Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Am J Physiol Renal Physiol ; 326(1): F20-F29, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37916289

RESUMO

We have previously shown that the long-acting ß2-adrenergic receptor (ß2-AR) agonist formoterol induced recovery from acute kidney injury in mice. To determine whether formoterol protected against diabetic nephropathy, the most common cause of end-stage kidney disease (ESKD), we used a high-fat diet (HFD), a murine type 2 diabetes model, and streptozotocin, a murine type 1 diabetes model. Following formoterol treatment, there was a marked recovery from and reversal of diabetic nephropathy in HFD mice compared with those treated with vehicle alone at the ultrastructural, histological, and functional levels. Similar results were seen after formoterol treatment in mice receiving streptozotocin. To investigate effects in humans, we performed a competing risk regression analysis with death as a competing risk to examine the association between Veterans with chronic kidney disease (CKD) and chronic obstructive pulmonary disease (COPD), who use ß2-AR agonists, and Veterans with CKD but no COPD, and progression to ESKD in a large national cohort of Veterans with stage 4 CKD between 2011 and 2013. Veterans were followed until 2016 or death. ESKD was defined as the initiation of dialysis and/or receipt of kidney transplant. We found that COPD was associated with a 25.6% reduction in progression from stage 4 CKD to ESKD compared with no COPD after adjusting for age, diabetes, sex, race-ethnicity, comorbidities, and medication use. Sensitivity analysis showed a 33.2% reduction in ESKD in Veterans with COPD taking long-acting formoterol and a 20.8% reduction in ESKD in Veterans taking other ß2-AR agonists compared with those with no COPD. These data indicate that ß2-AR agonists, especially formoterol, could be a treatment for diabetic nephropathy and perhaps other forms of CKD.NEW & NOTEWORTHY Diabetic nephropathy is the most common cause of ESKD. Formoterol, a long-acting ß2-adrenergic receptor (ß2-AR) agonist, reversed diabetic nephropathy in murine models of type 1 and 2 diabetes. In humans, there was an association with protection from progression of CKD in patients with COPD, by means of ß2-AR agonist intake, compared with those without COPD. These data indicate that ß2-AR agonists, especially formoterol, could be a new treatment for diabetic nephropathy and other forms of CKD.


Assuntos
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Nefropatias Diabéticas , Falência Renal Crônica , Doença Pulmonar Obstrutiva Crônica , Humanos , Animais , Camundongos , Nefropatias Diabéticas/tratamento farmacológico , Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Estreptozocina , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Fumarato de Formoterol/uso terapêutico , Falência Renal Crônica/tratamento farmacológico , Falência Renal Crônica/etiologia , Receptores Adrenérgicos/uso terapêutico
2.
J Biol Chem ; 297(3): 101079, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34391780

RESUMO

Phosphorylation (activation) and dephosphorylation (deactivation) of the slit diaphragm proteins NEPHRIN and NEPH1 are critical for maintaining the kidney epithelial podocyte actin cytoskeleton and, therefore, proper glomerular filtration. However, the mechanisms underlying these events remain largely unknown. Here we show that NEPHRIN and NEPH1 are novel receptor proteins for hepatocyte growth factor (HGF) and can be phosphorylated independently of the mesenchymal epithelial transition receptor in a ligand-dependent fashion through engagement of their extracellular domains by HGF. Furthermore, we demonstrate SH2 domain-containing protein tyrosine phosphatase-2-dependent dephosphorylation of these proteins. To establish HGF as a ligand, purified baculovirus-expressed NEPHRIN and NEPH1 recombinant proteins were used in surface plasma resonance binding experiments. We report high-affinity interactions of NEPHRIN and NEPH1 with HGF, although NEPHRIN binding was 20-fold higher than that of NEPH1. In addition, using molecular modeling we constructed peptides that were used to map specific HGF-binding regions in the extracellular domains of NEPHRIN and NEPH1. Finally, using an in vitro model of cultured podocytes and an ex vivo model of Drosophila nephrocytes, as well as chemically induced injury models, we demonstrated that HGF-induced phosphorylation of NEPHRIN and NEPH1 is centrally involved in podocyte repair. Taken together, this is the first study demonstrating a receptor-based function for NEPHRIN and NEPH1. This has important biological and clinical implications for the repair of injured podocytes and the maintenance of podocyte integrity.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Proteínas de Membrana/metabolismo , Animais , Linhagem Celular , Taxa de Filtração Glomerular/fisiologia , Fator de Crescimento de Hepatócito/fisiologia , Humanos , Junções Intercelulares/metabolismo , Rim/patologia , Glomérulos Renais/metabolismo , Proteínas de Membrana/genética , Camundongos , Peptídeos/metabolismo , Fosforilação , Podócitos/metabolismo , Ligação Proteica/fisiologia , Transdução de Sinais/fisiologia
3.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G1044-G1053, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33908271

RESUMO

Myosin 1c (Myo1c) is an unconventional myosin that modulates signaling pathways involved in tissue injury and repair. In this study, we observed that Myo1c expression is significantly upregulated in human chronic liver disease such as nonalcoholic steatohepatitis (NASH) and in animal models of liver fibrosis. High throughput data from the GEO-database identified similar Myo1c upregulation in mice and human liver fibrosis. Notably, transforming growth factor-ß1 (TGF-ß1) stimulation to hepatic stellate cells (HSCs), the liver pericyte and key cell type responsible for the deposition of extracellular matrix, upregulates Myo1c expression, whereas genetic depletion or pharmacological inhibition of Myo1c blunted TGF-ß-induced fibrogenic responses, resulting in repression of α-smooth muscle actin (α-SMA) and collagen type I α 1 chain (Col1α1) mRNA. Myo1c deletion also decreased fibrogenic processes such as cell proliferation, wound healing response, and contractility when compared with vehicle-treated HSCs. Importantly, phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) and mothers against decapentaplegic homolog 3 (SMAD3) were significantly blunted upon Myo1c inhibition in GRX cells as well as Myo1c knockout (Myo1c-KO) mouse embryonic fibroblasts (MEFs) upon TGF-ß stimulation. Using the genetic Myo1c-KO mice, we confirmed that Myo1c is critical for fibrogenesis, as Myo1c-KO mice were resistant to carbon tetrachloride (CCl4)-induced liver fibrosis. Histological and immunostaining analysis of liver sections showed that deposition of collagen fibers and α-SMA expression were significantly reduced in Myo1c-KO mice upon liver injury. Collectively, these results demonstrate that Myo1c mediates hepatic fibrogenesis by modulating TGF-ß signaling and suggest that inhibiting this process may have clinical application in treating liver fibrosis.NEW & NOTEWORTHY The incidences of liver fibrosis are growing at a rapid pace and have become one of the leading causes of end-stage liver disease. Although TGF-ß1 is known to play a prominent role in transforming cells to produce excessive extracellular matrix that lead to hepatic fibrosis, the therapies targeting TGF-ß1 have achieved very limited clinical impact. This study highlights motor protein myosin-1c-mediated mechanisms that serve as novel regulators of TGF-ß1 signaling and fibrosis.


Assuntos
Fibroblastos/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Miosina Tipo I/metabolismo , Animais , Cadeia alfa 1 do Colágeno Tipo I , Fibroblastos/patologia , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Camundongos , Miosina Tipo I/genética , Fosforilação , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
4.
J Biol Chem ; 294(26): 10104-10119, 2019 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-31073028

RESUMO

Although the slit diaphragm proteins in podocytes are uniquely organized to maintain glomerular filtration assembly and function, little is known about the underlying mechanisms that participate in trafficking these proteins to the correct location for development and homeostasis. Identifying these mechanisms will likely provide novel targets for therapeutic intervention to preserve podocyte function following glomerular injury. Analysis of structural variation in cases of human nephrotic syndrome identified rare heterozygous deletions of EXOC4 in two patients. This suggested that disruption of the highly-conserved eight-protein exocyst trafficking complex could have a role in podocyte dysfunction. Indeed, mRNA profiling of injured podocytes identified significant exocyst down-regulation. To test the hypothesis that the exocyst is centrally involved in podocyte development/function, we generated homozygous podocyte-specific Exoc5 (a central exocyst component that interacts with Exoc4) knockout mice that showed massive proteinuria and died within 4 weeks of birth. Histological and ultrastructural analysis of these mice showed severe glomerular defects with increased fibrosis, proteinaceous casts, effaced podocytes, and loss of the slit diaphragm. Immunofluorescence analysis revealed that Neph1 and Nephrin, major slit diaphragm constituents, were mislocalized and/or lost. mRNA profiling of Exoc5 knockdown podocytes showed that vesicular trafficking was the most affected cellular event. Mapping of signaling pathways and Western blot analysis revealed significant up-regulation of the mitogen-activated protein kinase and transforming growth factor-ß pathways in Exoc5 knockdown podocytes and in the glomeruli of podocyte-specific Exoc5 KO mice. Based on these data, we propose that exocyst-based mechanisms regulate Neph1 and Nephrin signaling and trafficking, and thus podocyte development and function.


Assuntos
Deleção de Genes , Glomérulos Renais/patologia , Síndrome Nefrótica/patologia , Podócitos/patologia , Proteínas de Transporte Vesicular/fisiologia , Animais , Apoptose , Movimento Celular , Exocitose , Humanos , Glomérulos Renais/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndrome Nefrótica/genética , Fosforilação , Podócitos/metabolismo , Transporte Proteico , Proteinúria/etiologia , Proteinúria/patologia , Transdução de Sinais
5.
Kidney Int ; 95(3): 708-716, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30709661

RESUMO

Definitive diagnosis of glomerular disease requires a kidney biopsy, an invasive procedure that may not be safe or feasible to perform in all patients. We developed a noninvasive, accurate, and economical diagnostic assay with easy commercial adaptability to detect recurrent focal segmental glomerulosclerosis (rFSGS) after kidney transplant. Since FSGS involves podocyte damage and death, our approach involved mRNA profiling of cultured podocytes treated with plasma from patients with rFSGS to identify upregulated genes involved in podocyte damage. For concept validation, three upregulated pro-apoptotic candidate genes (IL1ß, BMF, and IGFBP3) were selected, and their promoter regions were cloned into a luciferase-based reporter vector and transfected into podocytes to generate stable podocyte cell lines. Strikingly, when exposed to rFSGS patient plasma, these cell lines showed increased reporter activity; in contrast, no reporter activity was noted with plasma from patients with non-recurrent FSGS or membranous nephropathy. Area under the receiver operating characteristics curves (AUCs) for models discriminating between rFSGS and other nephropathies (non-recurrent FSGS and membranous nephropathy) and between rFSGS and non-recurrent FSGS ranged from 0.81 to 0.86, respectively. Estimated sensitivities and specificities for the diagnosis of rFSGS were greater than 80% for the IL1ß and BMF cell lines, and were slightly lower for the IGFBP3 cell line. Importantly, the novel approach outlined here for the diagnosis of rFSGS is widely applicable to the design of sensitive and specific diagnostic/prognostic assays for other glomerular diseases.


Assuntos
Bioensaio/métodos , Glomerulosclerose Segmentar e Focal/diagnóstico , Podócitos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Diagnóstico Diferencial , Perfilação da Expressão Gênica , Genes Reporter , Glomerulosclerose Segmentar e Focal/sangue , Glomerulosclerose Segmentar e Focal/complicações , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Falência Renal Crônica/etiologia , Falência Renal Crônica/cirurgia , Transplante de Rim , Luciferases/genética , Plasma/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/metabolismo , RNA-Seq , Curva ROC , Recidiva
6.
Kidney Int ; 96(1): 139-158, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31097328

RESUMO

Transforming growth factor-ß (TGF-ß) is known to play a critical role in the pathogenesis of many progressive podocyte diseases. However, the molecular mechanisms regulating TGF-ß signaling in podocytes remain unclear. Using a podocyte-specific myosin (Myo)1c knockout, we demonstrate whether Myo1c is critical for TGF-ß-signaling in podocyte disease pathogenesis. Specifically, podocyte-specific Myo1c knockout mice were resistant to fibrotic injury induced by Adriamycin or nephrotoxic serum. Further, loss of Myo1c also protected from injury in the TGF-ß-dependent unilateral ureteral obstruction mouse model of renal interstitial fibrosis. Mechanistic analyses showed that loss of Myo1c significantly blunted TGF-ß signaling through downregulation of canonical and non-canonical TGF-ß pathways. Interestingly, nuclear rather than the cytoplasmic Myo1c was found to play a central role in controlling TGF-ß signaling through transcriptional regulation. Differential expression analysis of nuclear Myo1c-associated gene promoters showed that nuclear Myo1c targeted the TGF-ß responsive gene growth differentiation factor (GDF)-15 and directly bound to the GDF-15 promoter. Importantly, GDF15 was found to be involved in podocyte pathogenesis, where GDF15 was upregulated in glomeruli of patients with focal segmental glomerulosclerosis. Thus, Myo1c-mediated regulation of TGF-ß-responsive genes is central to the pathogenesis of podocyte injury. Hence, inhibiting this process may have clinical application in treating podocytopathies.


Assuntos
Fator 15 de Diferenciação de Crescimento/genética , Nefropatias/patologia , Miosina Tipo I/metabolismo , Podócitos/patologia , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/metabolismo , Animais , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Feminino , Fibrose , Regulação da Expressão Gênica , Humanos , Nefropatias/induzido quimicamente , Masculino , Camundongos , Camundongos Knockout , Miosina Tipo I/genética , Podócitos/efeitos dos fármacos , Regiões Promotoras Genéticas , Transcrição Gênica
7.
Kidney Int ; 96(3): 656-673, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31262488

RESUMO

Podocytes have limited ability to recover from injury. Here, we demonstrate that increased mitochondrial biogenesis, to meet the metabolic and energy demand of a cell, accelerates podocyte recovery from injury. Analysis of events induced during podocyte injury and recovery showed marked upregulation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a transcriptional co-activator of mitochondrial biogenesis, and key components of the mitochondrial electron transport chain. To evaluate our hypothesis that increasing mitochondrial biogenesis enhanced podocyte recovery from injury, we treated injured podocytes with formoterol, a potent, specific, and long-acting ß2-adrenergic receptor agonist that induces mitochondrial biogenesis in vitro and in vivo. Formoterol increased mitochondrial biogenesis and restored mitochondrial morphology and the injury-induced changes to the organization of the actin cytoskeleton in podocytes. Importantly, ß2-adrenergic receptors were found to be present on podocyte membranes. Their knockdown attenuated formoterol-induced mitochondrial biogenesis. To determine the potential clinical relevance of these findings, mouse models of acute nephrotoxic serum nephritis and chronic (Adriamycin [doxorubicin]) glomerulopathy were used. Mice were treated with formoterol post-injury when glomerular dysfunction was established. Strikingly, formoterol accelerated the recovery of glomerular function by reducing proteinuria and ameliorating kidney pathology. Furthermore, formoterol treatment reduced cellular apoptosis and increased the expression of the mitochondrial biogenesis marker PGC-1α and multiple electron transport chain proteins. Thus, our results support ß2-adrenergic receptors as novel therapeutic targets and formoterol as a therapeutic compound for treating podocytopathies.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Fumarato de Formoterol/farmacologia , Glomerulonefrite/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Podócitos/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Fumarato de Formoterol/uso terapêutico , Técnicas de Silenciamento de Genes , Glomerulonefrite/induzido quimicamente , Glomerulonefrite/patologia , Humanos , Camundongos , Mitocôndrias/metabolismo , Biogênese de Organelas , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Podócitos/citologia , Podócitos/patologia , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais
8.
Kidney Int ; 96(4): 883-889, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31472902

RESUMO

Steroid-resistant nephrotic syndrome is a frequent cause of chronic kidney disease almost inevitably progressing to end-stage renal disease. More than 58 monogenic causes of SRNS have been discovered and majority of known steroid-resistant nephrotic syndrome causing genes are predominantly expressed in glomerular podocytes, placing them at the center of disease pathogenesis. Herein, we describe two unrelated families with steroid-resistant nephrotic syndrome with homozygous mutations in the KIRREL1 gene. One mutation showed high frequency in the European population (minor allele frequency 0.0011) and this patient achieved complete remission following treatment, but later progressed to chronic kidney disease. We found that mutant KIRREL1 proteins failed to localize to the podocyte cell membrane, indicating defective trafficking and impaired podocytes function. Thus, the KIRREL1 gene product has an important role in modulating the integrity of the slit diaphragm and maintaining glomerular filtration function.


Assuntos
Resistência a Medicamentos/genética , Glucocorticoides/farmacologia , Proteínas de Membrana/genética , Síndrome Nefrótica/genética , Insuficiência Renal Crônica/genética , Adolescente , Idade de Início , Linhagem Celular , Criança , Pré-Escolar , Consanguinidade , Análise Mutacional de DNA , Progressão da Doença , Feminino , Seguimentos , Frequência do Gene , Membrana Basal Glomerular/patologia , Membrana Basal Glomerular/ultraestrutura , Glucocorticoides/uso terapêutico , Homozigoto , Humanos , Masculino , Proteínas de Membrana/metabolismo , Microscopia Eletrônica de Transmissão , Mutação , Síndrome Nefrótica/tratamento farmacológico , Síndrome Nefrótica/patologia , Linhagem , Podócitos , Insuficiência Renal Crônica/patologia , Sequenciamento do Exoma
9.
Nephrology (Carlton) ; 24(5): 497-503, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30848004

RESUMO

Beta2-adrenergic receptor (ß2 -AR) is a G-protein-coupled adrenergic receptor family member, whose clinical significance has been extensively investigated in lung, cardiovascular and muscular diseases, but its role in kidney biology remains understudied. In this review, we discuss some of the recent studies, where the effect of agonist/antagonist-mediated activation/inhibition of ß2 -AR on disease pathogenesis process was studied, and highlighted the role of ß2 -AR in kidney biology. The expression of ß2 -AR has been noted in many kidney subunits including proximal tubules, glomeruli and podocytes. In vivo studies have shown that in cultured proximal tubules ß2 -AR is involved in Na-ATPase activity and transcellular Na-transport through protein kinase-C activation; whereas in cultured podocytes, it was associated with depolarization of the membrane. The animal studies further revealed that ß2 -AR activation by short-acting ß2 agonists attenuated monocyte activation, pro-inflammatory and pro-fibrotic responses through ß-arrestin2 dependent NF-kB inactivation in diabetic kidney disease; in contrast, activation by long-acting ß2 agonists restored mitochondrial and renal function in the acute kidney injury mice models through PGC-1α dependent mitochondrial biogenesis. In conclusion, the activation of ß2 -AR may present a rapidly developing therapeutic target for renal diseases.


Assuntos
Injúria Renal Aguda/metabolismo , Nefropatias Diabéticas/metabolismo , Rim/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/patologia , Injúria Renal Aguda/fisiopatologia , Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Animais , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Fármacos Renais/uso terapêutico , Transdução de Sinais
10.
Int J Mol Sci ; 21(1)2019 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-31906131

RESUMO

Podocytes have a unique structure that supports glomerular filtration function, and many glomerular diseases result in loss of this structure, leading to podocyte dysfunction and ESRD (end stage renal disease). These structural and functional changes involve a complex set of molecular and cellular mechanisms that remain poorly understood. To understand the molecular signature of podocyte injury, we performed transcriptome analysis of cultured human podocytes injured either with PAN (puromycin aminonucleoside) or doxorubicin/adriamycin (ADR). The pathway analysis through DE (differential expression) and gene-enrichment analysis of the injured podocytes showed Tumor protein p53 (P53) as one of the major signaling pathways that was significantly upregulated upon podocyte injury. Accordingly, P53 expression was also up-regulated in the glomeruli of nephrotoxic serum (NTS) and ADR-injured mice. To further confirm these observations, cultured podocytes were treated with the P53 inhibitor pifithrin-α, which showed significant protection from ADR-induced actin cytoskeleton damage. In conclusion, signaling pathways that are involved in podocyte pathogenesis and can be therapeutically targeted were identified by high-throughput transcriptomic analysis of injured podocytes.


Assuntos
Doxorrubicina/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Nefropatias/metabolismo , Podócitos/metabolismo , Puromicina Aminonucleosídeo/efeitos adversos , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Animais , Doxorrubicina/farmacologia , Humanos , Nefropatias/induzido quimicamente , Nefropatias/patologia , Camundongos , Fosforilação/efeitos dos fármacos , Podócitos/patologia , Puromicina Aminonucleosídeo/farmacologia
11.
Am J Physiol Renal Physiol ; 314(2): F280-F292, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29046299

RESUMO

Podocyte dysfunction and loss is an early event and a hallmark of proteinuric kidney diseases. A podocyte's normal function is maintained via its unique cellular architecture that relies on an intracellular network of filaments, including filamentous actin (F-actin) and microtubules, that provides mechanical support. Damage to this filamentous network leads to changes in cellular morphology and results in podocyte injury, dysfunction, and death. Conversely, stabilization of this network protects podocytes and ameliorates proteinuria. This suggests that stabilization of podocyte architecture via its filamentous network could be a key therapeutic strategy for proteinuric kidney diseases. However, development of podocyte-directed therapeutics, especially those that target the cell's filamentous network, is still lacking, partly because of unavailability of appropriate cellular assays for use in a drug discovery environment. Here, we describe a new high-content screening-based methodology and its implementation on podocytes to identify paullone derivatives as a novel group of podocyte-protective compounds. We find that three compounds, i.e., kenpaullone, 1-azakenpaullone, and alsterpaullone, dose dependently protect podocytes from puromycin aminonucleoside (PAN)-mediated injury in vitro by reducing PAN-induced changes in both the filamentous actin and microtubules, with alsterpaullone providing maximal protection. Mechanistic studies further show that alsterpaullone suppressed PAN-induced activation of signaling downstream of GSK3ß and p38 mitogen-activated protein kinase. In vivo it reduced ADR-induced glomerular injury in a zebrafish model. Together, these results identify paullone derivatives as novel podocyte-protective agents for future therapeutic development.


Assuntos
Benzazepinas/farmacologia , Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala , Indóis/farmacologia , Podócitos/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Fármacos Renais/farmacologia , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Doxorrubicina , Glicogênio Sintase Quinase 3 beta/metabolismo , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Nefropatias/patologia , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Microtúbulos/patologia , Podócitos/metabolismo , Podócitos/patologia , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
J Am Soc Nephrol ; 28(7): 2119-2132, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28202497

RESUMO

Aminopeptidase A (APA) is expressed in glomerular podocytes and tubular epithelia and metabolizes angiotensin II (AngII), a peptide known to promote glomerulosclerosis. In this study, we tested whether APA expression changes in response to progressive nephron loss or whether APA exerts a protective role against glomerular damage and during AngII-mediated hypertensive kidney injury. At advanced stages of FSGS, fawn-hooded hypertensive rat kidneys exhibited distinctly increased APA staining in areas of intact glomerular capillary loops. Moreover, BALB/c APA-knockout (KO) mice injected with a nephrotoxic serum showed persistent glomerular hyalinosis and albuminuria 96 hours after injection, whereas wild-type controls achieved virtually full recovery. We then tested the effect of 4-week infusion of AngII (400 ng/kg per minute) in APA-KO and wild-type mice. Although we observed no significant difference in achieved systolic BP, AngII-treated APA-KO mice developed a significant rise in albuminuria not observed in AngII-treated wild-type mice along with increased segmental and global sclerosis and/or collapse of juxtamedullary glomeruli, microcystic tubular dilation, and tubulointerstitial fibrosis. In parallel, AngII treatment significantly increased the kidney AngII content and attenuated the expression of podocyte nephrin in APA-KO mice but not in wild-type controls. These data show that deficiency of APA increases susceptibility to glomerular injury in BALB/c mice. The augmented AngII-mediated kidney injury observed in association with increased intrarenal AngII accumulation in the absence of APA suggests a protective metabolizing role of APA in AngII-mediated glomerular diseases.


Assuntos
Glutamil Aminopeptidase/deficiência , Nefropatias/enzimologia , Nefropatias/etiologia , Glomérulos Renais , Animais , Suscetibilidade a Doenças , Masculino , Camundongos , Camundongos Knockout , Ratos
13.
J Biol Chem ; 289(14): 9502-18, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24554715

RESUMO

Podocytes are specialized epithelial cells that are critical components of the glomerular filtration barrier, and their dysfunction leads to proteinuria and renal failure. Therefore, preserving podocyte function is therapeutically significant. In this study, we identified Neph1 signaling as a therapeutic target that upon inhibition prevented podocyte damage from a glomerular injury-inducing agent puromycin aminonucleoside (PAN). To specifically inhibit Neph1 signaling, we used a protein transduction approach, where the cytoplasmic domain of Neph1 (Neph1CD) tagged with a protein transduction domain trans-activator of transcription was transduced in cultured podocytes prior to treatment with PAN. The PAN-induced Neph1 phosphorylation was significantly reduced in Neph1CD-transduced cells; in addition, these cells were resistant to PAN-induced cytoskeletal damage. The biochemical analysis using subfractionation studies showed that unlike control cells Neph1 was retained in the lipid raft fractions in the transduced cells following treatment with PAN, indicating that transduction of Neph1CD in podocytes prevented PAN-induced mislocalization of Neph1. In accordance, the immunofluorescence analysis further suggested that Neph1CD-transduced cells had increased ability to retain endogenous Neph1 at the membrane in response to PAN-induced injury. Similar results were obtained when angiotensin was used as an injury-inducing agent. Consistent with these observations, maintaining high levels of Neph1 at the membrane using a podocyte cell line overexpressing chimeric Neph1 increased the ability of podocytes to resist PAN-induced injury and PAN-induced albumin leakage. Using a zebrafish in vivo PAN and adriamycin injury models, we further demonstrated the ability of transduced Neph1CD to preserve glomerular function. Collectively, these results support the conclusion that inhibiting Neph1 signaling is therapeutically significant in preventing podocyte damage from glomerular injury.


Assuntos
Membrana Basal Glomerular/lesões , Membrana Basal Glomerular/metabolismo , Proteínas de Membrana/metabolismo , Podócitos/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Antimetabólitos Antineoplásicos/efeitos adversos , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular , Membrana Basal Glomerular/patologia , Humanos , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/patologia , Proteínas de Membrana/genética , Fosforilação/genética , Podócitos/patologia , Puromicina Aminonucleosídeo/efeitos adversos , Puromicina Aminonucleosídeo/farmacologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
14.
Pharmacol Rep ; 76(3): 612-621, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38668812

RESUMO

BACKGROUND: Podocytes have a remarkable ability to recover from injury; however, little is known about the recovery mechanisms involved in this process. We recently showed that formoterol, a long-acting ß2-adrenergic receptor (ß2-AR) agonist, induced mitochondrial biogenesis (MB) in podocytes and led to renoprotection in mice. However, it is not clear whether this effect was mediated by formoterol acting through the ß2-AR or if it occurred through "off-target" effects. METHODS: We genetically deleted the ß2-AR specifically in murine podocytes and used these mice to determine whether formoterol acting through the podocyte ß2-AR alone is sufficient for recovery of renal filtration function following injury. The podocyte-specific ß2-AR knockout mice (ß2-ARfl/fl/PodCre) were generated by crossing ß2-AR floxed mice with podocin Cre (B6.Cg-Tg(NPHS2-cre)295Lbh/J) mice. These mice were then subjected to both acute and chronic glomerular injury using nephrotoxic serum (NTS) and adriamycin (ADR), respectively. The extent of injury was evaluated by measuring albuminuria and histological and immunostaining analysis of the murine kidney sections. RESULTS: A similar level of injury was observed in ß2-AR knockout and control mice; however, the ß2-ARfl/fl/PodCre mice failed to recover in response to formoterol. Functional evaluation of the ß2-ARfl/fl/PodCre mice following injury plus formoterol showed similar albuminuria and glomerular injury to control mice that were not treated with formoterol. CONCLUSIONS: These results indicate that the podocyte ß2-AR is a critical component of the recovery mechanism and may serve as a novel therapeutic target for treating podocytopathies.


Assuntos
Doxorrubicina , Fumarato de Formoterol , Camundongos Knockout , Podócitos , Receptores Adrenérgicos beta 2 , Animais , Camundongos , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Albuminúria/metabolismo , Doxorrubicina/farmacologia , Doxorrubicina/toxicidade , Fumarato de Formoterol/farmacologia , Camundongos Endogâmicos C57BL , Podócitos/metabolismo , Podócitos/efeitos dos fármacos , Podócitos/patologia , Receptores Adrenérgicos beta 2/metabolismo
15.
J Biol Chem ; 287(12): 9441-53, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22262837

RESUMO

Neph1 is present in podocytes, where it plays a critical role in maintaining the filtration function of the glomerulus, in part through signaling events mediated by its cytoplasmic domain that are involved in actin cytoskeleton organization. To understand the function of this protein, a detailed knowledge of the structure of the Neph1 cytoplasmic domain (Neph1-CD) is required. In this study, the solution structure of this domain was determined by small/wide angle x-ray scattering (SWAXS). Analysis of Neph1-CD by SWAXS suggested that this protein adopts a global shape with a radius of gyration and a maximum linear dimension of 21.3 and 70 Å, respectively. These parameters and the data from circular dichroism experiments were used to construct a structural model of this protein. The His-ZO-1-PDZ1 (first PDZ domain of zonula occludens) domain that binds Neph1-CD was also analyzed by SWAXS, to confirm that it adopts a global structure similar to its crystal structure. We used the SWAXS intensity profile, the structural model of Neph1-CD, and the crystal structure of ZO-1-PDZ1 to construct a structural model of the Neph1-CD·ZO-1-PDZ1 complex. Mapping of the intermolecular interactions suggested that in addition to the C-terminal residues Thr-His-Val, residues Lys-761 and Tyr-762 in Neph1 are also critical for stabilizing the complex. Estimated intensity values from the SWAXS data and in vivo and in vitro pull-down experiments demonstrated loss of binding to ZO-1 when these residues were individually mutated to alanines. Our findings present a structural model that provides novel insights into the molecular structure and function of Neph1-CD.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Podócitos/metabolismo , Sítios de Ligação , Proteínas de Membrana/genética , Estrutura Molecular , Domínios PDZ , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Podócitos/química , Ligação Proteica , Estrutura Secundária de Proteína , Espalhamento a Baixo Ângulo , Proteína da Zônula de Oclusão-1
16.
Kidney Int ; 84(6): 1154-65, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23715127

RESUMO

The targeting and organization of podocyte slit diaphragm proteins nephrin and neph1 is critical for development and maintenance of a functional glomerular filtration barrier. Myo1c is a non-muscle myosin motor protein that interacts directly with nephrin and neph1, and mediates their intracellular transport to the podocyte intercellular junction. Here we investigated the necessity of Myo1c in podocyte development using zebrafish as a model system. Immunofluorescence microscopy and in situ RNA hybridization analysis of zebrafish embryos showed that Myo1c is widely expressed in various tissues including the zebrafish glomerulus. Knockdown of the Myo1c gene in zebrafish using antisense morpholino derivatives resulted in an abnormal developmental phenotype that included pericardial edema and dilated renal tubules. Ultrastructural analysis of the glomerulus in Myo1c-depleted zebrafish showed abnormal podocyte morphology and absence of the slit diaphragm. Consistent with these observations, the glomerular filter permeability appeared altered in zebrafish in which Myo1c expression was attenuated. The specificity of Myo1c knockdown was confirmed by a rescue experiment in which co-injection of Myo1c morpholino derivatives with orthologous Myo1c mRNA prepared from mouse cDNA lessened phenotypic abnormalities including edema in Myo1c morphants. Thus, our results demonstrate that Myo1c is necessary for podocyte morphogenesis.


Assuntos
Glomérulos Renais/metabolismo , Miosina Tipo I/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Edema/genética , Edema/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Silenciamento de Genes , Genótipo , Barreira de Filtração Glomerular/metabolismo , Taxa de Filtração Glomerular , Glomérulos Renais/embriologia , Camundongos , Morfogênese , Morfolinos/metabolismo , Miosina Tipo I/genética , Permeabilidade , Fenótipo , Podócitos/metabolismo , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
18.
Cells ; 10(6)2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34073294

RESUMO

Unconventional myosins, linked to deafness, are also proposed to play a role in retinal cell physiology. However, their direct role in photoreceptor function remains unclear. We demonstrate that systemic loss of the unconventional myosin MYO1C in mice, specifically causes rhodopsin mislocalization, leading to impaired visual function. Electroretinogram analysis of Myo1c knockout (Myo1c-KO) mice showed a progressive loss of photoreceptor function. Immunohistochemistry and binding assays demonstrated MYO1C localization to photoreceptor inner and outer segments (OS) and identified a direct interaction of rhodopsin with MYO1C. In Myo1c-KO retinas, rhodopsin mislocalized to rod inner segments (IS) and cell bodies, while cone opsins in OS showed punctate staining. In aged mice, the histological and ultrastructural examination of the phenotype of Myo1c-KO retinas showed progressively shorter photoreceptor OS. These results demonstrate that MYO1C is important for rhodopsin localization to the photoreceptor OS, and for normal visual function.


Assuntos
Proteínas do Olho/metabolismo , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Rodopsina/metabolismo , Animais , Dineínas/genética , Eletrorretinografia/métodos , Camundongos , Fenótipo , Rodopsina/genética
19.
Biochem Biophys Res Commun ; 396(4): 950-5, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20460105

RESUMO

BMP-SMAD (bone morphogenetic protein) signaling pathways in association with APT play paramount roles in osteoblastic differentiation, bone formation and embryonic development of human and animals. However, the implications of potent components (BMP6, Smad1, Smad2 and APT) of this pathway in SCD (sickle cell disease) pathology with orthopedic complications (Ortho+SS) are poorly elucidated and substantially unknown. Here, we address the role of BMP6, Smad1, Smad2 and APT mRNA and protein expression in hMDDCs obtained from Ortho+SS patients, employing RT-PCR, qRT-PCR and immunoblotting. Interestingly, we observed that SCD pathology exhibited significantly up-regulated expression of those signaling components at the level of mRNA and protein. Furthermore, exogenous BMP6 induced apoptosis was observed to be significantly associated in Ortho+SS complication and markedly increased the percentage of cells undergoing apoptosis as compared to healthy group. Interestingly, the non-stimulated cells have shown higher apoptotic nuclei percentage than the stimulated cells in pathological condition. Thus, expression of BMP-SMAD signaling components augments apoptosis and up regulates the transcription of these genes and it suggests that induction is due to transcriptional regulation. Taken together, our findings provide evidence that BMP-SMAD signaling components along with APT were over expressed, mediates apoptosis and may play an important role in the SCD pathology with orthopedic complications.


Assuntos
Anemia Falciforme/metabolismo , Doenças Ósseas/metabolismo , Proteína Morfogenética Óssea 6/metabolismo , Células Dendríticas/metabolismo , Artropatias/metabolismo , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Anemia Falciforme/complicações , Apoptose , Doenças Ósseas/etiologia , Proteína Morfogenética Óssea 6/genética , Humanos , Artropatias/etiologia , Monócitos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Smad1/genética , Proteína Smad2/genética
20.
Genes (Basel) ; 11(2)2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-32045989

RESUMO

Sphingolipids are bioactive lipids associated with cellular membranes and plasma lipoproteins, and their synthesis and degradation are tightly regulated. We have previously determined that low plasma concentrations of certain ceramide species predict the development of nephropathy in diabetes patients with normal albumin excretion rates at baseline. Herein, we tested the hypothesis that altering the sphingolipid content of circulating lipoproteins can alter the metabolic and signaling pathways in podocytes, whose dysfunction leads to an impairment of glomerular filtration. Cultured human podocytes were treated with lipoproteins from healthy subjects enriched in vitro with C16 ceramide, or D-erythro 2-hydroxy C16 ceramide, a ceramide naturally found in skin. The RNA-Seq data demonstrated differential expression of genes regulating sphingolipid metabolism, sphingolipid signaling, and mTOR signaling pathways. A multiplex analysis of mTOR signaling pathway intermediates showed that the majority (eight) of the pathway phosphorylated proteins measured (eleven) were significantly downregulated in response to C16 ceramide-enriched HDL2 compared to HDL2 alone and hydroxy ceramide-enriched HDL2. In contrast, C16 ceramide-enriched HDL3 upregulated the phosphorylation of four intermediates in the mTOR pathway. These findings highlight a possible role for lipoprotein-associated sphingolipids in regulating metabolic and signaling pathways in podocytes and could lead to novel therapeutic targets in glomerular kidney diseases.


Assuntos
Ceramidas/metabolismo , Lipoproteínas/farmacologia , Podócitos/metabolismo , Esfingolipídeos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Transcriptoma/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Isótopos de Carbono , Linhagem Celular , Ceramidas/genética , HDL-Colesterol/farmacologia , Adesões Focais/efeitos dos fármacos , Adesões Focais/genética , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Fosforilação , Podócitos/efeitos dos fármacos , RNA-Seq , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Esfingolipídeos/genética , Serina-Treonina Quinases TOR/genética , Transcriptoma/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA