Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.803
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 178(5): 1102-1114.e17, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31442403

RESUMO

Caloric restriction is known to improve inflammatory and autoimmune diseases. However, the mechanisms by which reduced caloric intake modulates inflammation are poorly understood. Here we show that short-term fasting reduced monocyte metabolic and inflammatory activity and drastically reduced the number of circulating monocytes. Regulation of peripheral monocyte numbers was dependent on dietary glucose and protein levels. Specifically, we found that activation of the low-energy sensor 5'-AMP-activated protein kinase (AMPK) in hepatocytes and suppression of systemic CCL2 production by peroxisome proliferator-activator receptor alpha (PPARα) reduced monocyte mobilization from the bone marrow. Importantly, we show that fasting improves chronic inflammatory diseases without compromising monocyte emergency mobilization during acute infectious inflammation and tissue repair. These results reveal that caloric intake and liver energy sensors dictate the blood and tissue immune tone and link dietary habits to inflammatory disease outcome.


Assuntos
Restrição Calórica , Monócitos/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Adulto , Animais , Antígenos Ly/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Quimiocina CCL2/deficiência , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Feminino , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , PPAR alfa/deficiência , PPAR alfa/genética , PPAR alfa/metabolismo
2.
Immunity ; 57(5): 1087-1104.e7, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38640930

RESUMO

Macrophages are critical to turn noninflamed "cold tumors" into inflamed "hot tumors". Emerging evidence indicates abnormal cholesterol metabolites in the tumor microenvironment (TME) with unclear function. Here, we uncovered the inducible expression of cholesterol-25-hydroxylase (Ch25h) by interleukin-4 (IL-4) and interleukin-13 (IL-13) via the transcription factor STAT6, causing 25-hydroxycholesterol (25HC) accumulation. scRNA-seq analysis confirmed that CH25Hhi subsets were enriched in immunosuppressive macrophage subsets and correlated to lower survival rates in pan-cancers. Targeting CH25H abrogated macrophage immunosuppressive function to enhance infiltrating T cell numbers and activation, which synergized with anti-PD-1 to improve anti-tumor efficacy. Mechanically, lysosome-accumulated 25HC competed with cholesterol for GPR155 binding to inhibit the kinase mTORC1, leading to AMPKα activation and metabolic reprogramming. AMPKα also phosphorylated STAT6 Ser564 to enhance STAT6 activation and ARG1 production. Together, we propose CH25H as an immunometabolic checkpoint, which manipulates macrophage fate to reshape CD8+ T cell surveillance and anti-tumor response.


Assuntos
Hidroxicolesteróis , Lisossomos , Macrófagos , Microambiente Tumoral , Animais , Hidroxicolesteróis/metabolismo , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Humanos , Lisossomos/metabolismo , Microambiente Tumoral/imunologia , Fator de Transcrição STAT6/metabolismo , Adenilato Quinase/metabolismo , Camundongos Endogâmicos C57BL , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Transdução de Sinais , Reprogramação Metabólica
3.
Cell ; 172(4): 731-743.e12, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29425491

RESUMO

The noncanonical IKK family member TANK-binding kinase 1 (TBK1) is activated by pro-inflammatory cytokines, but its role in controlling metabolism remains unclear. Here, we report that the kinase uniquely controls energy metabolism. Tbk1 expression is increased in adipocytes of HFD-fed mice. Adipocyte-specific TBK1 knockout (ATKO) attenuates HFD-induced obesity by increasing energy expenditure; further studies show that TBK1 directly inhibits AMPK to repress respiration and increase energy storage. Conversely, activation of AMPK under catabolic conditions can increase TBK1 activity through phosphorylation, mediated by AMPK's downstream target ULK1. Surprisingly, ATKO also exaggerates adipose tissue inflammation and insulin resistance. TBK1 suppresses inflammation by phosphorylating and inducing the degradation of the IKK kinase NIK, thus attenuating NF-κB activity. Moreover, TBK1 mediates the negative impact of AMPK activity on NF-κB activation. These data implicate a unique role for TBK1 in mediating bidirectional crosstalk between energy sensing and inflammatory signaling pathways in both over- and undernutrition.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Metabolismo Energético , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/patologia , Tecido Adiposo/patologia , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Linhagem Celular Transformada , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Serina-Treonina Quinases/genética , Quinase Induzida por NF-kappaB
4.
Mol Cell ; 84(6): 1120-1138.e8, 2024 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-38377992

RESUMO

UFMylation is an emerging ubiquitin-like post-translational modification that regulates various biological processes. Dysregulation of the UFMylation pathway leads to human diseases, including cancers. However, the physiological role of UFMylation in T cells remains unclear. Here, we report that mice with conditional knockout (cKO) Ufl1, a UFMylation E3 ligase, in T cells exhibit effective tumor control. Single-cell RNA sequencing analysis shows that tumor-infiltrating cytotoxic CD8+ T cells are increased in Ufl1 cKO mice. Mechanistically, UFL1 promotes PD-1 UFMylation to antagonize PD-1 ubiquitination and degradation. Furthermore, AMPK phosphorylates UFL1 at Thr536, disrupting PD-1 UFMylation to trigger its degradation. Of note, UFL1 ablation in T cells reduces PD-1 UFMylation, subsequently destabilizing PD-1 and enhancing CD8+ T cell activation. Thus, Ufl1 cKO mice bearing tumors have a better response to anti-CTLA-4 immunotherapy. Collectively, our findings uncover a crucial role of UFMylation in T cells and highlight UFL1 as a potential target for cancer treatment.


Assuntos
Neoplasias , Receptor de Morte Celular Programada 1 , Animais , Humanos , Camundongos , Linfócitos T CD8-Positivos/metabolismo , Neoplasias/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
5.
Mol Cell ; 84(10): 1964-1979.e6, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38759628

RESUMO

The role of the mitochondrial electron transport chain (ETC) in regulating ferroptosis is not fully elucidated. Here, we reveal that pharmacological inhibition of the ETC complex I reduces ubiquinol levels while decreasing ATP levels and activating AMP-activated protein kinase (AMPK), the two effects known for their roles in promoting and suppressing ferroptosis, respectively. Consequently, the impact of complex I inhibitors on ferroptosis induced by glutathione peroxidase 4 (GPX4) inhibition is limited. The pharmacological inhibition of complex I in LKB1-AMPK-inactivated cells, or genetic ablation of complex I (which does not trigger apparent AMPK activation), abrogates the AMPK-mediated ferroptosis-suppressive effect and sensitizes cancer cells to GPX4-inactivation-induced ferroptosis. Furthermore, complex I inhibition synergizes with radiotherapy (RT) to selectively suppress the growth of LKB1-deficient tumors by inducing ferroptosis in mouse models. Our data demonstrate a multifaceted role of complex I in regulating ferroptosis and propose a ferroptosis-inducing therapeutic strategy for LKB1-deficient cancers.


Assuntos
Proteínas Quinases Ativadas por AMP , Complexo I de Transporte de Elétrons , Ferroptose , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Proteínas Serina-Treonina Quinases , Ferroptose/genética , Ferroptose/efeitos dos fármacos , Animais , Humanos , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Complexo I de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Camundongos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Linhagem Celular Tumoral , Neoplasias/genética , Neoplasias/patologia , Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Quinases Proteína-Quinases Ativadas por AMP/genética , Mitocôndrias/metabolismo , Mitocôndrias/genética , Mitocôndrias/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Transdução de Sinais , Feminino
6.
Mol Cell ; 83(4): 556-573.e7, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36696898

RESUMO

The protection of DNA replication forks under stress is essential for genome maintenance and cancer suppression. One mechanism of fork protection involves an elevation in intracellular Ca2+ ([Ca2+]i), which in turn activates CaMKK2 and AMPK to prevent uncontrolled fork processing by Exo1. How replication stress triggers [Ca2+]i elevation is unclear. Here, we report a role of cytosolic self-DNA (cytosDNA) and the ion channel TRPV2 in [Ca2+]i induction and fork protection. Replication stress leads to the generation of ssDNA and dsDNA species that, upon translocation into cytoplasm, trigger the activation of the sensor protein cGAS and the production of cGAMP. The subsequent binding of cGAMP to STING causes its dissociation from TRPV2, leading to TRPV2 derepression and Ca2+ release from the ER, which in turn activates the downstream signaling cascade to prevent fork degradation. This Ca2+-dependent genome protection pathway is also activated in response to replication stress caused by oncogene activation.


Assuntos
DNA , Nucleotidiltransferases , DNA/genética , DNA/metabolismo , Replicação do DNA , DNA de Cadeia Simples , Proteínas de Membrana , Nucleotidiltransferases/metabolismo , Transdução de Sinais/fisiologia , Canais de Cátion TRPV
7.
Mol Cell ; 82(24): 4700-4711.e12, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36384136

RESUMO

Maintenance of energy level to drive movements and material exchange with the environment is a basic principle of life. AMP-activated protein kinase (AMPK) senses energy level and is a major regulator of cellular energy responses. The gamma subunit of AMPK senses elevated ratio of AMP to ATP and allosterically activates the alpha catalytic subunit to phosphorylate downstream effectors. Here, we report that knockout of AMPKγ, but not AMPKα, suppressed phosphorylation of eukaryotic translation elongation factor 2 (eEF2) induced by energy starvation. We identified PPP6C as an AMPKγ-regulated phosphatase of eEF2. AMP-bound AMPKγ sequesters PPP6C, thereby blocking dephosphorylation of eEF2 and thus inhibiting translation elongation to preserve energy and to promote cell survival. Further phosphoproteomic analysis identified additional targets of PPP6C regulated by energy stress in an AMPKγ-dependent manner. Thus, AMPKγ senses cellular energy availability to regulate not only AMPKα kinase, but also PPP6C phosphatase and possibly other effectors.


Assuntos
Proteínas Quinases Ativadas por AMP , Biossíntese de Proteínas , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Fosforilação , Fator 2 de Elongação de Peptídeos/metabolismo
8.
Mol Cell ; 82(23): 4519-4536.e7, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36384137

RESUMO

Nutrient sensing and damage sensing are two fundamental processes in living organisms. While hyperglycemia is frequently linked to diabetes-related vulnerability to microbial infection, how body glucose levels affect innate immune responses to microbial invasion is not fully understood. Here, we surprisingly found that viral infection led to a rapid and dramatic decrease in blood glucose levels in rodents, leading to robust AMPK activation. AMPK, once activated, directly phosphorylates TBK1 at S511, which triggers IRF3 recruitment and the assembly of MAVS or STING signalosomes. Consistently, ablation or inhibition of AMPK, knockin of TBK1-S511A, or increased glucose levels compromised nucleic acid sensing, while boosting AMPK-TBK1 cascade by AICAR or TBK1-S511E knockin improves antiviral immunity substantially in various animal models. Thus, we identify TBK1 as an AMPK substrate, reveal the molecular mechanism coupling a dual sensing of glucose and nuclei acids, and report its physiological necessity in antiviral defense.


Assuntos
Proteínas Quinases Ativadas por AMP , Ácidos Nucleicos , Animais , Proteínas Quinases Ativadas por AMP/genética , Imunidade Inata , Antivirais , Glucose
9.
Mol Cell ; 81(13): 2722-2735.e9, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34077757

RESUMO

Lipid droplets are important for cancer cell growth and survival. However, the mechanism underlying the initiation of lipid droplet lipolysis is not well understood. We demonstrate here that glucose deprivation induces the binding of choline kinase (CHK) α2 to lipid droplets, which is sequentially mediated by AMPK-dependent CHKα2 S279 phosphorylation and KAT5-dependent CHKα2 K247 acetylation. Importantly, CHKα2 with altered catalytic domain conformation functions as a protein kinase and phosphorylates PLIN2 at Y232 and PLIN3 at Y251. The phosphorylated PLIN2/3 dissociate from lipid droplets and are degraded by Hsc70-mediated autophagy, thereby promoting lipid droplet lipolysis, fatty acid oxidation, and brain tumor growth. In addition, levels of CHKα2 S279 phosphorylation, CHKα2 K247 acetylation, and PLIN2/3 phosphorylation are positively correlated with one another in human glioblastoma specimens and are associated with poor prognosis in glioblastoma patients. These findings underscore the role of CHKα2 as a protein kinase in lipolysis and glioblastoma development.


Assuntos
Colina Quinase/metabolismo , Glioblastoma/enzimologia , Gotículas Lipídicas/enzimologia , Lipólise , Proteínas de Neoplasias/metabolismo , Proteínas Quinases/metabolismo , Acetilação , Linhagem Celular Tumoral , Colina Quinase/genética , Glioblastoma/genética , Humanos , Proteínas de Neoplasias/genética , Proteínas Quinases/genética
10.
Mol Cell ; 81(2): 370-385.e7, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33271062

RESUMO

The mechanisms of cellular energy sensing and AMPK-mediated mTORC1 inhibition are not fully delineated. Here, we discover that RIPK1 promotes mTORC1 inhibition during energetic stress. RIPK1 is involved in mediating the interaction between AMPK and TSC2 and facilitate TSC2 phosphorylation at Ser1387. RIPK1 loss results in a high basal mTORC1 activity that drives defective lysosomes in cells and mice, leading to accumulation of RIPK3 and CASP8 and sensitization to cell death. RIPK1-deficient cells are unable to cope with energetic stress and are vulnerable to low glucose levels and metformin. Inhibition of mTORC1 rescues the lysosomal defects and vulnerability to energetic stress and prolongs the survival of RIPK1-deficient neonatal mice. Thus, RIPK1 plays an important role in the cellular response to low energy levels and mediates AMPK-mTORC1 signaling. These findings shed light on the regulation of mTORC1 during energetic stress and unveil a point of crosstalk between pro-survival and pro-death pathways.


Assuntos
Proteína 5 Relacionada à Autofagia/genética , Proteína de Domínio de Morte Associada a Fas/genética , Intestino Grosso/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Animais Recém-Nascidos , Proteína 5 Relacionada à Autofagia/deficiência , Caspase 8/genética , Caspase 8/metabolismo , Morte Celular/genética , Proteína de Domínio de Morte Associada a Fas/deficiência , Regulação da Expressão Gênica , Glucose/antagonistas & inibidores , Glucose/farmacologia , Células HEK293 , Células HT29 , Humanos , Intestino Grosso/efeitos dos fármacos , Intestino Grosso/patologia , Células Jurkat , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Lisossomos/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Metformina/antagonistas & inibidores , Metformina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Transdução de Sinais , Sirolimo/farmacologia , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
11.
Mol Cell ; 81(11): 2317-2331.e6, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33909988

RESUMO

Aberrant energy status contributes to multiple metabolic diseases, including obesity, diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4 immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on Ser283, thereby disrupting its interaction with CMTM4 and subsequently triggering PD-L1 degradation. In addition, AMPK phosphorylates EZH2, which disrupts PRC2 function, leading to enhanced IFNs and antigen presentation gene expression. Through these mechanisms, AMPK agonists or ketogenic diets enhance the efficacy of anti-CTLA-4 immunotherapy and improve the overall survival rate in syngeneic mouse tumor models. Our findings reveal a pivotal role for AMPK in regulating the immune response to immune-checkpoint blockade and advocate for combining ketogenic diets or AMPK agonists with anti-CTLA4 immunotherapy to combat cancer.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Antígeno B7-H1/genética , Neoplasias da Mama/genética , Antígeno CTLA-4/genética , Neoplasias Colorretais/genética , Inibidores de Checkpoint Imunológico , Proteínas Quinases Ativadas por AMP/imunologia , Aloenxertos , Animais , Anticorpos Neutralizantes/farmacologia , Antineoplásicos/farmacologia , Antígeno B7-H1/imunologia , Compostos de Bifenilo/farmacologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/mortalidade , Neoplasias da Mama/terapia , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/imunologia , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/terapia , Dieta Cetogênica/métodos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/imunologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia/métodos , Proteínas com Domínio MARVEL/genética , Proteínas com Domínio MARVEL/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Pironas/farmacologia , Transdução de Sinais , Análise de Sobrevida , Tiofenos/farmacologia
12.
Mol Cell ; 81(18): 3803-3819.e7, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34547240

RESUMO

Mitochondrial dynamics regulated by mitochondrial fusion and fission maintain mitochondrial functions, whose alterations underline various human diseases. Here, we show that inositol is a critical metabolite directly restricting AMPK-dependent mitochondrial fission independently of its classical mode as a precursor for phosphoinositide generation. Inositol decline by IMPA1/2 deficiency elicits AMPK activation and mitochondrial fission without affecting ATP level, whereas inositol accumulation prevents AMPK-dependent mitochondrial fission. Metabolic stress or mitochondrial damage causes inositol decline in cells and mice to elicit AMPK-dependent mitochondrial fission. Inositol directly binds to AMPKγ and competes with AMP for AMPKγ binding, leading to restriction of AMPK activation and mitochondrial fission. Our study suggests that the AMP/inositol ratio is a critical determinant for AMPK activation and establishes a model in which AMPK activation requires inositol decline to release AMPKγ for AMP binding. Hence, AMPK is an inositol sensor, whose inactivation by inositol serves as a mechanism to restrict mitochondrial fission.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Inositol/metabolismo , Dinâmica Mitocondrial/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Linhagem Celular , Humanos , Inositol/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Células PC-3 , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Estresse Fisiológico/fisiologia
13.
Mol Cell ; 80(2): 263-278.e7, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33022274

RESUMO

Cancer metastasis accounts for the major cause of cancer-related deaths. How disseminated cancer cells cope with hostile microenvironments in secondary site for full-blown metastasis is largely unknown. Here, we show that AMPK (AMP-activated protein kinase), activated in mouse metastasis models, drives pyruvate dehydrogenase complex (PDHc) activation to maintain TCA cycle (tricarboxylic acid cycle) and promotes cancer metastasis by adapting cancer cells to metabolic and oxidative stresses. This AMPK-PDHc axis is activated in advanced breast cancer and predicts poor metastasis-free survival. Mechanistically, AMPK localizes in the mitochondrial matrix and phosphorylates the catalytic alpha subunit of PDHc (PDHA) on two residues S295 and S314, which activates the enzymatic activity of PDHc and alleviates an inhibitory phosphorylation by PDHKs, respectively. Importantly, these phosphorylation events mediate PDHc function in cancer metastasis. Our study reveals that AMPK-mediated PDHA phosphorylation drives PDHc activation and TCA cycle to empower cancer cells adaptation to metastatic microenvironments for metastasis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Ciclo do Ácido Cítrico , Complexo Piruvato Desidrogenase/metabolismo , Animais , Domínio Catalítico , Linhagem Celular Tumoral , Sobrevivência Celular , Ativação Enzimática , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Fosforilação , Fosfosserina/metabolismo , Transdução de Sinais , Estresse Fisiológico , Análise de Sobrevida
14.
Genes Dev ; 34(19-20): 1330-1344, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32912901

RESUMO

Despite being the frontline therapy for type 2 diabetes, the mechanisms of action of the biguanide drug metformin are still being discovered. In particular, the detailed molecular interplays between the AMPK and the mTORC1 pathway in the hepatic benefits of metformin are still ill defined. Metformin-dependent activation of AMPK classically inhibits mTORC1 via TSC/RHEB, but several lines of evidence suggest additional mechanisms at play in metformin inhibition of mTORC1. Here we investigated the role of direct AMPK-mediated serine phosphorylation of RAPTOR in a new RaptorAA mouse model, in which AMPK phospho-serine sites Ser722 and Ser792 of RAPTOR were mutated to alanine. Metformin treatment of primary hepatocytes and intact murine liver requires AMPK regulation of both RAPTOR and TSC2 to fully inhibit mTORC1, and this regulation is critical for both the translational and transcriptional response to metformin. Transcriptionally, AMPK and mTORC1 were both important for regulation of anabolic metabolism and inflammatory programs triggered by metformin treatment. The hepatic transcriptional response in mice on high-fat diet treated with metformin was largely ablated by AMPK deficiency under the conditions examined, indicating the essential role of this kinase and its targets in metformin action in vivo.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Metformina/farmacologia , Proteína Regulatória Associada a mTOR/genética , Transdução de Sinais/efeitos dos fármacos , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Genótipo , Hipoglicemiantes/farmacologia , Inflamação , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Metabolismo/efeitos dos fármacos , Metformina/uso terapêutico , Camundongos , Fosforilação/efeitos dos fármacos , Proteína Regulatória Associada a mTOR/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
15.
EMBO J ; 42(20): e113743, 2023 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-37661833

RESUMO

Mitochondria play essential roles in cancer cell adaptation to hypoxia, but the underlying mechanisms remain elusive. Through mitochondrial proteomic profiling, we here find that the prolyl hydroxylase EglN1 (PHD2) accumulates on mitochondria under hypoxia. EglN1 substrate-binding region in the ß2ß3 loop is responsible for its mitochondrial translocation and contributes to breast tumor growth. Furthermore, we identify AMP-activated protein kinase alpha (AMPKα) as an EglN1 substrate on mitochondria. The EglN1-AMPKα interaction is essential for their mutual mitochondrial translocation. After EglN1 prolyl-hydroxylates AMPKα under normoxia, they rapidly dissociate following prolyl-hydroxylation, leading to their immediate release from mitochondria. In contrast, hypoxia results in constant EglN1-AMPKα interaction and their accumulation on mitochondria, leading to the formation of a Ca2+ /calmodulin-dependent protein kinase 2 (CaMKK2)-EglN1-AMPKα complex to activate AMPKα phosphorylation, ensuring metabolic homeostasis and breast tumor growth. Our findings identify EglN1 as an oxygen-sensitive metabolic checkpoint signaling hypoxic stress to mitochondria through its ß2ß3 loop region, suggesting a potential therapeutic target for breast cancer.


Assuntos
Proteínas Quinases Ativadas por AMP , Neoplasias da Mama , Feminino , Humanos , Proteínas Quinases Ativadas por AMP/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Hipóxia , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Mitocôndrias/metabolismo , Proteômica
16.
Development ; 151(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38165175

RESUMO

The insect epidermis forms the exoskeleton and determines the body size of an organism. How the epidermis acts as a metabolic regulator to adapt to changes in dietary protein availability remains elusive. Here, we show that the Drosophila epidermis regulates tyrosine (Tyr) catabolism in response to dietary protein levels, thereby promoting metabolic homeostasis. The gene expression profile of the Drosophila larval body wall reveals that enzymes involved in the Tyr degradation pathway, including 4-hydroxyphenylpyruvate dioxygenase (Hpd), are upregulated by increased protein intake. Hpd is specifically expressed in the epidermis and is dynamically regulated by the internal Tyr levels. Whereas basal Hpd expression is maintained by insulin/IGF-1 signalling, Hpd induction on high-protein diet requires activation of the AMP-activated protein kinase (AMPK)-forkhead box O subfamily (FoxO) axis. Impairment of the FoxO-mediated Hpd induction in the epidermis leads to aberrant increases in internal Tyr and its metabolites, disrupting larval development on high-protein diets. Taken together, our findings uncover a crucial role of the epidermis as a metabolic regulator in coping with an unfavourable dietary environment.


Assuntos
Dieta Rica em Proteínas , Drosophila , Animais , Drosophila/metabolismo , Homeostase , Insulina/metabolismo , Epiderme/metabolismo , Proteínas Alimentares , Tirosina
17.
Mol Cell ; 76(4): 546-561.e8, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31561952

RESUMO

Through transcriptional control of the evolutionarily conserved heat shock, or proteotoxic stress, response, heat shock factor 1 (HSF1) preserves proteomic stability. Here, we show that HSF1, a physiological substrate for AMP-activated protein kinase (AMPK), constitutively suppresses this central metabolic sensor. By physically evoking conformational switching of AMPK, HSF1 impairs AMP binding to the γ subunits and enhances the PP2A-mediated de-phosphorylation, but it impedes the LKB1-mediated phosphorylation of Thr172, and retards ATP binding to the catalytic α subunits. These immediate and manifold regulations empower HSF1 to both repress AMPK under basal conditions and restrain its activation by diverse stimuli, thereby promoting lipogenesis, cholesterol synthesis, and protein cholesteroylation. In vivo, HSF1 antagonizes AMPK to control body fat mass and drive the lipogenic phenotype and growth of melanomas independently of its intrinsic transcriptional action. Thus, the physical AMPK-HSF1 interaction epitomizes a reciprocal kinase-substrate regulation whereby lipid metabolism and proteomic stability intertwine.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Metabolismo Energético , Fatores de Transcrição de Choque Térmico/metabolismo , Proteínas Quinases Ativadas por AMP/química , Proteínas Quinases Ativadas por AMP/genética , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Adiposidade , Animais , Sítios de Ligação , Proliferação de Células , Colesterol/biossíntese , Células HEK293 , Células HeLa , Fatores de Transcrição de Choque Térmico/deficiência , Fatores de Transcrição de Choque Térmico/genética , Humanos , Lipogênese , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Fosforilação , Conformação Proteica , Estabilidade Proteica , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Relação Estrutura-Atividade
18.
Mol Cell ; 74(6): 1123-1137.e6, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31053472

RESUMO

Abnormal processing of stressed replication forks by nucleases can cause fork collapse, genomic instability, and cell death. Despite its importance, it is poorly understood how the cell properly controls nucleases to prevent detrimental fork processing. Here, we report a signaling pathway that controls the activity of exonuclease Exo1 to prevent aberrant fork resection during replication stress. Our results indicate that replication stress elevates intracellular Ca2+ concentration ([Ca2+]i), leading to activation of CaMKK2 and the downstream kinase 5' AMP-activated protein kinase (AMPK). Following activation, AMPK directly phosphorylates Exo1 at serine 746 to promote 14-3-3 binding and inhibit Exo1 recruitment to stressed replication forks, thereby avoiding unscheduled fork resection. Disruption of this signaling pathway results in excessive ssDNA, chromosomal instability, and hypersensitivity to replication stress inducers. These findings reveal a link between [Ca2+]i and the replication stress response as well as a function of the Ca2+-CaMKK2-AMPK signaling axis in safeguarding fork structure to maintain genome stability.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/genética , Cálcio/metabolismo , Enzimas Reparadoras do DNA/genética , Reparo do DNA , Replicação do DNA , Exodesoxirribonucleases/genética , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Sinalização do Cálcio/genética , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/genética , Quinase 1 do Ponto de Checagem/metabolismo , Cromatina/química , Cromatina/metabolismo , Dano ao DNA , Enzimas Reparadoras do DNA/metabolismo , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Exodesoxirribonucleases/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Fosforilação , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Development ; 150(21)2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37823352

RESUMO

Dendritic outgrowth in immature neurons is enhanced by neuronal activity and is considered one of the mechanisms of neural circuit optimization. It is known that calcium signals affect transcriptional regulation and cytoskeletal remodeling necessary for dendritic outgrowth. Here, we demonstrate that activity-dependent calcium signaling also controls mitochondrial homeostasis via AMP-activated protein kinase (AMPK) in growing dendrites of differentiating mouse hippocampal neurons. We found that the inhibition of neuronal activity induced dendritic hypotrophy with abnormally elongated mitochondria. In growing dendrites, AMPK is activated by neuronal activity and dynamically oscillates in synchrony with calcium spikes, and this AMPK oscillation was inhibited by CaMKK2 knockdown. AMPK activation led to phosphorylation of MFF and ULK1, which initiate mitochondrial fission and mitophagy, respectively. Dendritic mitochondria in AMPK-depleted neurons exhibited impaired fission and mitophagy and displayed multiple signs of dysfunction. Genetic inhibition of fission led to dendritic hypoplasia that was reminiscent of AMPK-deficient neurons. Thus, AMPK activity is finely tuned by the calcium-CaMKK2 pathway and regulates mitochondrial homeostasis by facilitating removal of damaged components of mitochondria in growing neurons during normal brain development.


Assuntos
Proteínas Quinases Ativadas por AMP , Cálcio , Camundongos , Animais , Fosforilação , Proteínas Quinases Ativadas por AMP/genética , Cálcio/metabolismo , Neurônios/metabolismo , Mitocôndrias/metabolismo , Dendritos/metabolismo , Homeostase
20.
Mol Cell ; 70(1): 120-135.e8, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29625033

RESUMO

The Ser/Thr protein kinase mTOR controls metabolic pathways, including the catabolic process of autophagy. Autophagy plays additional, catabolism-independent roles in homeostasis of cytoplasmic endomembranes and whole organelles. How signals from endomembrane damage are transmitted to mTOR to orchestrate autophagic responses is not known. Here we show that mTOR is inhibited by lysosomal damage. Lysosomal damage, recognized by galectins, leads to association of galectin-8 (Gal8) with the mTOR apparatus on the lysosome. Gal8 inhibits mTOR activity through its Ragulator-Rag signaling machinery, whereas galectin-9 activates AMPK in response to lysosomal injury. Both systems converge upon downstream effectors including autophagy and defense against Mycobacterium tuberculosis. Thus, a novel galectin-based signal-transduction system, termed here GALTOR, intersects with the known regulators of mTOR on the lysosome and controls them in response to lysosomal damage. VIDEO ABSTRACT.


Assuntos
Autofagia , Galectinas/metabolismo , Lisossomos/enzimologia , Serina-Treonina Quinases TOR/metabolismo , Tuberculose/enzimologia , Proteínas Quinases Ativadas por AMP/metabolismo , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Galectinas/deficiência , Galectinas/genética , Células HEK293 , Células HeLa , Humanos , Lisossomos/microbiologia , Lisossomos/patologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos , Mycobacterium tuberculosis/patogenicidade , Transdução de Sinais , Células THP-1 , Serina-Treonina Quinases TOR/genética , Tuberculose/genética , Tuberculose/microbiologia , Tuberculose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA