Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 8056, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-38052799

RESUMO

Shear stress generated by urinary fluid flow is an important regulator of renal function. Its dysregulation is observed in various chronic and acute kidney diseases. Previously, we demonstrated that primary cilium-dependent autophagy allows kidney epithelial cells to adapt their metabolism in response to fluid flow. Here, we show that nuclear YAP/TAZ negatively regulates autophagy flux in kidney epithelial cells subjected to fluid flow. This crosstalk is supported by a primary cilium-dependent activation of AMPK and SIRT1, independently of the Hippo pathway. We confirm the relevance of the YAP/TAZ-autophagy molecular dialog in vivo using a zebrafish model of kidney development and a unilateral ureteral obstruction mouse model. In addition, an in vitro assay simulating pathological accelerated flow observed at early stages of chronic kidney disease (CKD) activates YAP, leading to a primary cilium-dependent inhibition of autophagic flux. We confirm this YAP/autophagy relationship in renal biopsies from patients suffering from diabetic kidney disease (DKD), the leading cause of CKD. Our findings demonstrate the importance of YAP/TAZ and autophagy in the translation of fluid flow into cellular and physiological responses. Dysregulation of this pathway is associated with the early onset of CKD.


Assuntos
Insuficiência Renal Crônica , Sirtuína 1 , Animais , Camundongos , Humanos , Sirtuína 1/genética , Proteínas Quinases Ativadas por AMP , Peixe-Zebra , Autofagia/fisiologia , Insuficiência Renal Crônica/genética , Células Epiteliais/fisiologia , Rim
3.
Kidney Int ; 102(1): 78-95, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35337891

RESUMO

Kidney mass and function are sexually determined, but the cellular events and the molecular mechanisms involved in this dimorphism are poorly characterized. By combining female and male mice with castration/replacement experiments, we showed that male mice exhibited kidney overgrowth from five weeks of age. This effect was organ specific, since liver and heart weight were comparable between males and females, regardless of age. Consistently, the androgen receptor was found to be expressed in the kidneys of males, but not in the liver. In growing mice, androgens led to kidney overgrowth by first inducing a burst of cell proliferation and then an increase of cell size. Remarkably, androgens were also required to maintain cell size in adults. In fact, orchiectomy resulted in smaller kidneys in a matter of few weeks. These changes paralleled the changes of the expression of ornithine decarboxylase and cyclin D1, two known mediators of kidney growth, whereas, unexpectedly, mTORC1 and Hippo pathways did not seem to be involved. Androgens also enhanced kidney autophagy, very likely by increasing transcription factor EB nuclear translocation. Functionally, the increase of tubular mass resulted in increased sodium/phosphate transport. These findings were relevant to humans. Remarkably, by studying living gender-paired kidney donors-recipients, we showed that tubular cell size increased three months after transplantation in men as compared to women, regardless of the donor gender. Thus, our results identify novel signaling pathways that may be involved in androgen-induced kidney growth and homeostasis and suggest that androgens determine kidney size after transplantation.


Assuntos
Androgênios , Caracteres Sexuais , Androgênios/farmacologia , Animais , Feminino , Homeostase , Humanos , Rim , Masculino , Camundongos , Tamanho do Órgão
4.
Kidney Int ; 99(3): 632-645, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33137337

RESUMO

Kidney function is crucially dependent on the complex three-dimensional structure of nephrons. Any distortion of their shape may lead to kidney dysfunction. Traditional histological methods present major limitations for three-dimensional tissue reconstruction. Here, we combined tissue clearing, multi-photon microscopy and digital tracing for the reconstruction of single nephrons under physiological and pathological conditions. Sets of nephrons differing in location, shape and size according to their function were identified. Interestingly, nephrons tend to lie in planes. When this technique was applied to a model of cystic kidney disease, cysts were found to develop only in specific nephron segments. Along the same segment, cysts are contiguous within normal non-dilated tubules. Moreover, the shapes of cysts varied according to the nephron segment. Thus, our findings provide a valuable strategy for visualizing the complex structure of kidneys at the single nephron level and, more importantly, provide a basis for understanding pathological processes such as cystogenesis.


Assuntos
Néfrons , Doenças Renais Policísticas , Humanos , Rim , Microscopia
5.
Nat Cell Biol ; 22(9): 1091-1102, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32868900

RESUMO

Organs and cells must adapt to shear stress induced by biological fluids, but how fluid flow contributes to the execution of specific cell programs is poorly understood. Here we show that shear stress favours mitochondrial biogenesis and metabolic reprogramming to ensure energy production and cellular adaptation in kidney epithelial cells. Shear stress stimulates lipophagy, contributing to the production of fatty acids that provide mitochondrial substrates to generate ATP through ß-oxidation. This flow-induced process is dependent on the primary cilia located on the apical side of epithelial cells. The interplay between fluid flow and lipid metabolism was confirmed in vivo using a unilateral ureteral obstruction mouse model. Finally, primary cilium-dependent lipophagy and mitochondrial biogenesis are required to support energy-consuming cellular processes such as glucose reabsorption, gluconeogenesis and cytoskeletal remodelling. Our findings demonstrate how primary cilia and autophagy are involved in the translation of mechanical forces into metabolic adaptation.


Assuntos
Autofagia/fisiologia , Cílios/metabolismo , Cílios/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Rim/metabolismo , Rim/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Linhagem Celular , Gluconeogênese/fisiologia , Glucose/metabolismo , Metabolismo dos Lipídeos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Estresse Mecânico
6.
Basic Res Cardiol ; 115(5): 51, 2020 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-32699940

RESUMO

The concentration of fibroblast growth factor 23 (FGF23) rises progressively in renal failure (RF). High FGF23 concentrations have been consistently associated with adverse cardiovascular outcomes or death, in chronic kidney disease (CKD), heart failure or liver cirrhosis. We identified the mechanisms whereby high concentrations of FGF23 can increase the risk of death of cardiovascular origin. We studied the effects of FGF23 and Klotho in adult rat ventricular cardiomyocytes (ARVMs) and on the heart of mice with CKD. We show that FGF23 increases the frequency of spontaneous calcium waves (SCWs), a marker of cardiomyocyte arrhythmogenicity, in ARVMs. FGF23 increased sarcoplasmic reticulum Ca2+ leakage, basal phosphorylation of Ca2+-cycling proteins including phospholamban and ryanodine receptor type 2. These effects are secondary to a decrease in phosphodiesterase 4B (PDE4B) in ARVMs and in heart of mice with RF. Soluble Klotho, a circulating form of the FGF23 receptor, prevents FGF23 effects on ARVMs by increasing PDE3A and PDE3B expression. Our results suggest that the combination of high FGF23 and low sKlotho concentrations decreases PDE activity in ARVMs, which favors the occurrence of ventricular arrhythmias and may participate in the high death rate observed in patients with CKD.


Assuntos
Arritmias Cardíacas/etiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Arritmias Cardíacas/metabolismo , Sinalização do Cálcio , Cardiomegalia/etiologia , AMP Cíclico/metabolismo , Acoplamento Excitação-Contração , Fator de Crescimento de Fibroblastos 23 , Proteínas Klotho , Masculino , Camundongos , Nefrectomia , Cultura Primária de Células , Ratos Wistar
7.
JCI Insight ; 5(9)2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32376805

RESUMO

The loss of functional nephrons after kidney injury triggers the compensatory growth of the remaining ones to allow functional adaptation. However, in some cases, these compensatory events activate signaling pathways that lead to pathological alterations and chronic kidney disease. Little is known about the identity of these pathways and how they lead to the development of renal lesions. Here, we combined mouse strains that differently react to nephron reduction with molecular and temporal genome-wide transcriptome studies to elucidate the molecular mechanisms involved in these events. We demonstrated that nephron reduction led to 2 waves of cell proliferation: the first one occurred during the compensatory growth regardless of the genetic background, whereas the second one occurred, after a quiescent phase, exclusively in the sensitive strain and accompanied the development of renal lesions. Similarly, clustering by coinertia analysis revealed the existence of 2 waves of gene expression. Interestingly, we identified type I interferon (IFN) response as an early (first-wave) and specific signature of the sensitive (FVB/N) mice. Activation of type I IFN response was associated with G1/S cell cycle arrest, which correlated with p21 nuclear translocation. Remarkably, the transient induction of type I IFN response by poly(I:C) injections during the compensatory growth resulted in renal lesions in otherwise-resistant C57BL6 mice. Collectively, these results suggest that the early molecular and cellular events occurring after nephron reduction determine the risk of developing late renal lesions and point to type I IFN response as a crucial event of the deterioration process.


Assuntos
Rim , Néfrons , Insuficiência Renal Crônica , Transdução de Sinais , Animais , Proliferação de Células , Progressão da Doença , Suscetibilidade a Doenças , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular , Interferon Tipo I/metabolismo , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Endogâmicos C57BL , Néfrons/metabolismo , Néfrons/patologia , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
8.
Sci Rep ; 9(1): 15597, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31666653

RESUMO

MicroRNAs (miRNAs) play an important role in the kidneys under physiological and pathological conditions, but their role in immune glomerulonephritis is unclear. miR-146a has been identified as a key player in innate immunity and inflammatory responses, and in the kidney, this miRNA is involved in the response of injured tubular cells. We studied the renal and immune phenotypes of miR-146a+/+ and miR-146a-/- mice at 12 months of age, and the results showed that miR-146a-/- mice developed autoimmunity during aging, as demonstrated by circulating antibodies targeting double-stranded DNA and an immune complex-mediated glomerulonephritis associated with a mild renal immune infiltrate. In addition, miR-146a-/- mice showed reduced expression of the transmembrane protein Kim1/Tim1, a key regulator of regulatory B cell (Breg) homeostasis, in the kidney and the immune cells. The numbers of memory B cells and plasmablasts were increased in miR-146a-/- mice compared with the numbers in wild-type mice, whereas Bregs were decreased in number and displayed an altered capacity to produce IL-10. Finally, we showed that miR-146a-/- mice develop an autoimmune syndrome with increasing age, and this syndrome includes immune complex glomerulonephritis, which might be due to altered B cell responses associated with Kim1/Tim1 deficiency. This study unravels a link between miR-146a and Kim1 and identifies miR-146a as a significant player in immune-mediated glomerulonephritis pathogenesis.


Assuntos
Complexo Antígeno-Anticorpo/imunologia , Glomerulonefrite/genética , Glomerulonefrite/imunologia , MicroRNAs/metabolismo , Animais , Linfócitos B Reguladores/metabolismo , Biomarcadores/metabolismo , Regulação da Expressão Gênica , Glomerulonefrite/metabolismo , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Homeostase , Camundongos , MicroRNAs/genética , Fenótipo
9.
BMC Genomics ; 19(1): 558, 2018 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-30060733

RESUMO

After the publication of this work [1], a mistake was noticed in the Eq. 1. Given an m × n expression matrix with m genes and samples of n tissues, the correct definition of the Gini index for gene i is.

10.
PLoS Genet ; 13(12): e1007093, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29240767

RESUMO

Congenital nephron number varies widely in the human population and individuals with low nephron number are at risk of developing hypertension and chronic kidney disease. The development of the kidney occurs via an orchestrated morphogenetic process where metanephric mesenchyme and ureteric bud reciprocally interact to induce nephron formation. The genetic networks that modulate the extent of this process and set the final nephron number are mostly unknown. Here, we identified a specific isoform of MITF (MITF-A), a bHLH-Zip transcription factor, as a novel regulator of the final nephron number. We showed that overexpression of MITF-A leads to a substantial increase of nephron number and bigger kidneys, whereas Mitfa deficiency results in reduced nephron number. Furthermore, we demonstrated that MITF-A triggers ureteric bud branching, a phenotype that is associated with increased ureteric bud cell proliferation. Molecular studies associated with an in silico analyses revealed that amongst the putative MITF-A targets, Ret was significantly modulated by MITF-A. Consistent with the key role of this network in kidney morphogenesis, Ret heterozygosis prevented the increase of nephron number in mice overexpressing MITF-A. Collectively, these results uncover a novel transcriptional network that controls branching morphogenesis during kidney development and identifies one of the first modifier genes of nephron endowment.


Assuntos
Rim/fisiologia , Fator de Transcrição Associado à Microftalmia/metabolismo , Néfrons/fisiologia , Animais , Feminino , Humanos , Rim/embriologia , Rim/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fator de Transcrição Associado à Microftalmia/genética , Morfogênese , Néfrons/anatomia & histologia , Néfrons/crescimento & desenvolvimento , Néfrons/metabolismo , Organogênese , Isoformas de Proteínas , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas c-ret/metabolismo , Ureter/metabolismo , Ureter/fisiologia
11.
Sci Rep ; 7(1): 1474, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28469279

RESUMO

Glycosylation is critical for the regulation of several cellular processes. One glycosylation pathway, the unusual O-linked ß-N-acetylglucosamine glycosylation (O-GlcNAcylation) has been shown to be required for proper mitosis, likely through a subset of proteins that are O-GlcNAcylated during metaphase. As lectins bind glycosylated proteins, we asked if specific lectins interact with mitotic O-GlcNAcylated proteins during metaphase to ensure correct cell division. Galectin-3, a small soluble lectin of the Galectin family, is an excellent candidate, as it has been previously described as a transient centrosomal component in interphase and mitotic epithelial cells. In addition, it has recently been shown to associate with basal bodies in motile cilia, where it stabilizes the microtubule-organizing center (MTOC). Using an experimental mouse model of chronic kidney disease and human epithelial cell lines, we investigate the role of Galectin-3 in dividing epithelial cells. Here we find that Galectin-3 is essential for metaphase where it associates with NuMA in an O-GlcNAcylation-dependent manner. We provide evidence that the NuMA-Galectin-3 interaction is important for mitotic spindle cohesion and for stable NuMA localization to the spindle pole, thus revealing that Galectin-3 is a novel contributor to epithelial mitotic progress.


Assuntos
Acetilglucosamina/metabolismo , Antígenos Nucleares/metabolismo , Células Epiteliais/metabolismo , Galectina 3/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Processamento de Proteína Pós-Traducional , Insuficiência Renal Crônica/metabolismo , Polos do Fuso/metabolismo , Animais , Antígenos Nucleares/genética , Proteínas Sanguíneas , Proteínas de Ciclo Celular , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/citologia , Galectina 3/genética , Galectinas , Glicosilação , Humanos , Interfase , Metáfase , Camundongos , Camundongos Knockout , Proteínas Associadas à Matriz Nuclear/genética , Ligação Proteica , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Polos do Fuso/ultraestrutura
12.
BMC Genomics ; 18(1): 277, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28376718

RESUMO

BACKGROUND: Gene expression data can be compromised by cells originating from other tissues than the target tissue of profiling. Failures in detecting such tissue heterogeneity have profound implications on data interpretation and reproducibility. A computational tool explicitly addressing the issue is warranted. RESULTS: We introduce BioQC, a R/Bioconductor software package to detect tissue heterogeneity in gene expression data. To this end BioQC implements a computationally efficient Wilcoxon-Mann-Whitney test and provides more than 150 signatures of tissue-enriched genes derived from large-scale transcriptomics studies. Simulation experiments show that BioQC is both fast and sensitive in detecting tissue heterogeneity. In a case study with whole-organ profiling data, BioQC predicted contamination events that are confirmed by quantitative RT-PCR. Applied to transcriptomics data of the Genotype-Tissue Expression (GTEx) project, BioQC reveals clustering of samples and suggests that some samples likely suffer from tissue heterogeneity. CONCLUSIONS: Our experience with gene expression data indicates a prevalence of tissue heterogeneity that often goes unnoticed. BioQC addresses the issue by integrating prior knowledge with a scalable algorithm. We propose BioQC as a first-line tool to ensure quality and reproducibility of gene expression data.


Assuntos
Perfilação da Expressão Gênica , Software , Algoritmos , Animais , Cães , Humanos , Camundongos , Especificidade de Órgãos , Reprodutibilidade dos Testes , Transcriptoma
13.
J Am Soc Nephrol ; 28(7): 2144-2157, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28270414

RESUMO

Mammalian target of rapamycin (mTOR) signaling is involved in a variety of kidney diseases. Clinical trials administering mTOR inhibitors to patients with FSGS, a prototypic podocyte disease, led to conflicting results, ranging from remission to deterioration of kidney function. Here, we combined complex genetic titration of mTOR complex 1 (mTORC1) levels in murine glomerular disease models, pharmacologic studies, and human studies to precisely delineate the role of mTOR in FSGS. mTORC1 target genes were significantly induced in microdissected glomeruli from both patients with FSGS and a murine FSGS model. Furthermore, a mouse model with constitutive mTORC1 activation closely recapitulated human FSGS. Notably, the complete knockout of mTORC1 by induced deletion of both Raptor alleles accelerated the progression of murine FSGS models. However, lowering mTORC1 signaling by deleting just one Raptor allele ameliorated the progression of glomerulosclerosis. Similarly, low-dose treatment with the mTORC1 inhibitor rapamycin efficiently diminished disease progression. Mechanistically, complete pharmacologic inhibition of mTOR in immortalized podocytes shifted the cellular energy metabolism toward reduced rates of oxidative phosphorylation and anaerobic glycolysis, which correlated with increased production of reactive oxygen species. Together, these data suggest that podocyte injury and loss is commonly followed by adaptive mTOR activation. Prolonged mTOR activation, however, results in a metabolic podocyte reprogramming leading to increased cellular stress and dedifferentiation, thus offering a treatment rationale for incomplete mTOR inhibition.


Assuntos
Glomerulosclerose Segmentar e Focal/etiologia , Glomerulosclerose Segmentar e Focal/prevenção & controle , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/fisiologia , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/fisiologia , Animais , Progressão da Doença , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos
14.
Nat Cell Biol ; 18(6): 657-67, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27214279

RESUMO

Autophagy is an adaptation mechanism that is vital for cellular homeostasis in response to various stress conditions. Previous reports indicate that there is a functional interaction between the primary cilium (PC) and autophagy. The PC, a microtubule-based structure present at the surface of numerous cell types, is a mechanical sensor. Here we show that autophagy induced by fluid flow regulates kidney epithelial cell volume in vitro and in vivo. PC ablation blocked autophagy induction and cell-volume regulation. In addition, inhibition of autophagy in ciliated cells impaired the flow-dependent regulation of cell volume. PC-dependent autophagy can be triggered either by mTOR inhibition or a mechanism dependent on the polycystin 2 channel. Only the LKB1-AMPK-mTOR signalling pathway was required for the flow-dependent regulation of cell volume by autophagy. These findings suggest that therapies regulating autophagy should be considered in developing treatments for PC-related diseases.


Assuntos
Autofagia , Fenômenos Fisiológicos Celulares , Cílios/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Tamanho Celular , Cães , Immunoblotting , Células Madin Darby de Rim Canino , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais , Serina-Treonina Quinases TOR/fisiologia
15.
J Am Soc Nephrol ; 27(12): 3690-3705, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27153926

RESUMO

In CKD, tubular cells may be involved in the induction of interstitial fibrosis, which in turn, leads to loss of renal function. However, the molecular mechanisms that link tubular cells to the interstitial compartment are not clear. Activation of the Stat3 transcription factor has been reported in tubular cells after renal damage, and Stat3 has been implicated in CKD progression. Here, we combined an experimental model of nephron reduction in mice from different genetic backgrounds and genetically modified animals with in silico and in vitro experiments to determine whether the selective activation of Stat3 in tubular cells is involved in the development of interstitial fibrosis. Nephron reduction caused Stat3 phosphorylation in tubular cells of lesion-prone mice but not in resistant mice. Furthermore, specific deletion of Stat3 in tubular cells significantly reduced the extent of interstitial fibrosis, which correlated with reduced fibroblast proliferation and matrix synthesis, after nephron reduction. Mechanistically, in vitro tubular Stat3 activation triggered the expression of a specific subset of paracrine profibrotic factors, including Lcn2, Pdgfb, and Timp1. Together, our results provide a molecular link between tubular and interstitial cells during CKD progression and identify Stat3 as a central regulator of this link and a promising therapeutic target.


Assuntos
Comunicação Celular , Túbulos Renais/citologia , Insuficiência Renal Crônica/fisiopatologia , Fator de Transcrição STAT3/fisiologia , Animais , Feminino , Camundongos
16.
Nat Commun ; 7: 10330, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26787103

RESUMO

In chronic kidney disease (CKD), proteinuria results in severe tubulointerstitial lesions, which ultimately lead to end-stage renal disease. Here we identify 4-phenylbutyric acid (PBA), a chemical chaperone already used in humans, as a novel therapeutic strategy capable to counteract the toxic effect of proteinuria. Mechanistically, we show that albumin induces tubular unfolded protein response via cytosolic calcium rise, which leads to tubular apoptosis by Lipocalin 2 (LCN2) modulation through ATF4. Consistent with the key role of LCN2 in CKD progression, Lcn2 gene inactivation decreases ER stress-induced apoptosis, tubulointerstitial lesions and mortality in proteinuric mice. More importantly, the inhibition of this pathway by PBA protects kidneys from morphological and functional degradation in proteinuric mice. These results are relevant to human CKD, as LCN2 is increased in proteinuric patients. In conclusion, our study identifies a therapeutic strategy susceptible to improve the benefit of RAS inhibitors in proteinuria-induced CKD progression.


Assuntos
Proteínas de Fase Aguda/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Nefropatias/etiologia , Nefropatias/metabolismo , Lipocalinas/metabolismo , Proteínas Oncogênicas/metabolismo , Proteinúria/complicações , Proteinúria/metabolismo , Proteínas de Fase Aguda/genética , Albuminas/farmacologia , Animais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Éxons/genética , Feminino , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipocalina-2 , Lipocalinas/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Mutantes , Proteínas Oncogênicas/genética , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Proteínas WT1/genética , Proteínas WT1/metabolismo
17.
Nephron ; 129(1): 42-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25531116

RESUMO

Cystic kidney disease is characterized by the progressive development of multiple fluid-filled cysts. Cysts can be acquired, or they may appear during development or in postnatal life due to specific gene defects and lead to renal failure. The most frequent form of this disease is the inherited polycystic kidney disease (PKD). Experimental models of PKD showed that an increase of cellular proliferation and apoptosis as well as defects in apico-basal and planar cell polarity or cilia play a critical role in cyst development. However, little is known about the mechanisms and the mediators involved in acquired cystic kidney diseases (ACKD). In this study, we used the nephron reduction as a model to study the mechanisms underlying cyst development in ACKD. We found that tubular dilations after nephron reduction recapitulated most of the morphological features of ACKD. The development of tubular dilations was associated with a dramatic increase of cell proliferation. In contrast, the apico-basal polarity and cilia did not seem to be affected. Interestingly, polycystin 1 and fibrocystin were markedly increased and polycystin 2 was decreased in cells lining the dilated tubules, whereas the expression of several other cystic genes did not change. More importantly, Pkd1 haploinsufficiency accelerated the development of tubular dilations after nephron reduction, a phenotype that was associated to a further increase of cell proliferation. These data were relevant to humans ACKD, as cystic genes expression and the rate of cell proliferation were also increased. In conclusion, our study suggests that the nephron reduction can be considered a suitable model to study ACKD and that dosage of genes involved in PKD is also important in ACKD.


Assuntos
Dosagem de Genes , Néfrons/patologia , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Canais de Cátion TRPP/genética , Adulto , Idoso , Animais , Polaridade Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Haploinsuficiência , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Nefrectomia , Doenças Renais Policísticas/etiologia , Rim Policístico Autossômico Recessivo/genética , Rim Policístico Autossômico Recessivo/patologia , Receptores de Superfície Celular/genética , Canais de Cátion TRPP/deficiência
18.
Nat Med ; 19(10): 1288-96, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24056770

RESUMO

In chronic kidney disease (CKD), loss of functional nephrons results in metabolic and mechanical stress in the remaining ones, resulting in further nephron loss. Here we show that Akt2 activation has an essential role in podocyte protection after nephron reduction. Glomerulosclerosis and albuminuria were substantially worsened in Akt2(-/-) but not in Akt1(-/-) mice as compared to wild-type mice. Specific deletion of Akt2 or its regulator Rictor in podocytes revealed that Akt2 has an intrinsic function in podocytes. Mechanistically, Akt2 triggers a compensatory program that involves mouse double minute 2 homolog (Mdm2), glycogen synthase kinase 3 (Gsk3) and Rac1. The defective activation of this pathway after nephron reduction leads to apoptosis and foot process effacement of the podocytes. We further show that AKT2 activation by mammalian target of rapamycin complex 2 (mTORC2) is also required for podocyte survival in human CKD. More notably, we elucidate the events underlying the adverse renal effect of sirolimus and provide a criterion for the rational use of this drug. Thus, our results disclose a new function of Akt2 and identify a potential therapeutic target for preserving glomerular function in CKD.


Assuntos
Falência Renal Crônica/metabolismo , Podócitos/citologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Animais , Progressão da Doença , Humanos , Falência Renal Crônica/patologia , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/fisiologia , Néfrons/metabolismo , Néfrons/fisiopatologia , Proteínas Proto-Oncogênicas c-akt/genética , Serina-Treonina Quinases TOR/fisiologia
19.
EMBO Mol Med ; 4(8): 825-39, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22711280

RESUMO

The molecular networks that control the progression of chronic kidney diseases (CKD) are poorly defined. We have recently shown that the susceptibility to development of renal lesions after nephron reduction is controlled by a locus on mouse chromosome 6 and requires epidermal growth factor receptor (EGFR) activation. Here, we identified microphthalmia-associated transcription factor A (MITF-A), a bHLH-Zip transcription factor, as a modifier of CKD progression. Sequence analysis revealed a strain-specific mutation in the 5' UTR that decreases MITF-A protein synthesis in lesion-prone friend virus B NIH (FVB/N) mice. More importantly, we dissected the molecular pathway by which MITF-A modulates CKD progression. MITF-A interacts with histone deacetylases to repress the transcription of TGF-α, a ligand of EGFR, and antagonizes transactivation by its related partner, transcription factor E3 (TFE3). Consistent with the key role of this network in CKD, Tgfa gene inactivation protected FVB/N mice from renal deterioration after nephron reduction. These data are relevant to human CKD, as we found that the TFE3/MITF-A ratio was increased in patients with damaged kidneys. Our study uncovers a novel transcriptional network and unveils novel potential prognostic and therapeutic targets for preventing human CKD progression.


Assuntos
Redes Reguladoras de Genes , Predisposição Genética para Doença , Nefropatias/genética , Fator de Transcrição Associado à Microftalmia/genética , Animais , Doença Crônica , Progressão da Doença , Feminino , Humanos , Masculino , Camundongos , Modelos Biológicos
20.
J Am Soc Nephrol ; 22(2): 327-35, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21183591

RESUMO

The mechanisms of progression of chronic kidney disease (CKD) are poorly understood. Epidemiologic studies suggest a strong genetic component, but the genes that contribute to the onset and progression of CKD are largely unknown. Here, we applied an experimental model of CKD (75% excision of total renal mass) to six different strains of mice and found that only the FVB/N strain developed renal lesions. We performed a genome-scan analysis in mice generated by back-crossing resistant and sensitive strains; we identified a major susceptibility locus (Ckdp1) on chromosome 6, which corresponds to regions on human chromosome 2 and 3 that link with CKD progression. In silico analysis revealed that the locus includes the gene encoding the EGF receptor (EGFR) ligand TGF-α. TGF-α protein levels markedly increased after nephron reduction exclusively in FVB/N mice, and this increase preceded the development of renal lesions. Furthermore, pharmacologic inhibition of EGFR prevented the development of renal lesions in the sensitive FVB/N strain. These data suggest that variable TGF-α expression may explain, in part, the genetic susceptibility to CKD progression. EGFR inhibition may be a therapeutic strategy to counteract the genetic predisposition to CKD.


Assuntos
Predisposição Genética para Doença , Nefropatias/genética , Fator de Crescimento Transformador alfa/fisiologia , Animais , Mapeamento Cromossômico , Doença Crônica , Nefropatias/etiologia , Falência Renal Crônica/etiologia , Falência Renal Crônica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Mutação , Néfrons/patologia , Fator de Crescimento Transformador alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...