Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Commun ; 10(1): 2461, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31165727

RESUMO

Epigenetic changes might provide the biological explanation for the long-lasting impact of metabolic alterations of diabetic kidney disease development. Here we examined cytosine methylation of human kidney tubules using Illumina Infinium 450 K arrays from 91 subjects with and without diabetes and varying degrees of kidney disease using a cross-sectional design. We identify cytosine methylation changes associated with kidney structural damage and build a model for kidney function decline. We find that the methylation levels of 65 probes are associated with the degree of kidney fibrosis at genome wide significance. In total 471 probes improve the model for kidney function decline. Methylation probes associated with kidney damage and functional decline enrich on kidney regulatory regions and associate with gene expression changes, including epidermal growth factor (EGF). Altogether, our work shows that kidney methylation differences can be detected in patients with diabetic kidney disease and improve kidney function decline models indicating that they are potentially functionally important.


Assuntos
Citosina/metabolismo , Metilação de DNA , Nefropatias Diabéticas/genética , Rim/metabolismo , Idoso , Estudos de Casos e Controles , Ilhas de CpG , Estudos Transversais , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Progressão da Doença , Epigênese Genética , Feminino , Fibrose , Regulação da Expressão Gênica , Taxa de Filtração Glomerular , Humanos , Rim/patologia , Masculino , Pessoa de Meia-Idade
2.
EBioMedicine ; 24: 267-276, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28970079

RESUMO

Chronic kidney disease (CKD) has diverse phenotypic manifestations including structural (such as fibrosis) and functional (such as glomerular filtration rate and albuminuria) alterations. Gene expression profiling has recently gained popularity as an important new tool for precision medicine approaches. Here we used unbiased and directed approaches to understand how gene expression captures different CKD manifestations in patients with diabetic and hypertensive CKD. Transcriptome data from ninety-five microdissected human kidney samples with a range of demographics, functional and structural changes were used for the primary analysis. Data obtained from 41 samples were available for validation. Using the unbiased Weighted Gene Co-Expression Network Analysis (WGCNA) we identified 16 co-expressed gene modules. We found that modules that strongly correlated with eGFR primarily encoded genes with metabolic functions. Gene groups that mainly encoded T-cell receptor and collagen pathways, showed the strongest correlation with fibrosis level, suggesting that these two phenotypic manifestations might have different underlying mechanisms. Linear regression models were then used to identify genes whose expression showed significant correlation with either structural (fibrosis) or functional (eGFR) manifestation and mostly corroborated the WGCNA findings. We concluded that gene expression is a very sensitive sensor of fibrosis, as the expression of 1654 genes correlated with fibrosis even after adjusting to eGFR and other clinical parameters. The association between GFR and gene expression was mostly mediated by fibrosis. In conclusion, our transcriptome-based CKD trait dissection analysis suggests that the association between gene expression and renal function is mediated by structural changes and that there may be differences in pathways that lead to decline in kidney function and the development of fibrosis, respectively.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Perfilação da Expressão Gênica/métodos , Hipertensão/complicações , Túbulos Renais/química , Insuficiência Renal Crônica/genética , Idoso , Diabetes Mellitus Tipo 2/genética , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Taxa de Filtração Glomerular , Humanos , Hipertensão/genética , Túbulos Renais/patologia , Túbulos Renais/fisiopatologia , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Análise de Regressão , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia
3.
Nat Med ; 23(4): 429-438, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28218918

RESUMO

African Americans have a heightened risk of developing chronic and end-stage kidney disease, an association that is largely attributed to two common genetic variants, termed G1 and G2, in the APOL1 gene. Direct evidence demonstrating that these APOL1 risk alleles are pathogenic is still lacking because the APOL1 gene is present in only some primates and humans; thus it has been challenging to demonstrate experimental proof of causality of these risk alleles for renal disease. Here we generated mice with podocyte-specific inducible expression of the APOL1 reference allele (termed G0) or each of the risk-conferring alleles (G1 or G2). We show that mice with podocyte-specific expression of either APOL1 risk allele, but not of the G0 allele, develop functional (albuminuria and azotemia), structural (foot-process effacement and glomerulosclerosis) and molecular (gene-expression) changes that closely resemble human kidney disease. Disease development was cell-type specific and likely reversible, and the severity correlated with the level of expression of the risk allele. We further found that expression of the risk-variant APOL1 alleles interferes with endosomal trafficking and blocks autophagic flux, which ultimately leads to inflammatory-mediated podocyte death and glomerular scarring. In summary, this is the first demonstration that the expression of APOL1 risk alleles is causal for altered podocyte function and glomerular disease in vivo.


Assuntos
Apolipoproteínas/genética , Glomérulos Renais/metabolismo , Lipoproteínas HDL/genética , Podócitos/metabolismo , Insuficiência Renal Crônica/genética , Albuminúria/genética , Alelos , Animais , Apolipoproteína L1 , Autofagia/genética , Azotemia/genética , Western Blotting , Endocitose/genética , Endossomos/metabolismo , Imunofluorescência , Predisposição Genética para Doença , Variação Genética , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/patologia , Células HEK293 , Células HeLa , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Glomérulos Renais/patologia , Glomérulos Renais/ultraestrutura , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Podócitos/ultraestrutura , Insuficiência Renal Crônica/patologia
4.
J Biol Chem ; 292(3): 837-846, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-27909050

RESUMO

Clear cell renal cell carcinoma (CCRCC) is an incurable malignancy in advanced stages and needs newer therapeutic targets. Transcriptomic analysis of CCRCCs and matched microdissected renal tubular controls revealed overexpression of NOTCH ligands and receptors in tumor tissues. Examination of the TCGA RNA-seq data set also revealed widespread activation of NOTCH pathway in a large cohort of CCRCC samples. Samples with NOTCH pathway activation were also clinically distinct and were associated with better overall survival. Parallel DNA methylation and copy number analysis demonstrated that both genetic and epigenetic alterations led to NOTCH pathway activation in CCRCC. NOTCH ligand JAGGED1 was overexpressed and associated with loss of CpG methylation of H3K4me1-associated enhancer regions. JAGGED2 was also overexpressed and associated with gene amplification in distinct CCRCC samples. Transgenic expression of intracellular NOTCH1 in mice with tubule-specific deletion of VHL led to dysplastic hyperproliferation of tubular epithelial cells, confirming the procarcinogenic role of NOTCH in vivo Alteration of cell cycle pathways was seen in murine renal tubular cells with NOTCH overexpression, and molecular similarity to human tumors was observed, demonstrating that human CCRCC recapitulates features and gene expression changes observed in mice with transgenic overexpression of the Notch intracellular domain. Treatment with the γ-secretase inhibitor LY3039478 led to inhibition of CCRCC cells in vitro and in vivo In summary, these data reveal the mechanistic basis of NOTCH pathway activation in CCRCC and demonstrate this pathway to a potential therapeutic target.


Assuntos
Neoplasias Renais/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Carcinoma de Células Renais , Ilhas de CpG , Metilação de DNA , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Feminino , Humanos , Proteína Jagged-1/genética , Proteína Jagged-1/metabolismo , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Neoplasias Renais/patologia , Masculino , Camundongos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Inibidores de Proteases/farmacologia , Receptor Notch1/genética
5.
Cell Rep ; 12(4): 636-47, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26190114

RESUMO

Diabetic nephropathy is a major cause of end-stage kidney disease. Characterized by progressive microvascular disease, most efforts have focused on injury to the glomerular endothelium. Recent work has suggested a role for the podocyte, a highly specialized component of the glomerular filtration barrier. Here, we demonstrate that the Drosophila nephrocyte, a cell analogous to the mammalian podocyte, displays defects that phenocopy aspects of diabetic nephropathy in animals fed chronic high dietary sucrose. Through functional studies, we identify an OGT-Polycomb-Knot-Sns pathway that links dietary sucrose to loss of the Nephrin ortholog Sns. Reducing OGT through genetic or drug means is sufficient to rescue loss of Sns, leading to overall extension of lifespan. We demonstrate upregulation of the Knot ortholog EBF2 in glomeruli of human diabetic nephropathy patients and a mouse ob/ob diabetes model. Furthermore, we demonstrate rescue of Nephrin expression and cell viability in ebf2(-/-) primary podocytes cultured in high glucose.


Assuntos
Nefropatias Diabéticas/metabolismo , Podócitos/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/patologia , Carboidratos da Dieta/efeitos adversos , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Humanos , Imunoglobulinas/genética , Imunoglobulinas/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Podócitos/patologia , Podócitos/fisiologia , Sacarose/toxicidade
6.
Nat Med ; 21(1): 37-46, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25419705

RESUMO

Renal fibrosis is the histological manifestation of a progressive, usually irreversible process causing chronic and end-stage kidney disease. We performed genome-wide transcriptome studies of a large cohort (n = 95) of normal and fibrotic human kidney tubule samples followed by systems and network analyses and identified inflammation and metabolism as the top dysregulated pathways in the diseased kidneys. In particular, we found that humans and mouse models with tubulointerstitial fibrosis had lower expression of key enzymes and regulators of fatty acid oxidation (FAO) and higher intracellular lipid deposition compared to controls. In vitro experiments indicated that inhibition of FAO in tubule epithelial cells caused ATP depletion, cell death, dedifferentiation and intracellular lipid deposition, phenotypes observed in fibrosis. In contrast, restoring fatty acid metabolism by genetic or pharmacological methods protected mice from tubulointerstitial fibrosis. Our results raise the possibility that correcting the metabolic defect in FAO may be useful for preventing and treating chronic kidney disease.


Assuntos
Ácidos Graxos/metabolismo , Fibrose/genética , Inflamação/genética , Nefropatias/genética , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Ácidos Graxos/genética , Fibrose/metabolismo , Fibrose/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Inflamação/metabolismo , Inflamação/patologia , Nefropatias/metabolismo , Nefropatias/patologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Camundongos , Oxirredução , Transdução de Sinais/genética
7.
J Am Soc Nephrol ; 26(3): 692-714, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25231882

RESUMO

Genome-wide association studies (GWASs) have identified multiple loci associated with the risk of CKD. Almost all risk variants are localized to the noncoding region of the genome; therefore, the role of these variants in CKD development is largely unknown. We hypothesized that polymorphisms alter transcription factor binding, thereby influencing the expression of nearby genes. Here, we examined the regulation of transcripts in the vicinity of CKD-associated polymorphisms in control and diseased human kidney samples and used systems biology approaches to identify potentially causal genes for prioritization. We interrogated the expression and regulation of 226 transcripts in the vicinity of 44 single nucleotide polymorphisms using RNA sequencing and gene expression arrays from 95 microdissected control and diseased tubule samples and 51 glomerular samples. Gene expression analysis from 41 tubule samples served for external validation. 92 transcripts in the tubule compartment and 34 transcripts in glomeruli showed statistically significant correlation with eGFR. Many novel genes, including ACSM2A/2B, FAM47E, and PLXDC1, were identified. We observed that the expression of multiple genes in the vicinity of any single CKD risk allele correlated with renal function, potentially indicating that genetic variants influence multiple transcripts. Network analysis of GFR-correlating transcripts highlighted two major clusters; a positive correlation with epithelial and vascular functions and an inverse correlation with inflammatory gene cluster. In summary, our functional genomics analysis highlighted novel genes and critical pathways associated with kidney function for future analysis.


Assuntos
Estudo de Associação Genômica Ampla , Insuficiência Renal Crônica/genética , Estudos de Casos e Controles , Humanos , Inflamação/metabolismo , Glomérulos Renais/metabolismo , Polimorfismo Genético , Insuficiência Renal Crônica/metabolismo , Uromodulina/genética , Urotélio/metabolismo
8.
Clin Cancer Res ; 20(16): 4349-60, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24916699

RESUMO

PURPOSE: Even though recent studies have shown that genetic changes at enhancers can influence carcinogenesis, most methylomic studies have focused on changes at promoters. We used renal cell carcinoma (RCC), an incurable malignancy associated with mutations in epigenetic regulators, as a model to study genome-wide patterns of DNA methylation at a high resolution. EXPERIMENTAL DESIGN: Analysis of cytosine methylation status of 1.3 million CpGs was determined by the HELP assay in RCC and healthy microdissected renal tubular controls. RESULTS: We observed that the RCC samples were characterized by widespread hypermethylation that preferentially affected gene bodies. Aberrant methylation was particularly enriched in kidney-specific enhancer regions associated with H3K4Me1 marks. Various important underexpressed genes, such as SMAD6, were associated with aberrantly methylated, intronic enhancers, and these changes were validated in an independent cohort. MOTIF analysis of aberrantly hypermethylated regions revealed enrichment for binding sites of AP2a, AHR, HAIRY, ARNT, and HIF1 transcription factors, reflecting contributions of dysregulated hypoxia signaling pathways in RCC. The functional importance of this aberrant hypermethylation was demonstrated by selective sensitivity of RCC cells to low levels of decitabine. Most importantly, methylation of enhancers was predictive of adverse prognosis in 405 cases of RCC in multivariate analysis. In addition, parallel copy-number analysis from MspI representations demonstrated novel copy-number variations that were validated in an independent cohort of patients. CONCLUSIONS: Our study is the first high-resolution methylome analysis of RCC, demonstrates that many kidney-specific enhancers are targeted by aberrant hypermethylation, and reveals the prognostic importance of these epigenetic changes in an independent cohort.


Assuntos
Carcinoma de Células Renais/genética , Carcinoma de Células Renais/mortalidade , Metilação de DNA , Elementos Facilitadores Genéticos/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/genética , Neoplasias Renais/mortalidade , Biomarcadores Tumorais/genética , Estudos de Casos e Controles , Células Cultivadas , Estudos de Coortes , Epigênese Genética/genética , Humanos , Especificidade de Órgãos , Prognóstico , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida
9.
Genome Biol ; 14(10): R108, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098934

RESUMO

BACKGROUND: One in eleven people is affected by chronic kidney disease, a condition characterized by kidney fibrosis and progressive loss of kidney function. Epidemiological studies indicate that adverse intrauterine and postnatal environments have a long-lasting role in chronic kidney disease development. Epigenetic information represents a plausible carrier for mediating this programming effect. Here we demonstrate that genome-wide cytosine methylation patterns of healthy and chronic kidney disease tubule samples obtained from patients show significant differences. RESULTS: We identify differentially methylated regions and validate these in a large replication dataset. The differentially methylated regions are rarely observed on promoters, but mostly overlap with putative enhancer regions, and they are enriched in consensus binding sequences for important renal transcription factors. This indicates their importance in gene expression regulation. A core set of genes that are known to be related to kidney fibrosis, including genes encoding collagens, show cytosine methylation changes correlating with downstream transcript levels. CONCLUSIONS: Our report raises the possibility that epigenetic dysregulation plays a role in chronic kidney disease development via influencing core pro-fibrotic pathways and can aid the development of novel biomarkers and future therapeutics.


Assuntos
Citosina/metabolismo , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Rim/metabolismo , Rim/patologia , Idoso , Sequência de Bases , Sítios de Ligação , Estudos de Casos e Controles , Análise por Conglomerados , Metilação de DNA , Fibrose , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Humanos , Pessoa de Meia-Idade , Motivos de Nucleotídeos , Matrizes de Pontuação de Posição Específica , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/patologia , Reprodutibilidade dos Testes , Fatores de Transcrição , Transcrição Gênica/efeitos dos fármacos
10.
J Am Soc Nephrol ; 24(2): 268-82, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23334396

RESUMO

Low levels of the adipocyte-secreted protein adiponectin correlate with albuminuria in both mice and humans, but whether adiponectin has a causative role in modulating renal disease is unknown. Here, we first generated a mouse model that allows induction of caspase-8-mediated apoptosis specifically in podocytes upon injection of a construct-specific agent. These POD-ATTAC mice exhibited significant kidney damage, mimicking aspects of human renal disease, such as foot process effacement, mesangial expansion, and glomerulosclerosis. After the initial induction, both podocytes and filtration function recovered. Next, we crossed POD-ATTAC mice with mice lacking or overexpressing adiponectin. POD-ATTAC mice lacking adiponectin developed irreversible albuminuria and renal failure; conversely, POD-ATTAC mice overexpressing adiponectin recovered more rapidly and exhibited less interstitial fibrosis. In conclusion, these results suggest that adiponectin is a renoprotective protein after podocyte injury. Furthermore, the POD-ATTAC mouse provides a platform for further studies, allowing precise timing of podocyte injury and regeneration.


Assuntos
Adiponectina/farmacologia , Podócitos/fisiologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Insuficiência Renal Crônica/tratamento farmacológico , Acetatos/farmacologia , Adiponectina/genética , Albuminúria/tratamento farmacológico , Albuminúria/patologia , Albuminúria/fisiopatologia , Animais , Apoptose/fisiologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Fibrose/tratamento farmacológico , Fibrose/patologia , Fibrose/fisiopatologia , Barreira de Filtração Glomerular/efeitos dos fármacos , Barreira de Filtração Glomerular/patologia , Barreira de Filtração Glomerular/fisiologia , Humanos , Indóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , PPAR gama/antagonistas & inibidores , Podócitos/patologia , Insuficiência Renal Crônica/patologia , Insuficiência Renal Crônica/fisiopatologia
11.
Diabetes ; 60(9): 2354-69, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21752957

RESUMO

OBJECTIVE: Diabetic kidney disease (DKD) is the single leading cause of kidney failure in the U.S., for which a cure has not yet been found. The aim of our study was to provide an unbiased catalog of gene-expression changes in human diabetic kidney biopsy samples. RESEARCH DESIGN AND METHODS: Affymetrix expression arrays were used to identify differentially regulated transcripts in 44 microdissected human kidney samples. DKD samples were significant for their racial diversity and decreased glomerular filtration rate (~25-35 mL/min). Stringent statistical analysis, using the Benjamini-Hochberg corrected two-tailed t test, was used to identify differentially expressed transcripts in control and diseased glomeruli and tubuli. Two different web-based algorithms were used to define differentially regulated pathways. RESULTS: We identified 1,700 differentially expressed probesets in DKD glomeruli and 1,831 in diabetic tubuli, and 330 probesets were commonly differentially expressed in both compartments. Pathway analysis highlighted the regulation of Ras homolog gene family member A, Cdc42, integrin, integrin-linked kinase, and vascular endothelial growth factor signaling in DKD glomeruli. The tubulointerstitial compartment showed strong enrichment for inflammation-related pathways. The canonical complement signaling pathway was determined to be statistically differentially regulated in both DKD glomeruli and tubuli and was associated with increased glomerulosclerosis even in a different set of DKD samples. CONCLUSIONS: Our studies have cataloged gene-expression regulation and identified multiple novel genes and pathways that may play a role in the pathogenesis of DKD or could serve as biomarkers.


Assuntos
Nefropatias Diabéticas/genética , Perfilação da Expressão Gênica , Glomérulos Renais/metabolismo , Adulto , Idoso , Estudos Transversais , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Glomérulos Renais/patologia , Masculino , Pessoa de Meia-Idade
12.
Kidney Int ; 79(8): 802-4, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21451535

RESUMO

Insufficient insulin secretion and insulin resistance are hallmarks of diabetes. Recent studies indicate that insulin plays an important role in maintaining the glomerular filtration barrier. Mima et al. report that glomeruli of diabetic and obese rats suffer from insulin resistance and altered insulin signaling. Protein kinase C inhibitors are able to overcome insulin resistance, offering new hopes for the treatment of the condition.


Assuntos
Nefropatias Diabéticas/fisiopatologia , Insulina/fisiologia , Glomérulos Renais/fisiopatologia , Animais , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/etiologia , Modelos Animais de Doenças , Humanos , Resistência à Insulina , Proteína Quinase C/antagonistas & inibidores , Ratos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA