Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cell ; 152(1-2): 290-303, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23332761

RESUMO

Autophagy is a stress response protecting cells from unfavorable conditions, such as nutrient starvation. The class III phosphatidylinositol-3 kinase, Vps34, forms multiple complexes and regulates both intracellular vesicle trafficking and autophagy induction. Here, we show that AMPK plays a key role in regulating different Vps34 complexes. AMPK inhibits the nonautophagy Vps34 complex by phosphorylating T163/S165 in Vps34 and therefore suppresses overall PI(3)P production and protects cells from starvation. In parallel, AMPK activates the proautophagy Vps34 complex by phosphorylating S91/S94 in Beclin1 to induce autophagy. Atg14L, an autophagy-essential gene present only in the proautophagy Vps34 complex, inhibits Vps34 phosphorylation but increases Beclin1 phosphorylation by AMPK. As such, Atg14L dictates the differential regulation (either inhibition or activation) of different Vps34 complexes in response to glucose starvation. Our study reveals an intricate molecular regulation of Vps34 complexes by AMPK in nutrient stress response and autophagy.


Assuntos
Autofagia , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Classe III de Fosfatidilinositol 3-Quinases/genética , Glucose/metabolismo , Camundongos , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Fosforilação , Proteínas Quinases/química , Proteínas Quinases/genética , Alinhamento de Sequência , Proteínas de Transporte Vesicular/metabolismo
2.
EMBO J ; 41(13): e110031, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35535466

RESUMO

Autophagy is a cellular degradative pathway that plays diverse roles in maintaining cellular homeostasis. Cellular stress caused by starvation, organelle damage, or proteotoxic aggregates can increase autophagy, which uses the degradative capacity of lysosomal enzymes to mitigate intracellular stresses. Early studies have shown a role for autophagy in the suppression of tumorigenesis. However, work in genetically engineered mouse models and in vitro cell studies have now shown that autophagy can be either cancer-promoting or inhibiting. Here, we summarize the effects of autophagy on cancer initiation, progression, immune infiltration, and metabolism. We also discuss the efforts to pharmacologically target autophagy in the clinic and highlight future areas for exploration.


Assuntos
Autofagia , Neoplasias , Animais , Carcinogênese , Transformação Celular Neoplásica , Homeostase , Camundongos
3.
Trends Biochem Sci ; 46(8): 687-700, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33593593

RESUMO

Autophagy is the primary catabolic program of the cell that promotes survival in response to metabolic stress. It is tightly regulated by a suite of kinases responsive to nutrient status, including mammalian target of rapamycin complex 1 (mTORC1), AMP-activated protein kinase (AMPK), protein kinase C-α (PKCα), MAPK-activated protein kinases 2/3 (MAPKAPK2/3), Rho kinase 1 (ROCK1), c-Jun N-terminal kinase 1 (JNK), and Casein kinase 2 (CSNK2). Here, we highlight recently uncovered mechanisms linking amino acid, glucose, and oxygen levels to autophagy regulation through mTORC1 and AMPK. In addition, we describe new pathways governing the autophagic machinery, including the Unc-51-like (ULK1), vacuolar protein sorting 34 (VPS34), and autophagy related 16 like 1 (ATG16L1) enzyme complexes. Novel downstream targets of ULK1 protein kinase are also discussed, such as the ATG16L1 subunit of the microtubule-associated protein 1 light chain 3 (LC3)-lipidating enzyme and the ATG14 subunit of the VPS34 complex. Collectively, we describe the complexities of the autophagy pathway and its role in maintaining cellular nutrient homeostasis during times of starvation.


Assuntos
Autofagia , Serina-Treonina Quinases TOR , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Nutrientes , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
4.
Nat Rev Mol Cell Biol ; 14(3): 133-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23361334

RESUMO

Mammalian target of rapamycin (mTOR) is a conserved Ser/Thr kinase that is part of mTOR complex 1 (mTORC1), a master regulator that couples amino acid availability to cell growth and autophagy. Multiple cues modulate mTORC1 activity, such as growth factors, stress, energy status and amino acids. Although amino acids are key environmental stimuli, exactly how they are sensed and how they activate mTORC1 is not fully understood. Recently, a model has emerged whereby mTORC1 activation occurs at the lysosome and is mediated through an amino acid sensing cascade involving RAG GTPases, Ragulator and vacuolar H(+)-ATPase (v-ATPase).


Assuntos
Aminoácidos/metabolismo , Lisossomos/metabolismo , Proteínas/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia , Proliferação de Células , Humanos , Leucina-tRNA Ligase/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Modelos Biológicos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos , Neuropeptídeos/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo
5.
Nat Methods ; 17(2): 232-239, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31768061

RESUMO

Autophagy is a degradative program that maintains cellular homeostasis. Autophagy defects have been described in numerous diseases. However, analysis of autophagy rates can be challenging, particularly in rare cell populations or in vivo, due to limitations in currently available tools for measuring autophagy induction. Here, we describe a method to monitor autophagy by measuring phosphorylation of the protein ATG16L1. We developed and characterized a monoclonal antibody that can detect phospho-ATG16L1 endogenously in mammalian cells. Importantly, phospho-ATG16L1 is only present on newly forming autophagosomes. Therefore, its levels are not affected by prolonged stress or late-stage autophagy blocks, which can confound autophagy analysis. Moreover, we show that ATG16L1 phosphorylation is a conserved signaling pathway activated by numerous autophagy-inducing stressors. The described antibody is suitable for western blot, immunofluorescence and immunohistochemistry, and measured phospho-ATG16L1 levels directly correspond to autophagy rates. Taken together, this phospho-antibody represents an exciting tool to study autophagy induction.


Assuntos
Anticorpos/imunologia , Autofagia , Animais , Proteínas de Transporte/metabolismo , Humanos , Fosforilação
6.
Genes Dev ; 29(22): 2362-76, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26588989

RESUMO

The mechanistic target of rapamycin (mTOR) is a central cell growth controller and forms two distinct complexes: mTORC1 and mTORC2. mTORC1 integrates a wide range of upstream signals, both positive and negative, to regulate cell growth. Although mTORC1 activation by positive signals, such as growth factors and nutrients, has been extensively investigated, the mechanism of mTORC1 regulation by stress signals is less understood. In this study, we identified the Nemo-like kinase (NLK) as an mTORC1 regulator in mediating the osmotic and oxidative stress signals. NLK inhibits mTORC1 lysosomal localization and thereby suppresses mTORC1 activation. Mechanistically, NLK phosphorylates Raptor on S863 to disrupt its interaction with the Rag GTPase, which is important for mTORC1 lysosomal recruitment. Cells with Nlk deletion or knock-in of the Raptor S863 phosphorylation mutants are defective in the rapid mTORC1 inhibition upon osmotic stress. Our study reveals a function of NLK in stress-induced mTORC1 modulation and the underlying biochemical mechanism of NLK in mTORC1 inhibition in stress response.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multiproteicos/metabolismo , Pressão Osmótica/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Ativação Enzimática , Deleção de Genes , Técnicas de Introdução de Genes , Células HEK293 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteínas Quinases Ativadas por Mitógeno/genética , Fosforilação , Proteína Regulatória Associada a mTOR
7.
EMBO Rep ; 20(7): e46885, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31267703

RESUMO

Autophagy is a highly regulated catabolic pathway that is potently induced by stressors including starvation and infection. An essential component of the autophagy pathway is an ATG16L1-containing E3-like enzyme, which is responsible for lipidating LC3B and driving autophagosome formation. ATG16L1 polymorphisms have been linked to the development of Crohn's disease (CD), and phosphorylation of CD-associated ATG16L1 T300A (caATG16L1) has been hypothesized to contribute to cleavage and autophagy dysfunction. Here we show that ULK1 kinase directly phosphorylates ATG16L1 in response to infection and starvation. Phosphorylated ATG16L1 localizes to the site of internalized bacteria and stable cell lines harbouring a phospho-dead mutant of ATG16L1 have impaired xenophagy, indicating a role for ATG16L1 phosphorylation in the promotion of anti-bacterial autophagy. In contrast to wild-type ATG16L1, ULK1-mediated phosphorylation of caATG16L1 drives its destabilization in response to stress. In summary, our results show that ATG16L1 is a novel target of ULK1 kinase and that ULK1 signalling to ATG16L1 is a double-edged sword, enhancing the function of the wild-type ATG16L1, but promoting degradation of caATG16L1.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Doença de Crohn/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macroautofagia , Mutação , Animais , Proteínas Relacionadas à Autofagia/genética , Células HCT116 , Células HEK293 , Humanos , Camundongos , Fosforilação , Estabilidade Proteica , Estresse Fisiológico
8.
Development ; 138(16): 3343-56, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21791526

RESUMO

The mammalian target of rapamycin (mTOR) is a kinase that responds to a myriad of signals, ranging from nutrient availability and energy status, to cellular stressors, oxygen sensors and growth factors. The finely tuned response of mTOR to these stimuli results in alterations to cell metabolism and cell growth. Recent studies of conditional knockouts of mTOR pathway components in mice have affirmed the role of mTOR signaling in energy balance, both at the cell and whole organism levels. Such studies have also highlighted a role for mTOR in stem cell homeostasis and lifespan determination. Here, we discuss the molecular mechanisms of TOR signaling and review recent in vitro and in vivo studies of mTOR tissue-specific activities in mammals.


Assuntos
Transdução de Sinais , Células-Tronco/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Proliferação de Células , Homeostase , Humanos , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Serina-Treonina Quinases TOR/antagonistas & inibidores
9.
Proc (Bayl Univ Med Cent) ; 36(6): 687-691, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37829233

RESUMO

Background: Prior studies have shown that programmed intermittent epidural bolus (PIEB) techniques, with or without patient-controlled epidural analgesia (PCEA) boluses, provide better pain relief, reduced motor block, and better patient satisfaction compared to continuous epidural infusion (CEI) techniques. We hypothesized that patients who had labor epidural analgesia (LEA) maintained with PIEB and PCEA would be less likely to receive a physician-administered rescue analgesia bolus compared to patients who had CEI and PCEA. Methods: We searched our electronic medical record for patients who had CEI and PCEA from August 1, 2021 to December 31, 2021 and for patients who had PIEB and PCEA from August 2, 2022 to December 31, 2022. Results: A total of 792 and 665 patients had maintenance of LEA with CEI/PCEA and PIEB/PCEA, respectively. A multivariate logistic regression was performed and, after adjusting for variables of interest, patients who had PIEB and PCEA were less likely to receive one or more physician-administered rescue analgesia boluses (odds ratio 0.504; 95% confidence interval 0.392, 0.649; P < 0.001) compared to patients who had CEI and PCEA. Conclusion: PIEB/PCEA was associated with fewer physician-administered boluses of rescue analgesia compared to CEI/PCEA when used for LEA.

10.
Cell Death Differ ; 29(3): 585-599, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34588632

RESUMO

Mutations in susceptibility alleles correlate with gut-inflammatory diseases, such as Crohn's disease; however, this does not often impact the disease progression indicating the existence of compensatory genes. We show that a reduction in Foxo3a expression in IL-10-deficient mice results in a spontaneous and aggressive Crohn's- like disease with 100% penetrance, which is rescued by deletion of myeloid cells, T cells and inhibition of mTORC1. In Foxo3a-/- IL-10-/- mice, there is poor cell death of myeloid cells in the gut, leading to increased accumulation of myeloid and T cells in the gut. Myeloid cells express high levels of inflammatory cytokines, and regulatory T cells are dysfunctional despite increased abundance. Foxo3a signaling represses the transcription of glutaminase (GLS/GLS2) to prevent over-consumption of glutamine by activated T cells and its conversion to glutamate that contributes to the TCA cycle and mTORC1 activation. Finally, we show that Foxo3a restricts the abundance of colitogenic microbiota in IL-10-deficient mice. Thus, by suppressing glutaminolysis in activated T cells Foxo3a mediates a critical checkpoint that prevents the development of fulminant gut inflammatory disease.


Assuntos
Colite , Proteína Forkhead Box O3/metabolismo , Interleucina-10 , Animais , Colite/genética , Colite/prevenção & controle , Inflamação , Interleucina-10/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Linfócitos T
11.
PLoS Genet ; 4(9): e1000176, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18773095

RESUMO

Chromosomal abnormalities, such as structural and numerical abnormalities, are a common occurrence in cancer. The close association of homologous chromosomes during interphase, a phenomenon termed somatic chromosome pairing, has been observed in cancerous cells, but the functional consequences of somatic pairing have not been established. Gene expression profiling studies revealed that somatic pairing of chromosome 19 is a recurrent chromosomal abnormality in renal oncocytoma, a neoplasia of the adult kidney. Somatic pairing was associated with significant disruption of gene expression within the paired regions and resulted in the deregulation of the prolyl-hydroxylase EGLN2 [corrected] a key protein that regulates the oxygen-dependent degradation of hypoxia-inducible factor (HIF). Overexpression of EGLN2 [corrected] in renal oncocytoma increased ubiquitin-mediated destruction of HIF and concomitantly suppressed the expression of several HIF-target genes, including the pro-death BNIP3L gene. The transcriptional changes that are associated with somatic pairing of chromosome 19 mimic the transcriptional changes that occur following DNA amplification. Therefore, in addition to numerical and structural chromosomal abnormalities, alterations in chromosomal spatial dynamics should be considered as genomic events that are associated with tumorigenesis. The identification of EGLN2 as a significantly deregulated gene that maps within the paired chromosome region directly implicates defects in the oxygen-sensing network to the biology of renal oncocytoma.


Assuntos
Adenoma Oxífilo/genética , Adenoma Oxífilo/metabolismo , Pareamento Cromossômico/genética , Cromossomos Humanos Par 19 , Dioxigenases/genética , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Proteínas Nucleares/genética , Oxigênio/metabolismo , Pró-Colágeno-Prolina Dioxigenase/genética , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Hipóxia Celular/genética , Linhagem Celular Tumoral , Cromossomos Humanos Par 19/metabolismo , Dioxigenases/metabolismo , Regulação para Baixo , Perfilação da Expressão Gênica , Humanos , Fator 1 Induzível por Hipóxia/genética , Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia , Proteínas Nucleares/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo
12.
Mol Cell Biol ; 27(1): 157-69, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17060462

RESUMO

The product of the von Hippel-Lindau gene (VHL) acts as the substrate-recognition component of an E3 ubiquitin ligase complex that ubiquitylates the catalytic alpha subunit of hypoxia-inducible factor (HIF) for oxygen-dependent destruction. Although emerging evidence supports the notion that deregulated accumulation of HIF upon the loss of VHL is crucial for the development of clear-cell renal cell carcinoma (CC-RCC), the molecular events downstream of HIF governing renal oncogenesis remain unclear. Here, we show that the expression of a homophilic adhesion molecule, E-cadherin, a major constituent of epithelial cell junctions whose loss is associated with the progression of epithelial cancers, is significantly down-regulated in primary CC-RCC and CC-RCC cell lines devoid of VHL. Reintroduction of wild-type VHL in CC-RCC (VHL(-/-)) cells markedly reduced the expression of E2 box-dependent E-cadherin-specific transcriptional repressors Snail and SIP1 and concomitantly restored E-cadherin expression. RNA interference-mediated knockdown of HIFalpha in CC-RCC (VHL(-/-)) cells likewise increased E-cadherin expression, while functional hypoxia or expression of VHL mutants incapable of promoting HIFalpha degradation attenuated E-cadherin expression, correlating with the disengagement of RNA polymerase II from the endogenous E-cadherin promoter/gene. These findings reveal a critical HIF-dependent molecular pathway connecting VHL, an established "gatekeeper" of the renal epithelium, with a major epithelial tumor suppressor, E-cadherin.


Assuntos
Caderinas/biossíntese , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Proteínas de Ligação a RNA/fisiologia , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor Von Hippel-Lindau/fisiologia , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Humanos , Rim/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Polimerase II/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição da Família Snail , Frações Subcelulares/metabolismo
13.
Autophagy ; 15(8): 1489-1491, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31107135

RESUMO

Macroautophagy/autophagy is a conserved degradative pathway that host cells use to deal with invading pathogens. Despite significant overlap with starvation-induced autophagy, the early signaling that potentiates anti-bacterial autophagy is still unclear. Here we report AMPK, an upstream kinase regulating starvation-mediated autophagy induction, is activated in response to bacterial infection. AMPK inhibits MTORC1, an autophagy repressor, and activates autophagic ULK1 and PIK3C3/VPS34 complexes. Although AMPK-mediated inhibition of MTORC1 is not accompanied by the induction of bulk autophagy, AMPK regulation is critical for selectively targeting the bacteria for degradation. Moreover, AMPK signaling is triggered by the detection of bacteria-derived outer membrane vesicles and does not require bacterial invasion. Together, these data characterize and highlight the significance of AMPK signaling in priming the autophagic response to bacterial infection. Abbreviations: AMPK: AMP-activated protein kinase; MTORC1: MTOR complex 1; ULK1: Unc-51 like kinase 1; PIK3C3/VPS34: Phosphatidylinositol 3-kinase catalytic subunit type 3.


Assuntos
Proteínas Quinases Ativadas por AMP , Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Membrana Externa Bacteriana , Classe III de Fosfatidilinositol 3-Quinases
14.
Cell Rep ; 26(8): 2150-2165.e5, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30784596

RESUMO

The autophagy pathway is an essential facet of the innate immune response, capable of rapidly targeting intracellular bacteria. However, the initial signaling regulating autophagy induction in response to pathogens remains largely unclear. Here, we report that AMPK, an upstream activator of the autophagy pathway, is stimulated upon detection of pathogenic bacteria, before bacterial invasion. Bacterial recognition occurs through the detection of outer membrane vesicles. We found that AMPK signaling relieves mTORC1-mediated repression of the autophagy pathway in response to infection, positioning the cell for a rapid induction of autophagy. Moreover, activation of AMPK and inhibition of mTORC1 in response to bacteria is not accompanied by an induction of bulk autophagy. However, AMPK signaling is required for the selective targeting of bacteria-containing vesicles by the autophagy pathway through the activation of pro-autophagic kinase complexes. These results demonstrate a key role for AMPK signaling in coordinating the rapid autophagic response to bacteria.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Membrana Externa Bacteriana/metabolismo , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Macroautofagia , Proteínas Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Animais , Células Cultivadas , Células HCT116 , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Células MCF-7 , Macrófagos/metabolismo , Macrófagos/microbiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Salmonella/patogenicidade
15.
Sci Signal ; 10(510)2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29259100

RESUMO

Huntington's disease (HD) is a neurodegenerative disease caused by an expansion in the huntingtin protein (also called Htt) that induces neuronal cell death with age. We found that the treatment of 12-month-old symptomatic heterozygous and homozygous zQ175 huntingtin knockin mice for 12 weeks with CTEP, a negative allosteric modulator of metabotropic glutamate receptor 5 (mGluR5), reduced the size and number of huntingtin aggregates, attenuated caspase-3 activity, and reduced both neuronal apoptosis and neuronal loss in brain tissue. Both motor and cognitive impairments were improved in CTEP-treated zQ175 mice. The reduction in huntingtin protein aggregate burden by CTEP correlated with the activation of an autophagy pathway mediated by the kinase GSK3ß, the transcription factor ZBTB16, and the autophagy factor ATG14. Inhibition of mGluR5 with CTEP also reduced the inhibitory phosphorylation of the autophagosome biogenesis-related kinase ULK1, increased the phosphorylation of the autophagy factor ATG13, and increased the abundance of the autophagy-related protein Beclin1 in homozygous zQ175 mice. The findings suggest that mGluR5 antagonism may activate autophagy through convergent mechanisms to promote the clearance of mutant huntingtin aggregates and might be therapeutic in HD patients.


Assuntos
Autofagia/efeitos dos fármacos , Proteína Huntingtina/genética , Doença de Huntington/tratamento farmacológico , Doença de Huntington/genética , Imidazóis/uso terapêutico , Piridinas/uso terapêutico , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Introdução de Genes , Humanos , Proteína Huntingtina/metabolismo , Imidazóis/farmacologia , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Piridinas/farmacologia
16.
Nat Commun ; 6: 8283, 2015 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-26387534

RESUMO

Defective hepatic insulin receptor (IR) signalling is a pathogenic manifestation of metabolic disorders including obesity and diabetes. The endo/lysosomal trafficking system may coordinate insulin action and nutrient homeostasis by endocytosis of IR and the autophagic control of intracellular nutrient levels. Here we show that class III PI3K--a master regulator of endocytosis, endosomal sorting and autophagy--provides negative feedback on hepatic insulin signalling. The ultraviolet radiation resistance-associated gene protein (UVRAG)-associated class III PI3K complex interacts with IR and is stimulated by insulin treatment. Acute and chronic depletion of hepatic Vps15, the regulatory subunit of class III PI3K, increases insulin sensitivity and Akt signalling, an effect that requires functional IR. This is reflected by FoxO1-dependent transcriptional defects and blunted gluconeogenesis in Vps15 mutant cells. On depletion of Vps15, the metabolic syndrome in genetic and diet-induced models of insulin resistance and diabetes is alleviated. Thus, feedback regulation of IR trafficking and function by class III PI3K may be a therapeutic target in metabolic conditions of insulin resistance.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Proteína VPS15 de Distribuição Vacuolar/metabolismo , Animais , Diabetes Mellitus/metabolismo , Retroalimentação Fisiológica , Homeostase , Humanos , Resistência à Insulina , Fígado/enzimologia , Masculino , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteína VPS15 de Distribuição Vacuolar/genética
17.
Science ; 347(6218): 194-8, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25567907

RESUMO

The mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) integrates environmental and intracellular signals to regulate cell growth. Amino acids stimulate mTORC1 activation at the lysosome in a manner thought to be dependent on the Rag small guanosine triphosphatases (GTPases), the Ragulator complex, and the vacuolar H(+)-adenosine triphosphatase (v-ATPase). We report that leucine and glutamine stimulate mTORC1 by Rag GTPase-dependent and -independent mechanisms, respectively. Glutamine promoted mTORC1 translocation to the lysosome in RagA and RagB knockout cells and required the v-ATPase but not the Ragulator. Furthermore, we identified the adenosine diphosphate ribosylation factor-1 GTPase to be required for mTORC1 activation and lysosomal localization by glutamine. Our results uncover a signaling cascade to mTORC1 activation independent of the Rag GTPases and suggest that mTORC1 is differentially regulated by specific amino acids.


Assuntos
Glutamina/metabolismo , Leucina/metabolismo , Lisossomos/enzimologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator 1 de Ribosilação do ADP/genética , Fator 1 de Ribosilação do ADP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Ativação Enzimática , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Transporte Proteico , Proteína Regulatória Associada a mTOR
18.
Cell Res ; 24(1): 42-57, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24343578

RESUMO

The ability of cells to respond to changes in nutrient availability is essential for the maintenance of metabolic homeostasis and viability. One of the key cellular responses to nutrient withdrawal is the upregulation of autophagy. Recently, there has been a rapid expansion in our knowledge of the molecular mechanisms involved in the regulation of mammalian autophagy induction in response to depletion of key nutrients. Intracellular amino acids, ATP, and oxygen levels are intimately tied to the cellular balance of anabolic and catabolic processes. Signaling from key nutrient-sensitive kinases mTORC1 and AMP-activated protein kinase (AMPK) is essential for the nutrient sensing of the autophagy pathway. Recent advances have shown that the nutrient status of the cell is largely passed on to the autophagic machinery through the coordinated regulation of the ULK and VPS34 kinase complexes. Identification of extensive crosstalk and feedback loops converging on the regulation of ULK and VPS34 can be attributed to the importance of these kinases in autophagy induction and maintaining cellular homeostasis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/fisiologia , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Homeostase/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Transdução de Sinais
19.
Nat Commun ; 5: 4241, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24980141

RESUMO

The Rag family proteins are Ras-like small GTPases that have a critical role in amino-acid-stimulated mTORC1 activation by recruiting mTORC1 to lysosome. Despite progress in the mechanistic understanding of Rag GTPases in mTORC1 activation, little is known about the physiological function of Rag GTPases in vivo. Here we show that loss of RagA and RagB (RagA/B) in cardiomyocytes results in hypertrophic cardiomyopathy and phenocopies lysosomal storage diseases, although mTORC1 activity is not substantially impaired in vivo. We demonstrate that despite upregulation of lysosomal protein expression by constitutive activation of the transcription factor EB (TFEB) in RagA/B knockout mouse embryonic fibroblasts, lysosomal acidification is compromised owing to decreased v-ATPase level in the lysosome fraction. Our study uncovers RagA/B GTPases as key regulators of lysosomal function and cardiac protection.


Assuntos
Cardiomiopatia Hipertrófica/enzimologia , Lisossomos/enzimologia , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Animais , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/prevenção & controle , Feminino , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Miócitos Cardíacos/enzimologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
20.
Autophagy ; 9(12): 1983-95, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24013218

RESUMO

Autophagy is a cellular defense response to stress conditions, such as nutrient starvation. The type III phosphatidylinositol (PtdIns) 3-kinase, whose catalytic subunit is PIK3C3/VPS34, plays a critical role in intracellular membrane trafficking and autophagy induction. PIK3C3 forms multiple complexes and the ATG14-containing PIK3C3 is specifically involved in autophagy induction. Mechanistic target of rapamycin (MTOR) complex 1, MTORC1, is a key cellular nutrient sensor and integrator to stimulate anabolism and inhibit catabolism. Inactivation of TORC1 by nutrient starvation plays a critical role in autophagy induction. In this report we demonstrated that MTORC1 inactivation is critical for the activation of the autophagy-specific (ATG14-containing) PIK3C3 kinase, whereas it has no effect on ATG14-free PIK3C3 complexes. MTORC1 inhibits the PtdIns 3-kinase activity of ATG14-containing PIK3C3 by phosphorylating ATG14, which is required for PIK3C3 inhibition by MTORC1 both in vitro and in vivo. Our data suggest a mechanistic link between amino acid starvation and autophagy induction via the direct activation of the autophagy-specific PIK3C3 kinase.


Assuntos
Autofagia , Classe III de Fosfatidilinositol 3-Quinases/metabolismo , Fome/fisiologia , Estresse Fisiológico/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Aminoácidos/deficiência , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Células Cultivadas , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Complexos Multiproteicos/metabolismo , Fagossomos/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa