Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
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
2.
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
3.
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
4.
Cancer Res ; 63(21): 7284-90, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14612525

RESUMO

P-Glycoprotein (P-gp) encoded by the MDR gene is one of the main factors in multidrug resistance. Its expression in cancer cells, which compromises cancer outcome, can be enhanced by some stress signals. Energy depletion, frequently observed in malignant cells, was shown to induce chemoresistance and could be one of these signals. To test this hypothesis, we studied the effect of glucose deprivation on P-gp expression in a rat hepatoma cell line (Fao). Incubation of Fao cells with a glucose-free medium enhanced P-gp mRNA and protein expression in a time-dependent manner, up to 400% at 40 h. This effect was associated with a stimulation of [(3)H]vinblastine efflux by P-gp despite impaired glycosylation. It was reproduced by inducers of endoplasmic reticulum stress response, such as 2-deoxyglucose (DG), tunicamycin, and thapsigargin. P-gp mRNA induction by DG was preceded by an increase in activator protein binding activity, c-Jun expression, and phosphorylation. In contrast, nuclear factor-kappaB binding activity was unaffected by DG. The antioxidant N-acetylcysteine partially reversed the increase in P-gp mRNA and protein levels induced by DG, as well as the enhancement of c-Jun phosphorylation and activator protein binding activity. Finally, transient transfection of the cells with a deleted mutant of c-Jun, Tam 67, abolished the DG-induced P-gp mRNA expression and mdr1b promoter activation. In conclusion, glucose deprivation enhances P-gp expression and transport function in liver cancer cells. This effect is mediated by endoplasmic reticulum stress response and involves MDR transcriptional induction through c-Jun activation. These results emphasize the importance of glucose metabolism in chemoresistance.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Carcinoma Hepatocelular/metabolismo , Retículo Endoplasmático/fisiologia , Glucose/deficiência , Neoplasias Hepáticas/metabolismo , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Desoxiglucose/farmacologia , Resistência a Múltiplos Medicamentos/genética , Glucose/metabolismo , Glicosilação , Humanos , Neoplasias Hepáticas/genética , NF-kappa B/fisiologia , Estresse Oxidativo/fisiologia , Fragmentos de Peptídeos/genética , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fator de Transcrição AP-1/fisiologia , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética
5.
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
6.
J Clin Invest ; 120(11): 4065-76, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20921623

RESUMO

Mechanisms of progression of chronic kidney disease (CKD), a major health care burden, are poorly understood. EGFR stimulates CKD progression, but the molecular networks that mediate its biological effects remain unknown. We recently showed that the severity of renal lesions after nephron reduction varied substantially among mouse strains and required activation of EGFR. Here, we utilized two mouse strains that react differently to nephron reduction--FVB/N mice, which develop severe renal lesions, and B6D2F1 mice, which are resistant to early deterioration--coupled with genome-wide expression to elucidate the molecular nature of CKD progression. Our results showed that lipocalin 2 (Lcn2, also known as neutrophil gelatinase-associated lipocalin [NGAL]), the most highly upregulated gene in the FVB/N strain, was not simply a marker of renal lesions, but an active player in disease progression. In fact, the severity of renal lesions was dramatically reduced in Lcn2-/- mice. We discovered that Lcn2 expression increased upon EGFR activation and that Lcn2 mediated its mitogenic effect during renal deterioration. EGFR inhibition prevented Lcn2 upregulation and lesion development in mice expressing a dominant negative EGFR isoform, and hypoxia-inducible factor 1α (Hif-1α) was crucially required for EGFR-induced Lcn2 overexpression. Consistent with this, cell proliferation was dramatically reduced in Lcn2-/- mice. These data are relevant to human CKD, as we found that LCN2 was increased particularly in patients who rapidly progressed to end-stage renal failure. Together our results uncover what we believe to be a novel function for Lcn2 and a critical pathway leading to progressive renal failure and cystogenesis.


Assuntos
Proteínas de Fase Aguda/metabolismo , Progressão da Doença , Lipocalinas/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Insuficiência Renal Crônica/patologia , Proteínas de Fase Aguda/genética , Adulto , Idoso , Animais , Linhagem Celular , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipocalina-2 , Lipocalinas/genética , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas Oncogênicas/genética , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia , Proteínas Proto-Oncogênicas/genética , Insuficiência Renal Crônica/fisiopatologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA