Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 136(15)2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37439249

RESUMO

The non-receptor tyrosine kinase SRC is overexpressed and/or hyperactivated in various human cancers, and facilitates cancer progression by promoting invasion and metastasis. However, the mechanisms underlying SRC upregulation are poorly understood. In this study, we demonstrate that transforming growth factor-ß (TGF-ß) induces SRC expression at the transcriptional level by activating an intragenic the SRC enhancer. In the human breast epithelial cell line MCF10A, TGF-ß1 stimulation upregulated one of the SRC promotors, the 1A promoter, resulting in increased SRC mRNA and protein levels. Chromatin immunoprecipitation (ChIP)-sequencing analysis revealed that the SMAD complex is recruited to three enhancer regions ∼15 kb upstream and downstream of the SRC promoter, and one of them is capable of activating the SRC promoter in response to TGF-ß. JUN, a member of the activator protein (AP)-1 family, localises to the enhancer and regulates TGF-ß-induced SRC expression. Furthermore, TGF-ß-induced SRC upregulation plays a crucial role in epithelial-mesenchymal transition (EMT)-associated cell migration by activating the SRC-focal adhesion kinase (FAK) circuit. Overall, these results suggest that TGF-ß-induced SRC upregulation promotes cancer cell invasion and metastasis in a subset of human malignancies.


Assuntos
Transição Epitelial-Mesenquimal , Fator de Crescimento Transformador beta , Humanos , Fator de Crescimento Transformador beta/metabolismo , Transição Epitelial-Mesenquimal/genética , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Linhagem Celular , Proteína-Tirosina Quinases de Adesão Focal , Movimento Celular/fisiologia , Linhagem Celular Tumoral
2.
J Biol Chem ; 298(3): 101630, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35085554

RESUMO

Cancer invasion and metastasis are the major causes of cancer patient mortality. Various growth factors, including hepatocyte growth factor (HGF), are known to promote cancer invasion and metastasis, but the regulatory mechanisms involved are not fully understood. Here, we show that HGF-promoted migration and invasion of breast cancer cells are regulated by CUB domain-containing protein 1 (CDCP1), a transmembrane activator of SRC kinase. In metastatic human breast cancer cell line MDA-MB-231, which highly expresses the HGF receptor MET and CDCP1, we show that CDCP1 knockdown attenuated HGF-induced MET activation, followed by suppression of lamellipodia formation and cell migration/invasion. In contrast, in the low invasive/nonmetastatic breast cancer cell line T47D, which had no detectable MET and CDCP1 expression, ectopic MET expression stimulated the HGF-dependent activation of invasive activity, and concomitant CDCP1 expression activated SRC and further promoted invasive activity. In these cells, CDCP1 expression dramatically activated HGF-induced membrane remodeling, which was accompanied by activation of the small GTPase Rac1. Analysis of guanine nucleotide exchange factors revealed that ARHGEF7 was specifically required for CDCP1-dependent induction of HGF-induced invasive ability. Furthermore, immunofluorescence staining demonstrated that CDCP1 coaccumulated with ARHGEF7. Finally, we confirmed that the CDCP1-SRC axis was also crucial for HGF and ARHGEF7-RAC1 signaling in MDA-MB-231 cells. Altogether, these results demonstrate that the CDCP1-SRC-ARHGEF7-RAC1 pathway plays an important role in the HGF-induced invasion of a subset of breast cancer cells.


Assuntos
Antígenos de Neoplasias , Neoplasias da Mama , Fator de Crescimento de Hepatócito , Fatores de Troca de Nucleotídeo Guanina Rho , Quinases da Família src , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Moléculas de Adesão Celular , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Transdução de Sinais , Quinases da Família src/genética , Quinases da Família src/metabolismo
3.
Commun Biol ; 4(1): 357, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33742113

RESUMO

The naked mole-rat (NMR; Heterocephalus glaber) exhibits cancer resistance and an exceptionally long lifespan of approximately 30 years, but the mechanism(s) underlying increased longevity in NMRs remains unclear. In the present study, we report unique mechanisms underlying cholesterol metabolism in NMR cells, which may be responsible for their anti-senescent properties. NMR fibroblasts expressed ß-catenin abundantly; this high expression was linked to increased accumulation of cholesterol-enriched lipid droplets. Ablation of ß-catenin or inhibition of cholesterol synthesis abolished lipid droplet formation and induced senescence-like phenotypes accompanied by increased oxidative stress. ß-catenin ablation downregulated apolipoprotein F and the LXR/RXR pathway, which are involved in cholesterol transport and biogenesis. Apolipoprotein F ablation also suppressed lipid droplet accumulation and promoted cellular senescence, indicating that apolipoprotein F mediates ß-catenin signaling in NMR cells. Thus, we suggest that ß-catenin in NMRs functions to offset senescence by regulating cholesterol metabolism, which may contribute to increased longevity in NMRs.


Assuntos
Senescência Celular , Colesterol/metabolismo , Fibroblastos/metabolismo , Ratos-Toupeira/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Gotículas Lipídicas/metabolismo , Longevidade , Camundongos , Ratos-Toupeira/genética , Células NIH 3T3 , Estresse Oxidativo
4.
Mol Cell ; 80(3): 437-451.e6, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33157014

RESUMO

Amino-acid-induced lysosomal mechanistic target of rapamycin complex 1 (mTORC1) localization through the Rag GTPases is a critical step for its activation by Rheb GTPase. However, how the mTORC1 interacts with Rheb on the lysosome remains elusive. We report that amino acids enhance the polyubiquitination of Rheb (Ub-Rheb), which shows a strong binding preference for mTORC1 and supports its activation, while the Ub-Rheb is subjected to subsequent degradation. Mechanistically, we identified ATXN3 as a Ub-Rheb deubiquitinase whose lysosomal localization is blocked by active Rag heterodimer in response to amino acid stimulation. Consistently, cells lacking functional Rag heterodimer on the lysosome accumulate Ub-Rheb, and blockade of its degradation instigates robust lysosomal mTORC1 localization and its activation without the Ragulator-Rag system. Thus, polyubiquitination of Rheb is an important post-translational modification, which facilitates the binding of mTORC1 to Rheb on the lysosome and is another crosstalk between the amino acid and growth factor signaling for mTORC1 activation.


Assuntos
Ataxina-3/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Aminoácidos/metabolismo , Animais , Ataxina-3/fisiologia , Linhagem Celular , Enzimas Desubiquitinantes/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Ligação Proteica/fisiologia , Proteína Enriquecida em Homólogo de Ras do Encéfalo/fisiologia , Proteínas Repressoras/metabolismo , Transdução de Sinais/fisiologia , Ubiquitinação
5.
Biochem Biophys Res Commun ; 528(1): 146-153, 2020 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-32451084

RESUMO

The naked mole-rat (NMR, Heterocephalus glaber) is the longest-living known rodent species, with a maximum lifespan of over 30 years. NMRs exhibit negligible senescence, exceptional resistance to cancer, and high basal autophagy activity compared with mouse. The molecular mechanisms and physiological roles underlying the high basal autophagy activity in NMRs remain to be elucidated. We identified that the Atg12-Atg5 conjugate, a critical component of autophagosome formation, was highly expressed in NMR skin fibroblasts (NSFs) compared with that in mouse skin fibroblasts. Phenotypic analysis of Atg5 knockdown NSFs revealed that high basal autophagy activity in NSFs was associated with abundant expression of the Atg12-Atg5 conjugate. Atg5 knockdown in NSFs led to accumulation of dysfunctional mitochondria, and suppressed cell proliferation and cell adhesion ability, promoting apoptosis/anoikis accompanied by upregulation of the apoptosis-related genes, Bax and Noxa. Furthermore, inhibition of the p53/Rb pro-apoptotic pathway with SV40 large T antigen abolished Atg5 knockdown-induced increases in apoptosis/anoikis. Taken together, these findings suggest that high basal autophagy activity in NMR cells, mediated by Atg5, contributes to suppression of p53/Rb-induced apoptosis, which could benefit the longevity of NMR cells.


Assuntos
Anoikis , Apoptose , Proteína 5 Relacionada à Autofagia/metabolismo , Autofagia , Fibroblastos/metabolismo , Proteína do Retinoblastoma/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteína 12 Relacionada à Autofagia/metabolismo , Adesão Celular , Proliferação de Células , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Técnicas de Silenciamento de Genes , Masculino , Mitocôndrias/metabolismo , Ratos-Toupeira , Pele/citologia , Regulação para Cima
6.
Artigo em Inglês | MEDLINE | ID: mdl-32085898

RESUMO

Upregulation of the Src tyrosine kinase is implicated in the progression of cancer. The oncogenic potential of Src is suppressed via several negative regulation systems including degradation via the ubiquitin-proteasome pathway. Here, we show that ubiquitination of Src promotes its secretion via small extracellular vesicles (sEVs) to suppress its oncogenic potential. In MDCK cells expressing a modified Src that can be activated by hydroxytamoxifen, activated Src was transported to late endosomes/lysosomes and secreted via sEVs. The secretion of Src was suppressed by ablation of Cbl E3-ligase, suggesting the contribution of ubiquitination to this process. Activated Src was ubiquitinated at multiple sites, and Lys429 was identified as a critical site for sEV-mediated secretion. Mutation of Src at Lys429 (R429) caused resistance to ubiquitination and decreased its secretion via sEVs. The activated R429 mutant was also transported to late endosomes/lysosomes, whereas its incorporation into intraluminal vesicles was reduced. Activation of the R429 mutant induced a greater FAK activation than that of wild-type Src, thereby potentiating Src-induced invasive phenotypes, such as invadopodia formation and invasive activity. These findings demonstrate that ubiquitination of activated Src at Lys429 promotes its secretion via sEVs, suggesting a potential strategy to suppress the oncogenic function of upregulated Src.

7.
J Cell Physiol ; 234(2): 1730-1744, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30144071

RESUMO

Glaucoma, a progressive and irreversible optic neuropathy, is one of the leading causes of vision impairment worldwide. Elevation of intraocular pressure (IOP) due to transforming growth factor-ß (TGF-ß)-induced dysfunction of the trabecular meshwork is a risk factor for glaucoma, but the underlying molecular mechanisms remain elusive. Here, we show that Src kinase is involved in TGF-ß-induced IOP elevation. We observed that dasatinib, a potent Src inhibitor, suppressed TGF-ß2-induced IOP in rat eyes. Mechanistic analyses in human trabecular meshwork cells showed that TGF-ß2 activated Src signaling and concomitantly increased cytoskeletal remodeling, cell adhesion, and extracellular matrix (ECM) accumulation. Src was activated via TGF-ß2-induced upregulation of the Src scaffolding protein CasL, which mediates the assembly of focal adhesions, cytoskeletal remodeling, and ECM deposition. Activation of Src suppressed the expression of tissue plasminogen activator, thereby attenuating ECM degradation. Furthermore, the Src inhibitor ameliorated TGF-ß2-induced changes in the contractile and adhesive characteristics of trabecular meshwork cells, and ECM deposition. These findings underscore the crucial role of Src activity in TGF-ß-induced IOP elevation and identify Src signaling as a potential therapeutic target in glaucoma.


Assuntos
Glaucoma/enzimologia , Pressão Intraocular , Malha Trabecular/enzimologia , Fator de Crescimento Transformador beta2 , Quinases da Família src/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Cultivadas , Dasatinibe/farmacologia , Modelos Animais de Doenças , Ativação Enzimática , Glaucoma/induzido quimicamente , Glaucoma/patologia , Glaucoma/fisiopatologia , Humanos , Pressão Intraocular/efeitos dos fármacos , Masculino , Inibidores de Proteínas Quinases/farmacologia , Ratos Endogâmicos BN , Transdução de Sinais , Ativador de Plasminogênio Tecidual/genética , Ativador de Plasminogênio Tecidual/metabolismo , Malha Trabecular/efeitos dos fármacos , Malha Trabecular/patologia , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
8.
J Immunol ; 200(11): 3790-3800, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29686050

RESUMO

Amino acid metabolism plays important roles in innate immune cells, including macrophages. Recently, we reported that a lysosomal adaptor protein, Lamtor1, which serves as the scaffold for amino acid-activated mechanistic target of rapamycin complex 1 (mTORC1), is critical for the polarization of M2 macrophages. However, little is known about how Lamtor1 affects the inflammatory responses that are triggered by the stimuli for TLRs. In this article, we show that Lamtor1 controls innate immune responses by regulating the phosphorylation and nuclear translocation of transcription factor EB (TFEB), which has been known as the master regulator for lysosome and autophagosome biogenesis. Furthermore, we show that nuclear translocation of TFEB occurs in alveolar macrophages of myeloid-specific Lamtor1 conditional knockout mice and that these mice are hypersensitive to intratracheal administration of LPS and bleomycin. Our observation clarified that the amino acid-sensing pathway consisting of Lamtor1, mTORC1, and TFEB is involved in the regulation of innate immune responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/imunologia , Imunidade Inata/imunologia , Lisossomos/imunologia , Proteínas/imunologia , Aminoácidos/imunologia , Animais , Autofagia/imunologia , Linhagem Celular , Núcleo Celular/imunologia , Macrófagos/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação/imunologia , Transporte Proteico/imunologia , Células RAW 264.7 , Transdução de Sinais/imunologia , Serina-Treonina Quinases TOR/imunologia
9.
Nat Commun ; 7: 13130, 2016 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-27731330

RESUMO

Macrophages play crucial roles in host defence and tissue homoeostasis, processes in which both environmental stimuli and intracellularly generated metabolites influence activation of macrophages. Activated macrophages are classified into M1 and M2 macrophages. It remains unclear how intracellular nutrition sufficiency, especially for amino acid, influences on macrophage activation. Here we show that a lysosomal adaptor protein Lamtor1, which forms an amino-acid sensing complex with lysosomal vacuolar-type H+-ATPase (v-ATPase), and is the scaffold for amino acid-activated mTORC1 (mechanistic target of rapamycin complex 1), is critically required for M2 polarization. Lamtor1 deficiency, amino-acid starvation, or inhibition of v-ATPase and mTOR result in defective M2 polarization and enhanced M1 polarization. Furthermore, we identified liver X receptor (LXR) as the downstream target of Lamtor1 and mTORC1. Production of 25-hydroxycholesterol is dependent on Lamtor1 and mTORC1. Our findings demonstrate that Lamtor1 plays an essential role in M2 polarization, coupling immunity and metabolism.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Aminoácidos/imunologia , Citocinas/imunologia , Macrófagos/imunologia , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Serina-Treonina Quinases TOR/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Aminoácidos/deficiência , Animais , Diferenciação Celular , Linhagem da Célula/imunologia , Citocinas/genética , Feminino , Regulação da Expressão Gênica , Receptores X do Fígado/genética , Receptores X do Fígado/imunologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrolídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/imunologia , Camundongos , Camundongos Transgênicos , Naftiridinas/farmacologia , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/imunologia , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , ATPases Vacuolares Próton-Translocadoras/genética , ATPases Vacuolares Próton-Translocadoras/imunologia
10.
PLoS One ; 9(2): e88891, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24558442

RESUMO

The mechanistic target of rapamycin (mTOR) functions as a component of two large complexes, mTORC1 and mTORC2, which play crucial roles in regulating cell growth and homeostasis. However, the molecular mechanisms by which mTOR controls cell proliferation remain elusive. Here we show that the FoxO3a transcription factor is coordinately regulated by mTORC1 and mTORC2, and plays a crucial role in controlling cell proliferation. To dissect mTOR signaling, mTORC1 was specifically inactivated by depleting p18, an essential anchor of mTORC1 on lysosomes. mTORC1 inactivation caused a marked retardation of cell proliferation, which was associated with upregulation of cyclin-dependent kinase inhibitors (CDKIs). Although Akt was activated by mTORC1 inactivation, FoxO3a was upregulated via an epigenetic mechanism and hypophosphorylated at Ser314, which resulted in its nuclear accumulation. Consistently, mTORC1 inactivation induced downregulation of serum- and glucocorticoid-inducible kinase 1 (SGK1), the kinase responsible for Ser314 phosphorylation. Expression of FoxO3a mutated at Ser314 suppressed cell proliferation by inducing CDKI expression. SGK1 overexpression suppressed CDKI expression in p18-deficient cells, whereas SGK1 knockdown induced CDKI expression in wild-type cells, resulting in the suppression of cell proliferation. These results suggest that mTORC1, in coordination with mTORC2, controls cell proliferation by regulating FoxO3a gene expression and SGK1-mediated phosphorylation of FoxO3a at Ser314.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Proliferação de Células , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Proteína Forkhead Box O3 , Regulação da Expressão Gênica , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Fosforilação , Regulação para Cima
11.
Methods Enzymol ; 535: 249-63, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24377928

RESUMO

p18/LAMTOR1 is a membrane protein specifically localized to the surface of late endosomes/lysosomes that serves as an anchor for the "Ragulator" complex, which contains p14/LAMTOR2, MP1/LAMTOR3, HBXIP, and C7orf59. The Ragulator interacts with RagAB/CD GTPases and V-ATPase and plays crucial roles for activation of mammalian target of rapamycin complex 1 (mTORC1) on the lysosomal surface. Activated mTORC1 orchestrates various cellular functions, for example, macromolecule biosynthesis, energy metabolism, autophagy, cell growth, responses to growth factors, and the trafficking and maturation of lysosomes. The Ragulator can also regulate a branch of the MAPK pathway by recruiting MEK1 to MP1/LAMTOR3. These findings suggest that p18/LAMTOR1 creates a core platform for intracellular signaling pathways that function via late endosomes/lysosomes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas de Transporte/metabolismo , Sistema de Sinalização das MAP Quinases , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Fracionamento Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Luminescentes/biossíntese , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Microdomínios da Membrana , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Células PC12 , Mapeamento de Interação de Proteínas , Transporte Proteico , Ratos , Proteínas Recombinantes de Fusão/biossíntese , Transdução de Sinais , Proteína Vermelha Fluorescente
12.
J Cell Sci ; 126(Pt 16): 3575-84, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23781028

RESUMO

The lysosomal adaptor protein p18 is an essential anchor of a scaffolding complex for the mTORC1 and MAPK pathways, which play crucial roles in controlling cell growth and energy homeostasis. To elucidate the in vivo function of the p18-mediated pathway, we conditionally ablated p18 in the mouse epidermis. Mutant mice were born with severe defects in formation of the stratum corneum and died within 12 h after birth due to dehydration caused by loss of skin barrier function. Mutant epidermal cells can grow and differentiate into granular cells, but exhibit functional defects in corneocyte maturation. Electron microscopy identified abnormal immature cells, overlying the mutant granular cells, which accumulated autophagosomes, glycogen granules and dead nuclei. Cell culture analysis showed that loss of p18 attenuated lysosome function, resulting in accumulation of immature lysosomes and autophagosomes. Analyses of lysosome behavior revealed that p18 is required for functional interaction between lysosomes and target organelles including autophagosomes. These findings suggest that p18-mediated pathways control lysosome-mediated catabolic processes, which are crucial for the development of mouse epidermis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Lisossomos/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Recém-Nascidos , Autofagia/fisiologia , Células Epidérmicas , Homeostase , Queratinócitos/citologia , Queratinócitos/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Nucleosídeo NM23 Difosfato Quinases/genética , Transdução de Sinais
13.
J Biol Chem ; 286(37): 32651-60, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21784859

RESUMO

Mammalian target of rapamycin (mTOR) is a kinase that plays a key role in a wide array of cellular processes and exists in two distinct functional complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Although mTORC2 is primarily activated by growth factors, mTORC1 is regulated by numerous extracellular and intracellular signals such as nutrients, growth factors, and cellular redox. Previous study has shown that cysteine oxidants sufficiently activate mTORC1 activity under amino acid-depleted conditions and that a reducing agent effectively suppresses amino acid-induced mTORC1 activity, thereby raising the possibility that redox-sensitive mechanisms underlie amino acid-dependent mTORC1 regulation. However, the molecular mechanism by which redox regulates mTORC1 activity is not well understood. In this study, we show that the redox-sensitive regulation of mTORC1 occurs via Rheb but not the Rag small GTPase. Enhancing cellular redox potential with cysteine oxidants significantly increases Rheb GTP levels. Importantly, modulation of the cellular redox potential with a cysteine oxidant or reducing agent failed to alter mTORC1 activity in TSC1(-/-) or TSC2(-/-) mouse embryonic fibroblast cells. Furthermore, a cysteine oxidant has little effect on mTOR localization but sufficiently activates mTORC1 activity in both p18(-/-) and control mouse embryonic fibroblast cells, suggesting that the redox-sensitive regulation of mTORC1 occurs independent of the Ragulator·Rag complex. Taken together, our results suggest that the TSC complex plays an important role in redox-sensitive mTORC1 regulation and argues for the activation of mTORC1 in places other than the lysosome upon inhibition of the TSC complex.


Assuntos
Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuropeptídeos/metabolismo , Proteínas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Lisossomos/genética , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Complexos Multiproteicos , Neuropeptídeos/genética , Oxirredução , Transporte Proteico/fisiologia , Proteínas/genética , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
14.
J Cell Sci ; 124(Pt 10): 1726-38, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21525037

RESUMO

Podosomes and invadopodia are actin-rich membrane protrusions that play a crucial role in cell adhesion and migration, and extracellular matrix remodeling in normal and cancer cells. The formation of podosomes and invadopodia is promoted by upregulation of some oncogenic molecules and is closely related to the invasive potential of cancer cells. However, the molecular mechanisms underlying the podosome and invadopodium formation still remain unclear. Here, we show that a guanine nucleotide exchange factor (GEF) for Rho family GTPases (Arhgef5) is crucial for Src-induced podosome formation. Using an inducible system for Src activation, we found that Src-induced podosome formation depends upon the Src SH3 domain, and identified Arhgef5 as a Src SH3-binding protein. RNA interference (RNAi)-mediated depletion of Arhgef5 caused robust inhibition of Src-dependent podosome formation. Overexpression of Arhgef5 promoted actin stress fiber remodeling through activating RhoA, and the activation of RhoA or Cdc42 was required for Src-induced podosome formation. Arhgef5 was tyrosine-phosphorylated by Src and bound to Src to positively regulate its activity. Furthermore, the pleckstrin homology (PH) domain of Arhgef5 was required for podosome formation, and Arhgef5 formed a ternary complex with Src and phosphoinositide 3-kinase when Src and/or Arhgef5 were upregulated. These findings provide novel insights into the molecular mechanisms of podosome and invadopodium formation induced by Src upregulation.


Assuntos
Extensões da Superfície Celular/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Quinases da Família src/metabolismo , Animais , Adesão Celular , Ativação Enzimática , Camundongos , Células NIH 3T3 , Fosfatidilinositol 3-Quinases/metabolismo , Ligação Proteica , Fatores de Troca de Nucleotídeo Guanina Rho , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP , Domínios de Homologia de src
15.
Mol Biol Cell ; 19(9): 3801-11, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18596233

RESUMO

For the zonula adherens (ZA) to be established by linear arrangement of adherens junctions (AJs) in epithelial sheet cells, critical for the epithelial cell sheet formation and intercellular barrier function, myosin-2 is supposedly integrated into the ZA with the result of overlapping localization of E-cadherin/actin/myosin-2. Here, we immunofluorescently showed that myosin-2 failed to be integrated into the ZA in cultured epithelial-type ZO1(ko)/2(kd) Eph4 cells lacking ZO-1 and -2 (zonula occludens-1 and -2) by knockout and knockdown, respectively. Instead, a linearized but fragmented arrangement of AJs was formed in the way that it was positive for E-cadherin/actin, but negative for myosin-2 (designated prezonula-AJ). Transfection of full-length ZO-1 or ZO-2, or ZO-1 lacking its PDZ (PSD-95/discs large/zonula occludens-1)-1/2 domains (but not one lacking PDZ-1/2/3) into ZO1(ko)/2(kd) Eph4 cells restored the junctional integration of myosin-2 with prezonula-AJ to establish the ZA. Transfection of dominant-active RhoA or Rho-kinase (ROCK), as well as administration of lysophosphatidic acid or Y27632, which activates RhoA or inhibits ROCK, respectively, suggested that RhoA regulated the junctional integration of myosin-2 into ZA in a manner such that ROCK played a necessary but not-sufficient role. Fluorescence resonance energy transfer analyses revealed that spatiotemporal Rho-activation occurred in a ZO-1/2-dependent way to establish ZA from primordial forms in epithelial cells.


Assuntos
Junções Aderentes/metabolismo , Epitélio/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Miosinas/metabolismo , Fosfoproteínas/metabolismo , Actinas/metabolismo , Amidas/farmacologia , Animais , Caderinas/metabolismo , Células Epiteliais , Transferência Ressonante de Energia de Fluorescência , Camundongos , Modelos Biológicos , Piridinas/farmacologia , Ratos , Proteína da Zônula de Oclusão-1 , Proteína da Zônula de Oclusão-2
16.
Mol Cell ; 30(4): 426-36, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18498747

RESUMO

The tyrosine kinase c-Src is upregulated in various human cancers irrespective of its negative regulator Csk, but the regulatory mechanisms remain unclear. Here, we show that a lipid raft-anchored Csk adaptor, Cbp/PAG, is directly involved in controlling the oncogenicity of c-Src. Using Csk-deficient cells that can be transformed by c-Src overexpression, we found that Cbp expression is markedly downregulated by c-Src activation and re-expression of Cbp efficiently suppresses c-Src transformation as well as tumorigenesis. Cbp-deficient cells are more susceptible to v-Src transformation than their parental cells. Upon phosphorylation, Cbp specifically binds to activated c-Src and sequesters it in lipid rafts, resulting in an efficient suppression of c-Src function independent of Csk. In some human cancer cells and tumors, Cbp is downregulated and the introduction of Cbp significantly suppresses tumorigenesis. These findings indicate a potential role for Cbp as a suppressor of c-Src-mediated tumor progression.


Assuntos
Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Proteína Tirosina Quinase CSK , Fracionamento Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/fisiologia , Técnicas de Transferência de Genes , Humanos , Microdomínios da Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , Neoplasias/metabolismo , Fosfoproteínas/genética , Proteínas Tirosina Quinases/genética , Quinases da Família src
17.
Dev Biol ; 315(2): 474-88, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18262517

RESUMO

Src family kinases (SFKs) have been implicated in the regulation of cell motility. To verify their in vivo roles during development, we generated mutant mice in which Csk, a negative regulator of SFKs, was inactivated in neural crest lineages using the Protein zero promoter in a Cre-loxP system. Inactivation of Csk caused deformities in various tissues of neural crest origins, including facial dysplasia and corneal opacity. In the cornea, the stromal collagen fibril was disorganized and there was an overproduction of collagen 1a1 and several metalloproteases. The corneal endothelium failed to overlie the central region of the eye and the peripheral endothelium displayed a disorganized cytoskeleton. Corneal mesenchymal cells cultured from mutant mice showed attenuated cell motility. In these cells, p130 Crk-associated substrate (Cas) was hyperphosphorylated and markedly downregulated. The expression of a dominant negative Cas (Cas Delta SD) could suppress the cell motility defects. Fluorescence resonance energy transfer analysis revealed that activation of Rac1 and Cdc42 was depolarized in Csk-inactivated cells, which was restored by the expression of either Csk or Cas Delta SD. These results demonstrate that the SFKs/Csk circuit plays crucial roles in corneal development by controlling stromal organization and by ensuring cell motility via the Cas-Rac/Cdc42 pathways.


Assuntos
Colágeno/metabolismo , Córnea/anormalidades , Crista Neural/citologia , Crista Neural/enzimologia , Proteínas Tirosina Quinases/deficiência , Animais , Proteína Tirosina Quinase CSK , Movimento Celular/fisiologia , Córnea/citologia , Córnea/embriologia , Córnea/metabolismo , Transferência Ressonante de Energia de Fluorescência , Mesoderma/citologia , Mesoderma/embriologia , Mesoderma/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Proteína P0 da Mielina/genética , Fenótipo , Regiões Promotoras Genéticas , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/fisiologia , Interferência de RNA , Proteína p130 Retinoblastoma-Like/metabolismo , Quinases da Família src
18.
Genes Cells ; 13(1): 1-12, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18173743

RESUMO

The c-src proto-oncogene product, c-Src, is frequently over-expressed and activated in various human malignant cancers, implicating a role for c-Src in cancer progression. To verify the role of c-Src, we analyzed the transforming ability of c-Src in mouse embryonic fibroblasts that lack Csk, a negative regulator of Src family kinases. Although Csk deficiency is not sufficient for cell transformation, c-Src over-expression induced characteristic transformed phenotypes including anchorage-independent growth and tumorigenecity. These phenotypes were dose-dependently inhibited by the re-expression of Csk, indicating that there is a certain threshold for c-Src transformation, which is determined by the c-Src : Csk ratio. In contrast to v-Src, c-Src induced the phosphorylation of a limited number of cellular proteins and elicited a restricted change in gene expression profiles. The activation of some critical targets for v-Src transformation, such as STAT3, was not significantly induced by c-Src transformation. Several genes that are involved in cancer progression, that is, cyclin D1 and HIF-1alpha, were induced by v-Src, but not by c-Src. Furthermore, v-Src tumors exhibited aggressive growth and extensive angiogenesis, while c-Src tumors grew more slowly accompanied by the induction of hematomas. These findings demonstrate that c-Src has the potential to induce cell transformation, but it requires coordination with an additional pathway(s) to promote tumor progression in vivo.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fibroblastos/citologia , Proteína Oncogênica pp60(v-src)/fisiologia , Proteínas Tirosina Quinases/fisiologia , Animais , Proteína Tirosina Quinase CSK , Transformação Celular Neoplásica/patologia , Ciclina D1/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Hematoma/metabolismo , Hematoma/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Proteínas Tirosina Quinases/genética , Proto-Oncogene Mas , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Transplante Heterólogo , Quinases da Família src
19.
EMBO J ; 26(5): 1234-44, 2007 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-17304209

RESUMO

Carboxy-terminal Src kinase (Csk) is a negative regulator of Src family kinases, which play pivotal roles in controlling cell adhesion, migration, and cancer progression. To elucidate the in vivo role of Csk in epithelial tissues, we conditionally inactivated Csk in squamous epithelia using the keratin-5 promoter/Cre-loxP system in mice. The mutant mice developed apparent defects in the skin, esophagus, and forestomach, with concomitant hyperplasia and chronic inflammation. Histology of the mutant epidermis revealed impaired cell-cell adhesion in basal cell layers. Analysis of primary keratinocytes showed that the defective cell-cell adhesion was caused by cytoskeletal remodeling via activation of the Rac1 pathway. Mutant keratinocytes also showed elevated expression of mesenchymal proteins, matrix metalloproteinases (MMPs), and the proinflammatory cytokine TNF-alpha. Inhibition of the expression of TNF-alpha and MMP9 by the anti-inflammatory reagent FK506 could cure the epidermal hyperplasia, suggesting a causal link between inflammation and epidermal hyperplasia. These observations demonstrate that the Src/Csk circuit plays crucial roles in development and maintenance of epithelia by controlling cytoskeletal organization as well as phenotypic conversion linked to inflammatory events.


Assuntos
Queratinócitos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Pele/metabolismo , Animais , Proteína Tirosina Quinase CSK , Adesão Celular , Células Cultivadas , Citoesqueleto/metabolismo , Esôfago/citologia , Esôfago/metabolismo , Immunoblotting , Imuno-Histoquímica , Imunossupressores/farmacologia , Inflamação/metabolismo , Inflamação/patologia , Queratina-5/genética , Queratina-5/metabolismo , Queratinócitos/citologia , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mutação , Fenótipo , Proteínas Tirosina Quinases/genética , Transdução de Sinais/efeitos dos fármacos , Pele/citologia , Pele/ultraestrutura , Tacrolimo/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
20.
Neurosci Res ; 57(2): 210-9, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17137665

RESUMO

Src family tyrosine kinases are essential for neural development, but their in vivo functions remain elusive because of functional compensation among family members. To elucidate the roles of individual Src family members in vivo, we generated transgenic mice expressing the neuronal form of c-Src (n-Src), Fyn, and their constitutively active forms in cerebellar Purkinje cells using the L7 promoter. The expression of the constitutively active n-Src retarded the postnatal development of Purkinje cells and disrupted dendritic morphogenesis, whereas the wild-type n-Src had only moderate effects. Neither wild-type nor constitutively active Fyn over-expression significantly affected Purkinje-cell morphology. The aberrant Purkinje cells in n-Src transgenic mice retained multiple dendritic shafts extending in non-polarized directions and were located heterotopically in the molecular layer. Ultrastructural observation of the dendritic shafts revealed that the microtubules of n-Src transgenic mice were more densely and irregularly arranged, and had structural deformities. In primary culture, Purkinje cells from n-Src transgenic mice developed abnormally thick dendritic shafts and large growth-cone-like structures with poorly extended dendrites, which could be rescued by treatment with a selective inhibitor of Src family kinases, PP2. These results suggest that n-Src activity regulates the dendritic morphogenesis of Purkinje cells through affecting microtubule organization.


Assuntos
Cerebelo/citologia , Dendritos/fisiologia , Genes src/fisiologia , Células de Purkinje/patologia , Animais , Animais Recém-Nascidos , Calbindinas , Células Cultivadas , Dendritos/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes src/genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão/métodos , Proteínas do Tecido Nervoso/fisiologia , Proteínas Proto-Oncogênicas c-fyn/genética , Células de Purkinje/ultraestrutura , Proteína G de Ligação ao Cálcio S100/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA