Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 371, 2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38191531

RESUMO

Aurora Kinase A (AURKA) promotes cell proliferation and is overexpressed in different types of polycystic kidney disease (PKD). To understand AURKA's role in regulating renal cyst development we conditionally deleted the gene in mouse models of Autosomal Dominant PKD (ADPKD) and Joubert Syndrome, caused by Polycystin 1 (Pkd1) and Inositol polyphosphate-5-phosphatase E (Inpp5e) mutations respectively. We show that while Aurka is dispensable for collecting duct development and homeostasis, its deletion prevents cyst formation in both disease models. Cross-comparison of transcriptional changes implicated AKT signaling in cyst prevention and we show that (i) AURKA and AKT physically interact, (ii) AURKA regulates AKT activity in a kinase-independent manner and (iii) inhibition of AKT can reduce disease severity. AKT activation also regulates Aurka expression, creating a feed-forward loop driving renal cystogenesis. We find that the AURKA kinase inhibitor Alisertib stabilises the AURKA protein, agonizing its cystogenic functions. These studies identify AURKA as a master regulator of renal cyst development in different types of PKD, functioning in-part via AKT.


Assuntos
Aurora Quinase A , Cistos , Doenças Renais Policísticas , Rim Policístico Autossômico Dominante , Animais , Camundongos , Aurora Quinase A/genética , Monoéster Fosfórico Hidrolases , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/prevenção & controle , Proteínas Proto-Oncogênicas c-akt/genética
2.
Mol Pharm ; 19(3): 843-852, 2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35133169

RESUMO

Folate receptor (FR) overexpression in a wide range of solid tumors provides an opportunity to develop novel, targeted cancer therapeutics. In this study, we investigated whether prebinding the chemotherapeutic methotrexate (MTX) to folate-binding protein (FBP), the soluble form of FR, would enable the protein to serve as a targeted therapeutic vector, enhancing uptake into tumor cells and improving therapeutic efficacy. In an in vivo study, using an FR-overexpressing KB xenograft model in SCID mice, modest improvement in inhibiting tumor growth was observed for the MTX/FBP mixtures as compared to saline control and free MTX. Surprisingly, FBP alone inhibited tumor growth compared to saline control, free MTX, and FBP/MTX. In order to better understand this effect, we investigated the cytotoxicity of micromolar concentrations of FBP in vitro using the KB, HeLa, and A549 cancer cell lines. Our results revealed concentration-dependent apoptosis (24 h; 10-50 µM) in all three cell lines accompanied by a time- and concentration-dependent reduction (6, 12, and 24 h; 10-50 µM) in metabolic activity and compromised cell plasma membrane integrity. This study demonstrates an apoptosis pathway for cytotoxicity of FBP, an endogenous serum protein, in cancer cell lines with widely varying levels of FR expression. Furthermore, in vivo tumor growth suppression for xenograft KB tumors in SCID mice was observed. These studies suggest novel strategies for the elimination of cancer cells employing endogenous, serum transport proteins.


Assuntos
Proteínas de Transporte , Ácido Fólico , Animais , Proteínas de Transporte/metabolismo , Receptores de Folato com Âncoras de GPI , Ácido Fólico/metabolismo , Humanos , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Camundongos , Camundongos SCID
3.
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
4.
J Biol Chem ; 294(45): 16684-16697, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31543504

RESUMO

Macrophage phagocytosis is required for effective clearance of invading bacteria and other microbes. Coordinated phosphoinositide signaling is critical both for phagocytic particle engulfment and subsequent phagosomal maturation to a degradative organelle. Phosphatidylinositol 3-phosphate (PtdIns(3)P) is a phosphoinositide that is rapidly synthesized and degraded on phagosomal membranes, where it recruits FYVE domain- and PX motif-containing proteins that promote phagosomal maturation. However, the molecular mechanisms that regulate PtdIns(3)P removal from the phagosome have remained unclear. We report here that a myotubularin PtdIns(3)P 3-phosphatase, myotubularin-related protein-4 (MTMR4), regulates macrophage phagocytosis. MTMR4 overexpression reduced and siRNA-mediated Mtmr4 silencing increased levels of cell-surface immunoglobulin receptors (i.e. Fcγ receptors (FcγRs)) on RAW 264.7 macrophages, associated with altered pseudopodal F-actin. Furthermore, MTMR4 negatively regulated the phagocytosis of IgG-opsonized particles, indicating that MTMR4 inhibits FcγR-mediated phagocytosis, and was dynamically recruited to phagosomes of macrophages during phagocytosis. MTMR4 overexpression decreased and Mtmr4-specific siRNA expression increased the duration of PtdIns(3)P on phagosomal membranes. Macrophages treated with Mtmr4-specific siRNA were more resistant to Mycobacterium marinum-induced phagosome arrest, associated with increased maturation of mycobacterial phagosomes, indicating that extended PtdIns(3)P signaling on phagosomes in the Mtmr4-knockdown cells permitted trafficking of phagosomes to acidic late endosomal and lysosomal compartments. In conclusion, our findings indicate that MTMR4 regulates PtdIns(3)P degradation in macrophages and thereby controls phagocytosis and phagosomal maturation.


Assuntos
Fagocitose , Fagossomos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Actinas/metabolismo , Animais , Endossomos/metabolismo , Humanos , Imunoglobulina G/imunologia , Lisossomos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Mycobacterium marinum/patogenicidade , Proteínas Tirosina Fosfatases não Receptoras/antagonistas & inibidores , Proteínas Tirosina Fosfatases não Receptoras/genética , Células RAW 264.7 , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de IgG/metabolismo , Transdução de Sinais
5.
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
6.
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
7.
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
8.
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
9.
Cancer Res ; 73(16): 5066-79, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23801747

RESUMO

It is now clear that progression from localized prostate cancer to incurable castrate-resistant prostate cancer (CRPC) is driven by continued androgen receptor (AR), signaling independently of androgen. Thus, there remains a strong rationale to suppress AR activity as the single most important therapeutic goal in CRPC treatment. Although the expression of ligand-independent AR splice variants confers resistance to AR-targeted therapy and progression to lethal castrate-resistant cancer, the molecular regulators of AR activity in CRPC remain unclear, in particular those pathways that potentiate the function of mutant AR in CRPC. Here, we identify FHL2 as a novel coactivator of ligand-independent AR variants that are important in CRPC. We show that the nuclear localization of FHL2 and coactivation of the AR is driven by calpain cleavage of the cytoskeletal protein filamin, a pathway that shows differential activation in prostate epithelial versus prostate cancer cell lines. We further identify a novel FHL2-AR-filamin transcription complex, revealing how deregulation of this axis promotes the constitutive, ligand-independent activation of AR variants, which are present in CRPC. Critically, the calpain-cleaved filamin fragment and FHL2 are present in the nucleus only in CRPC and not benign prostate tissue or localized prostate cancer. Thus, our work provides mechanistic insight into the enhanced AR activation, most notably of the recently identified AR variants, including AR-V7 that drives CRPC progression. Furthermore, our results identify the first disease-specific mechanism for deregulation of FHL2 nuclear localization during cancer progression. These results offer general import beyond prostate cancer, given that nuclear FHL2 is characteristic of other human cancers where oncogenic transcription factors that drive disease are activated like the AR in prostate cancer.


Assuntos
Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Células COS , Calpaína/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Chlorocebus aethiops , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Filaminas/genética , Filaminas/metabolismo , Humanos , Ligantes , Masculino , Neoplasias de Próstata Resistentes à Castração/patologia , Ativação Transcricional
10.
FEBS Lett ; 586(18): 2846-57, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22828281

RESUMO

Phosphoinositides regulate numerous cellular events via the recruitment and activation of multiple lipid-binding effector proteins. The precise temporal and spatial regulation of phosphoinositide signals by the co-ordinated activities of phosphoinositide kinases and phosphatases is essential for homeostasis and development. Mutations in two inositol polyphosphate 5-phosphatases, INPP5E and OCRL, cause the cerebrorenal syndromes of Joubert and Lowe's, respectively. INPP5E and OCRL exhibit overlapping phosphoinositide substrate specificity and subcellular localisation, including an association with the primary cilia. Here, we review recent studies that identify a new role for these enzymes in the regulation of primary cilia function. Joubert syndrome has been extensively linked to primary cilia defects, and Lowe's may represent a new class of 'ciliopathy associated' syndromes.


Assuntos
Cílios/patologia , Cílios/fisiologia , Monoéster Fosfórico Hidrolases/metabolismo , Humanos , Inositol Polifosfato 5-Fosfatases , Mutação , Fosfatidilinositóis/metabolismo , Monoéster Fosfórico Hidrolases/genética , Síndrome
11.
Subcell Biochem ; 58: 215-79, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22403078

RESUMO

Phosphoinositide phosphatases comprise several large enzyme families with over 35 mammalian enzymes identified to date that degrade many phosphoinositide signals. Growth factor or insulin stimulation activates the phosphoinositide 3-kinase that phosphorylates phosphatidylinositol (4,5)-bisphosphate [PtdIns(4,5)P(2)] to form phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P(3)], which is rapidly dephosphorylated either by PTEN (phosphatase and tensin homologue deleted on chromosome 10) to PtdIns(4,5)P(2), or by the 5-phosphatases (inositol polyphosphate 5-phosphatases), generating PtdIns(3,4)P(2). 5-phosphatases also hydrolyze PtdIns(4,5)P(2) forming PtdIns(4)P. Ten mammalian 5-phosphatases have been identified, which regulate hematopoietic cell proliferation, synaptic vesicle recycling, insulin signaling, and embryonic development. Two 5-phosphatase genes, OCRL and INPP5E are mutated in Lowe and Joubert syndrome respectively. SHIP [SH2 (Src homology 2)-domain inositol phosphatase] 2, and SKIP (skeletal muscle- and kidney-enriched inositol phosphatase) negatively regulate insulin signaling and glucose homeostasis. SHIP2 polymorphisms are associated with a predisposition to insulin resistance. SHIP1 controls hematopoietic cell proliferation and is mutated in some leukemias. The inositol polyphosphate 4-phosphatases, INPP4A and INPP4B degrade PtdIns(3,4)P(2) to PtdIns(3)P and regulate neuroexcitatory cell death, or act as a tumor suppressor in breast cancer respectively. The Sac phosphatases degrade multiple phosphoinositides, such as PtdIns(3)P, PtdIns(4)P, PtdIns(5)P and PtdIns(3,5)P(2) to form PtdIns. Mutation in the Sac phosphatase gene, FIG4, leads to a degenerative neuropathy. Therefore the phosphatases, like the lipid kinases, play major roles in regulating cellular functions and their mutation or altered expression leads to many human diseases.


Assuntos
Neoplasias da Mama/enzimologia , Leucemia/enzimologia , Síndrome Oculocerebrorrenal/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Diglicerídeos/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Inositol Polifosfato 5-Fosfatases , Leucemia/genética , Leucemia/patologia , Síndrome Oculocerebrorrenal/genética , Síndrome Oculocerebrorrenal/patologia , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/genética , Sistemas do Segundo Mensageiro
12.
J Biol Chem ; 286(34): 29758-70, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21712384

RESUMO

Phosphoinositide 3-kinase (PI3K) regulates cell polarity and migration by generating phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P(3)) at the leading edge of migrating cells. The serine-threonine protein kinase Akt binds to PI(3,4,5)P(3), resulting in its activation. Active Akt promotes spatially regulated actin cytoskeletal remodeling and thereby directed cell migration. The inositol polyphosphate 5-phosphatases (5-ptases) degrade PI(3,4,5)P(3) to form PI(3,4)P(2), which leads to diminished Akt activation. Several 5-ptases, including SKIP and SHIP2, inhibit actin cytoskeletal reorganization by opposing PI3K/Akt signaling. In this current study, we identify a molecular co-chaperone termed silencer of death domains (SODD/BAG4) that forms a complex with several 5-ptase family members, including SKIP, SHIP1, and SHIP2. The interaction between SODD and SKIP exerts an inhibitory effect on SKIP PI(3,4,5)P(3) 5-ptase catalytic activity and consequently enhances the recruitment of PI(3,4,5)P(3)-effectors to the plasma membrane. In contrast, SODD(-/-) mouse embryonic fibroblasts exhibit reduced Akt-Ser(473) and -Thr(308) phosphorylation following EGF stimulation, associated with increased SKIP PI(3,4,5)P(3)-5-ptase activity. SODD(-/-) mouse embryonic fibroblasts exhibit decreased EGF-stimulated F-actin stress fibers, lamellipodia, and focal adhesion complexity, a phenotype that is rescued by the expression of constitutively active Akt1. Furthermore, reduced cell migration was observed in SODD(-/-) macrophages, which express the three 5-ptases shown to interact with SODD (SKIP, SHIP1, and SHIP2). Therefore, this study identifies SODD as a novel regulator of PI3K/Akt signaling to the actin cytoskeleton.


Assuntos
Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Fibras de Estresse/metabolismo , Actinas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Movimento Celular/fisiologia , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Adesões Focais/genética , Adesões Focais/metabolismo , Inositol Polifosfato 5-Fosfatases , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Monoéster Fosfórico Hidrolases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Pseudópodes/genética , Pseudópodes/metabolismo , Fibras de Estresse/genética
13.
J Biol Chem ; 286(26): 23407-18, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21550974

RESUMO

Neuron polarization is essential for the formation of one axon and multiple dendrites, establishing the neuronal circuitry. Phosphoinositide 3-kinase (PI3K) signaling promotes axon selection and elongation. Here we report in hippocampal neurons siRNA knockdown of the proline-rich inositol polyphosphate 5-phosphatase (PIPP), which degrades PI3K-generated PtdIns(3,4,5)P(3), results in multiple hyperelongated axons consistent with a polarization defect. We identify collapsin response mediator protein 2 (CRMP2), which regulates axon selection by promoting WAVE1 delivery via Kinesin-1 motors to the axon growth cone, as a PIPP-interacting protein by Y2H screening, direct binding studies, and coimmunoprecipitation of an endogenous PIPP, CRMP2, and Kinesin-1 complex from brain lysates. The C-terminal growth cone-targeting domain of PIPP facilitates its interaction with CRMP2. PIPP growth cone localization is CRMP2-dependent. PIPP knockdown in PC12 cells promotes neurite elongation, WAVE1 and Kinesin-1 growth cone localization, whereas knockdown of CRMP2 exhibits the opposite phenotype, with shorter neurites and decreased WAVE1/Kinesin-1 at the growth cone. In contrast, CRMP2 overexpression promotes neurite elongation, a phenotype rescued by full-length PIPP, or expression of the CRMP2-binding PIPP domain. Therefore this study identifies PIPP and CRMP2 exert opposing roles in promoting axon selection and neurite elongation and the complex between these proteins serves to regulate the localization of effectors that promote neurite extension.


Assuntos
Cones de Crescimento/metabolismo , Hipocampo/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuritos/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Hipocampo/citologia , Inositol Polifosfato 5-Fosfatases , Peptídeos e Proteínas de Sinalização Intercelular , Cinesinas/genética , Cinesinas/metabolismo , Masculino , Complexos Multiproteicos/genética , Proteínas do Tecido Nervoso/genética , Células PC12 , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolases/genética , Ratos , Ratos Sprague-Dawley , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo
14.
J Cell Sci ; 123(Pt 18): 3071-83, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20736309

RESUMO

Phosphatidylinositol 3-phosphate [PtdIns(3)P] regulates endocytic trafficking and the sorting of receptors through early endosomes, including the rapid recycling of transferrin (Tfn). However, the phosphoinositide phosphatase that selectively opposes this function is unknown. The myotubularins are a family of eight catalytically active and six inactive enzymes that hydrolyse PtdIns(3)P to form PtdIns. However, the role each myotubularin family member plays in regulating endosomal PtdIns(3)P and thereby endocytic trafficking is not well established. Here, we identify the myotubularin family member MTMR4, which localizes to early endosomes and also to Rab11- and Sec15-positive recycling endosomes. In cells with MTMR4 knockdown, or following expression of the catalytically inactive MTMR4, MTMR4(C407A), the number of PtdIns(3)P-decorated endosomes significantly increased. MTMR4 overexpression delayed the exit of Tfn from early endosomes and its recycling to the plasma membrane. By contrast, expression of MTMR4(C407A), which acts as a dominant-negative construct, significantly accelerated Tfn recycling. However, in MTMR4 knockdown cells Tfn recycling was unchanged, suggesting that other MTMs might also contribute to recycling. MTMR4 regulated the subcellular distribution of Rab11 and, in cells with RNAi-mediated knockdown of MTMR4, Rab11 was directed away from the pericentriolar recycling compartment. The subcellular distribution of VAMP3, a v-SNARE protein that resides in recycling endosomes and endosome-derived transport vesicles, was also regulated by MTMR4. Therefore, MTMR4 localizes at the interface of early and recycling endosomes to regulate trafficking through this pathway.


Assuntos
Endossomos/enzimologia , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Animais , Transporte Biológico , Células COS , Linhagem Celular , Chlorocebus aethiops , Endossomos/genética , Endossomos/metabolismo , Humanos , Fosfatos de Fosfatidilinositol/metabolismo , Transporte Proteico , Proteínas Tirosina Fosfatases não Receptoras/genética
15.
Methods Mol Biol ; 462: 223-39, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19160673

RESUMO

Phosphatidylinositol 3,4,5 trisphosphate [PtdIns(3,4,5)P3] is a potent membrane-bound signaling molecule transiently synthesized by phosphoinositide 3-kinase (PI3-kinase) in response to extracellular agonists. PtdIns(3,4,5)P3 signals need to be strictly controlled. PtdIns(3,4,5)P3 recruits and binds effectors that function in oncogenic signaling pathways. PtdIns(3,4,5)P3 activates cell proliferation, growth, and migration as well as regulating insulin signaling. The inositol polyphosphate 5-phosphatase family of enzymes dephosphorylate and thereby modulate PtdIns(3,4,5)P3 levels, attenuating PI3-kinase-dependent signaling. PtdIns(3,4,5)P3 5-phosphatase enzyme activity can be assessed in vitro by analysis of the hydrolysis of radiolabeled or fluorescently labeled PtdIns(3,4,5)P3 and in vivo by visualization of the recruitment and turnover of the PtdIns(3,4,5)P3-specific biosensor GFP-PH/ ARNO or other PtdIns(3,4,5)P3 binding proteins at the plasma membrane.


Assuntos
Monoéster Fosfórico Hidrolases/análise , Monoéster Fosfórico Hidrolases/metabolismo , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Imunofluorescência , Imunoprecipitação , Microscopia Confocal , Monoéster Fosfórico Hidrolases/isolamento & purificação , Proteínas Recombinantes/análise , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Coloração e Rotulagem , Transfecção
16.
J Biol Chem ; 281(11): 7666-83, 2006 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-16407297

RESUMO

Four and a half LIM protein 1 (FHL1/SLIM1) is highly expressed in skeletal and cardiac muscle; however, the function of FHL1 remains unknown. Yeast two-hybrid screening identified slow type skeletal myosin-binding protein C as an FHL1 binding partner. Myosin-binding protein C is the major myosin-associated protein in striated muscle that enhances the lateral association and stabilization of myosin thick filaments and regulates actomyosin interactions. The interaction between FHL1 and myosin-binding protein C was confirmed using co-immunoprecipitation of recombinant and endogenous proteins. Recombinant FHL2 and FHL3 also bound myosin-binding protein C. FHL1 impaired co-sedimentation of myosin-binding protein C with reconstituted myosin filaments, suggesting FHL1 may compete with myosin for binding to myosin-binding protein C. In intact skeletal muscle and isolated myofibrils, FHL1 localized to the I-band, M-line, and sarcolemma, co-localizing with myosin-binding protein C at the sarcolemma in intact skeletal muscle. Furthermore, in isolated myofibrils FHL1 staining at the M-line appeared to extend partially into the C-zone of the A-band, where it co-localized with myosin-binding protein C. Overexpression of FHL1 in differentiating C2C12 cells induced "sac-like" myotube formation (myosac), associated with impaired Z-line and myosin thick filament assembly. This phenotype was rescued by co-expression of myosin-binding protein C. FHL1 knockdown using RNAi resulted in impaired myosin thick filament formation associated with reduced incorporation of myosin-binding protein C into the sarcomere. This study identified FHL1 as a novel regulator of myosin-binding protein C activity and indicates a role for FHL1 in sarcomere assembly.


Assuntos
Proteínas de Transporte/fisiologia , Proteínas Musculares/fisiologia , Miosinas/química , Sarcômeros/metabolismo , Actomiosina/química , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Proteínas de Transporte/metabolismo , Diferenciação Celular , Proliferação de Células , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Glutationa Transferase/metabolismo , Humanos , Imuno-Histoquímica , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas com Domínio LIM , Camundongos , Microscopia de Fluorescência , Dados de Sequência Molecular , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miócitos Cardíacos/metabolismo , Oligonucleotídeos/química , Peptídeos/química , Fenótipo , Ligação Proteica , Estrutura Terciária de Proteína , Interferência de RNA , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Transfecção , Técnicas do Sistema de Duplo-Híbrido
17.
Int J Biochem Cell Biol ; 37(11): 2260-5, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15964236

RESUMO

Phosphoinositides are membrane-bound signaling molecules that recruit, activate and localize target effectors to intracellular membranes regulating apoptosis, cell proliferation, insulin signaling and membrane trafficking. The SH2 domain containing inositol polyphosphate 5-phosphatase-2 (SHIP2) hydrolyzes phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) generating phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2). Overexpression of SHIP2 inhibits insulin-stimulated phosphoinositide 3-kinase (PI3K) dependent signaling events. Analysis of diabetic human subjects has revealed an association between SHIP2 gene polymorphisms and type 2 diabetes mellitus. Genetic ablation of SHIP2 in mice has generated conflicting results. SHIP2 knockout mice were originally reported to show lethal neonatal hypoglycemia resulting from insulin hypersensitivity, but in addition to inactivating the SHIP2 gene, the Phox2a gene was also inadvertently deleted. Another SHIP2 knockout mouse has now been generated which inactivates the SHIP2 gene but leaves Phox2a intact. These animals show normal insulin and glucose tolerance but are highly resistant to weight gain on high fat diets, exhibiting an obesity-resistant phenotype. Therefore, SHIP2 remains a significant therapeutic target for the treatment of both obesity and type 2 diabetes.


Assuntos
Fosfatidilinositóis/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Domínios de Homologia de src , Animais , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/genética , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/genética , Polimorfismo Genético , Sistemas do Segundo Mensageiro/fisiologia
18.
Blood ; 102(3): 940-8, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12676785

RESUMO

The platelet receptor for the von Willebrand factor (VWF) glycoprotein Ib-IX-V (GPIb-IX-V) complex mediates platelet adhesion at sites of vascular injury. The cytoplasmic tail of the GPIbalpha subunit interacts with the actin-binding protein, filamin, anchoring the receptor in the cytoskeleton. In motile cells, the second messenger phosphatidylinositol 3,4,5 trisphosphate (PtdIns(3,4,5)P3) induces submembraneous actin remodeling. The inositol polyphosphate 5-phosphatase, Src homology 2 domain-containing inositol polyphosphate 5-phosphatase-2 (SHIP-2), hydrolyzes PtdIns(3,4,5)P3 forming phosphatidylinositol 3,4 bisphosphate (PtdIns(3,4)P2) and regulates membrane ruffling via complex formation with filamin. In this study we investigate the intracellular location and association of SHIP-2 with filamin, actin, and the GPIb-IX-V complex in platelets. Immunoprecipitation of SHIP-2 from the Triton-soluble fraction of unstimulated platelets demonstrated association between SHIP-2, filamin, actin, and GPIb-IX-V. SHIP-2 associated with filamin or GPIb-IX-V was active and demonstrated PtdIns(3,4,5)P3 5-phosphatase activity. Following thrombin or VWF-induced platelet activation, detection of the SHIP-2, filamin, and receptor complex decreased in the Triton-soluble fraction, although in control studies the level of SHIP-2, filamin, or GPIb-IX-V immunoprecipitated by their respective antibodies did not change following platelet activation. In activated platelets spreading on a VWF matrix, SHIP-2 localized intensely with actin at the central actin ring and colocalized with actin and filamin at filopodia and lamellipodia. In spread platelets, GPIb-IX-V localized to the center of the platelet and showed little colocalization with filamin at the plasma membrane. These studies demonstrate a functionally active complex between SHIP-2, filamin, actin, and GPIb-IX-V that may orchestrate the localized hydrolysis of PtdIns(3,4,5)P3 and thereby regulate cortical and submembraneous actin.


Assuntos
Plaquetas/citologia , Proteínas do Citoesqueleto/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Actinas/metabolismo , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Tamanho Celular , Proteínas Contráteis/metabolismo , Citoesqueleto/metabolismo , Filaminas , Humanos , Proteínas dos Microfilamentos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Ativação Plaquetária , Ligação Proteica
19.
IUBMB Life ; 53(1): 25-36, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12018404

RESUMO

Recent studies have identified the inositol polyphosphate 5-phosphatases as a large family of signal modifying enzymes comprising 10 mammalian and 4 yeast family members. A number of investigations including gene-targeted deletion of 5-phosphatases in mice have demonstrated that these enzymes regulate many important cellular events including hematopoietic cell proliferation and activation, insulin signaling, endocytosis, and actin polymerization.


Assuntos
Monoéster Fosfórico Hidrolases/metabolismo , Animais , Sinalização do Cálcio , Marcação de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Fosfatos de Inositol/metabolismo , Inositol Polifosfato 5-Fosfatases , Insulina/metabolismo , Modelos Biológicos , Síndrome Oculocerebrorrenal/enzimologia , Síndrome Oculocerebrorrenal/genética , Monoéster Fosfórico Hidrolases/genética , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Transdução de Sinais , Vesículas Sinápticas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA