Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Mol Genet ; 29(1): 31-48, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31625572

RESUMO

Polycystic kidney disease (PKD) results in the formation of renal cysts that can impair function leading to renal failure. DNA damage accumulates in renal epithelial cells in PKD, but the molecular mechanisms are unclear and are investigated here. Phosphoinositide 3-kinase (PI3K)/AKT signaling activates mammalian target of rapamycin complex 1 (mTORC1) and hyperactivation of mTORC1 is a common event in PKD; however, mTORC1 inhibitors have yielded disappointing results in clinical trials. Here, we demonstrate AKT and mTORC1 hyperactivation in two representative murine PKD models (renal epithelial-specific Inpp5e knockout and collecting duct-specific Pkd1 deletion) and identify a downstream signaling network that contributes to DNA damage accumulation. Inpp5e- and Pkd1-null renal epithelial cells showed DNA damage including double-stranded DNA breaks associated with increased replication fork numbers, multinucleation and centrosome amplification. mTORC1 activated CAD, which promotes de novo pyrimidine synthesis, to sustain cell proliferation. AKT, but not mTORC1, inhibited the DNA repair/replication fork origin firing regulator TOPBP1, which impacts on DNA damage and cell proliferation. Notably, Inpp5e- and Pkd1-null renal epithelial cell spheroid formation defects were rescued by AKT inhibition. These data reveal that AKT hyperactivation contributes to DNA damage accumulation in multiple forms of PKD and cooperates with mTORC1 to promote cell proliferation. Hyperactivation of AKT may play a causal role in PKD by regulating DNA damage and cell proliferation, independent of mTORC1, and AKT inhibition may be a novel therapeutic approach for PKD.


Assuntos
Dano ao DNA/fisiologia , Doenças Renais Policísticas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proliferação de Células/genética , Proliferação de Células/fisiologia , Células Cultivadas , Dano ao DNA/genética , Eletroforese em Gel de Poliacrilamida , Immunoblotting , Imuno-Histoquímica , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Doenças Renais Policísticas/genética , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
2.
Hum Mol Genet ; 28(2): 230-244, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30265301

RESUMO

Polycystic kidney disease (PKD) results from excessive renal epithelial cell proliferation, leading to the formation of large fluid filled cysts which impair renal function and frequently lead to renal failure. Hyperactivation of numerous signaling pathways is hypothesized to promote renal epithelial cell hyperproliferation including mTORC1, extracellular signal-regulated kinase (ERK) and WNT signaling. ß-catenin and its target genes are overexpressed in some PKD models and expression of activated ß-catenin induces cysts in mice; however, ß-catenin murine knockout studies indicate it may also inhibit cystogenesis. Therefore, it remains unclear whether ß-catenin is pro- or anti-cystogenic and whether its role is canonical WNT signaling-dependent. Here, we investigate whether ß-catenin deletion in a PKD model with hyperactived ß-catenin signaling affects disease progression to address whether increased ß-catenin drives PKD. We used renal epithelial cell specific Inpp5e-null PKD mice which we report exhibit increased ß-catenin and target gene expression in the cystic kidneys. Surprisingly, co-deletion of ß-catenin with Inpp5e in renal epithelial cells exacerbated polycystic kidney disease and renal failure compared to Inpp5e deletion alone, but did not normalize ß-catenin target gene expression. ß-catenin/Inpp5e double-knockout kidneys exhibited increased cyst initiation, cell proliferation and MEK/ERK signaling compared to Inpp5e-null, associated with increased fibrosis, which may collectively contribute to accelerated disease. Therefore, increased ß-catenin and WNT target gene expression are not necessarily cyst promoting. Rather ß-catenin may play a dual and context-dependent role in PKD and in the presence of other cyst-inducing mutations (Inpp5e-deletion); ß-catenin loss may exacerbate disease in a WNT target gene-independent manner.


Assuntos
Doenças Renais Policísticas/metabolismo , beta Catenina/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Progressão da Doença , Deleção de Genes , Expressão Gênica , Rim/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Monoéster Fosfórico Hidrolases/genética , Doenças Renais Policísticas/enzimologia , Doenças Renais Policísticas/genética , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/genética
3.
J Cell Biol ; 216(1): 247-263, 2017 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-27998989

RESUMO

Human ciliopathies, including Joubert syndrome (JBTS), arise from cilia dysfunction. The inositol polyphosphate 5-phosphatase INPP5E localizes to cilia and is mutated in JBTS. Murine Inpp5e ablation is embryonically lethal and recapitulates JBTS, including neural tube defects and polydactyly; however, the underlying defects in cilia signaling and the function of INPP5E at cilia are still emerging. We report Inpp5e-/- embryos exhibit aberrant Hedgehog-dependent patterning with reduced Hedgehog signaling. Using mouse genetics, we show increasing Hedgehog signaling via Smoothened M2 expression rescues some Inpp5e-/- ciliopathy phenotypes and "normalizes" Hedgehog signaling. INPP5E's phosphoinositide substrates PI(4,5)P2 and PI(3,4,5)P3 accumulated at the transition zone (TZ) in Hedgehog-stimulated Inpp5e-/- cells, which was associated with reduced recruitment of TZ scaffolding proteins and reduced Smoothened levels at cilia. Expression of wild-type, but not 5-phosphatase-dead, INPP5E restored TZ molecular organization and Smoothened accumulation at cilia. Therefore, we identify INPP5E as an essential point of convergence between Hedgehog and phosphoinositide signaling at cilia that maintains TZ function and Hedgehog-dependent embryonic development.


Assuntos
Anormalidades Múltiplas/enzimologia , Cerebelo/anormalidades , Cílios/enzimologia , Embrião de Mamíferos/enzimologia , Anormalidades do Olho/enzimologia , Doenças Renais Císticas/enzimologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Retina/anormalidades , Epitélio Pigmentado da Retina/enzimologia , Sistemas do Segundo Mensageiro , Anormalidades Múltiplas/genética , Animais , Linhagem Celular , Cerebelo/enzimologia , Modelos Animais de Doenças , Anormalidades do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Doenças Renais Císticas/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Monoéster Fosfórico Hidrolases/deficiência , Monoéster Fosfórico Hidrolases/genética , Retina/enzimologia , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Fatores de Tempo , Transfecção , Proteína Gli2 com Dedos de Zinco
4.
Hum Mol Genet ; 25(11): 2295-2313, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27056978

RESUMO

Polycystic kidney disease (PKD) is a common cause of renal failure with few effective treatments. INPP5E is an inositol polyphosphate 5-phosphatase that dephosphorylates phosphoinositide 3-kinase (PI3K)-generated PI(3,4,5)P3 and is mutated in ciliopathy syndromes. Germline Inpp5e deletion is embryonically lethal, attributed to cilia stability defects, and is associated with polycystic kidneys. However, the molecular mechanisms responsible for PKD development upon Inpp5e loss remain unknown. Here, we show conditional inactivation of Inpp5e in mouse kidney epithelium results in severe PKD and renal failure, associated with a partial reduction in cilia number and hyperactivation of PI3K/Akt and downstream mammalian target of rapamycin complex 1 (mTORC1) signaling. Treatment with an mTORC1 inhibitor improved kidney morphology and function, but did not affect cilia number or length. Therefore, we identify Inpp5e as an essential inhibitor of the PI3K/Akt/mTORC1 signaling axis in renal epithelial cells, and demonstrate a critical role for Inpp5e-dependent mTORC1 regulation in PKD suppression.


Assuntos
Rim/metabolismo , Complexos Multiproteicos/genética , Monoéster Fosfórico Hidrolases/genética , Doenças Renais Policísticas/genética , Serina-Treonina Quinases TOR/genética , Animais , Ciliopatias/tratamento farmacológico , Ciliopatias/genética , Ciliopatias/patologia , Modelos Animais de Doenças , Elafina/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Mutação em Linhagem Germinativa , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Doenças Renais Policísticas/tratamento farmacológico , Doenças Renais Policísticas/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Deleção de Sequência , Transdução de Sinais/efeitos dos fármacos , Sirolimo/administração & dosagem , Serina-Treonina Quinases TOR/antagonistas & inibidores
5.
J Cell Sci ; 128(2): 364-72, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25395580

RESUMO

Mutations in inositol polyphosphate 5-phosphatase E (INPP5E) cause the ciliopathies known as Joubert and MORM syndromes; however, the role of INPP5E in ciliary biology is not well understood. Here, we describe an interaction between INPP5E and AURKA, a centrosomal kinase that regulates mitosis and ciliary disassembly, and we show that this interaction is important for the stability of primary cilia. Furthermore, AURKA phosphorylates INPP5E and thereby increases its 5-phosphatase activity, which in turn promotes transcriptional downregulation of AURKA, partly through an AKT-dependent mechanism. These findings establish the first direct link between AURKA and phosphoinositide signaling and suggest that the function of INPP5E in cilia is at least partly mediated by its interactions with AURKA.


Assuntos
Aurora Quinase A/metabolismo , Cílios/metabolismo , Fosfatidilinositóis/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Aurora Quinase A/genética , Cerebelo/anormalidades , Cerebelo/patologia , Cílios/genética , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Regulação da Expressão Gênica , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Mitose/genética , Mutação , Monoéster Fosfórico Hidrolases/genética , Mapas de Interação de Proteínas/genética , Retina/anormalidades , Retina/patologia , Transdução de Sinais
6.
Curr Top Microbiol Immunol ; 362: 247-314, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23086422

RESUMO

Phosphoinositide signalling molecules interact with a plethora of effector proteins to regulate cell proliferation and survival, vesicular trafficking, metabolism, actin dynamics and many other cellular functions. The generation of specific phosphoinositide species is achieved by the activity of phosphoinositide kinases and phosphatases, which phosphorylate and dephosphorylate, respectively, the inositol headgroup of phosphoinositide molecules. The phosphoinositide phosphatases can be classified as 3-, 4- and 5-phosphatases based on their specificity for dephosphorylating phosphates from specific positions on the inositol head group. The SAC phosphatases show less specificity for the position of the phosphate on the inositol ring. The phosphoinositide phosphatases regulate PI3K/Akt signalling, insulin signalling, endocytosis, vesicle trafficking, cell migration, proliferation and apoptosis. Mouse knockout models of several of the phosphoinositide phosphatases have revealed significant physiological roles for these enzymes, including the regulation of embryonic development, fertility, neurological function, the immune system and insulin sensitivity. Importantly, several phosphoinositide phosphatases have been directly associated with a range of human diseases. Genetic mutations in the 5-phosphatase INPP5E are causative of the ciliopathy syndromes Joubert and MORM, and mutations in the 5-phosphatase OCRL result in Lowe's syndrome and Dent 2 disease. Additionally, polymorphisms in the 5-phosphatase SHIP2 confer diabetes susceptibility in specific populations, whereas reduced protein expression of SHIP1 is reported in several human leukaemias. The 4-phosphatase, INPP4B, has recently been identified as a tumour suppressor in human breast and prostate cancer. Mutations in one SAC phosphatase, SAC3/FIG4, results in the degenerative neuropathy, Charcot-Marie-Tooth disease. Indeed, an understanding of the precise functions of phosphoinositide phosphatases is not only important in the context of normal human physiology, but to reveal the mechanisms by which these enzyme families are implicated in an increasing repertoire of human diseases.


Assuntos
Fosfatidilinositóis/metabolismo , Monoéster Fosfórico Hidrolases/fisiologia , Animais , Humanos , Inositol Polifosfato 5-Fosfatases , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases
7.
EMBO Rep ; 10(5): 487-93, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19325558

RESUMO

Akt is a crucial phosphoinositide 3-kinase (PI(3)K) effector that regulates cell proliferation and survival. PI(3)K-generated signals, PtdIns(3,4,5)P(3) and PtdIns(3,4)P(2), direct Akt plasma membrane engagement. Pathological Akt plasma membrane association promotes oncogenesis. PtdIns(3,4)P(2) is degraded by inositol polyphosphate 4-phosphatase-1 (4-ptase-1) forming PtdIns(3)P; however, the role of 4-ptase-1 in regulating the activation and function of Akt is unclear. In mouse embryonic fibroblasts lacking 4-ptase-1 ((-/-)MEFs), the Akt-pleckstrin homology (PH) domain was constitutively membrane-associated both in serum-starved and agonist-stimulated cells, in contrast to (+/+)MEFs, in which it was detected only at the plasma membrane following serum stimulation. Epidermal growth factor (EGF) stimulation resulted in increased Ser(473) and Thr(308)-Akt phosphorylation and activation of Akt-dependent signalling in (-/-)MEFs, relative to (+/+)MEFs. Significantly, loss of 4-ptase-1 resulted in increased cell proliferation and decreased apoptosis. SV40-transformed (-/-)MEFs showed increased anchorage-independent cell growth and formed tumours in nude mice. This study provides the first evidence, to our knowledge, that 4-ptase-1 controls the activation of Akt and thereby cell proliferation, survival and tumorigenesis.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Monoéster Fosfórico Hidrolases/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cicloeximida/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos , Camundongos , Monoéster Fosfórico Hidrolases/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estaurosporina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...