabetic nephropathy. Thus, mice with podocyte-specific deletion of IRE1α demonstrate more severe diabetic nephropathy and attenuation of the glomerular UPR and autophagy, implying a protective effect of IRE1α. These results are consistent with data in human diabetic nephropathy and highlight the potential for therapeutically targeting these pathways.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Endorribonucleases , Podócitos , Proteínas Serina-Treonina Quinases , Animais , Humanos , Masculino , Camundongos , Albuminúria/etiologia , Albuminúria/genética , Autofagia/genética , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Endorribonucleases/metabolismo , Endorribonucleases/genética , Deleção de Genes , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Resposta a Proteínas não Dobradas
20.
J Biol Chem ; 287(8): 5446-58, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22203681

RESUMO

Expression and activation of the Ste20-like kinase, SLK, is increased during kidney development and recovery from ischemic acute kidney injury. SLK promotes apoptosis, and it may regulate cell survival during injury or repair. This study addresses the role of phosphorylation in the regulation of kinase activity. We mutated serine and threonine residues in the putative activation segment of the SLK catalytic domain and expressed wild type (WT) and mutant proteins in COS-1 or glomerular epithelial cells. Compared with SLK WT, the T183A, S189A, and T183A/S189A mutants showed reduced in vitro kinase activity. SLK WT, but not mutants, increased activation-specific phosphorylation of c-Jun N-terminal kinase (JNK) and p38 kinase. Similarly, SLK WT stimulated activator protein-1 reporter activity, but activation of activator protein-1 by the three SLK mutants was ineffective. To test if homodimerization of SLK affects phosphorylation, the cDNA encoding SLK amino acids 1-373 (which include the catalytic domain) was fused with a cDNA for a modified FK506-binding protein, Fv (Fv-SLK 1-373). After transfection, the addition of AP20187 (an FK506 analog) induced regulated dimerization of Fv-SLK 1-373. AP20187-stimulated dimerization enhanced the kinase activity of Fv-SLK 1-373 WT. In contrast, kinase activity of Fv-SLK 1-373 T183A/S189A was weak and was not enhanced after dimerization. Finally, apoptosis was increased after expression of Fv-SLK 1-373 WT but not T183A/S189A. Thus, phosphorylation of Thr-183 and Ser-189 plays a key role in the activation and signaling of SLK and could represent a target for novel therapeutic approaches to renal injury.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Animais , Biocatálise , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Ativação Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mutagênese , Mutação , Fosforilação , Multimerização Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Estrutura Quaternária de Proteína , Ratos , Fator de Transcrição AP-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA