Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.291
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nat Rev Mol Cell Biol ; 22(1): 54-70, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33093673

RESUMO

Mitochondria contain about 1,000-1,500 proteins that fulfil multiple functions. Mitochondrial proteins originate from two genomes: mitochondrial and nuclear. Hence, proper mitochondrial function requires synchronization of gene expression in the nucleus and in mitochondria and necessitates efficient import of mitochondrial proteins into the organelle from the cytosol. Furthermore, the mitochondrial proteome displays high plasticity to allow the adaptation of mitochondrial function to cellular requirements. Maintenance of this complex and adaptable mitochondrial proteome is challenging, but is of crucial importance to cell function. Defects in mitochondrial proteostasis lead to proteotoxic insults and eventually cell death. Different quality control systems monitor the mitochondrial proteome. The cytosolic ubiquitin-proteasome system controls protein transport across the mitochondrial outer membrane and removes damaged or mislocalized proteins. Concomitantly, a number of mitochondrial chaperones and proteases govern protein folding and degrade damaged proteins inside mitochondria. The quality control factors also regulate processing and turnover of native proteins to control protein import, mitochondrial metabolism, signalling cascades, mitochondrial dynamics and lipid biogenesis, further ensuring proper function of mitochondria. Thus, mitochondrial protein quality control mechanisms are of pivotal importance to integrate mitochondria into the cellular environment.


Assuntos
Lipogênese , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Proteínas Mitocondriais/metabolismo , Proteoma/metabolismo , Animais , Humanos , Transporte Proteico , Proteoma/análise
2.
Cell ; 174(6): 1342-1344, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30193107

RESUMO

Synthetically re-designing eukaryotic metabolism has proven immensely challenging, raising the question of whether evolution has metabolically hardwired eukaryotic cells. Yu et al. now report that, through orchestrating multiple genetic changes and laboratory evolution, Saccharomyces metabolism can be reprogrammed from its evolutionary objective of producing ethanol to produce large amounts of free fatty acids.


Assuntos
Alcoolismo , Etanol , Fermentação , Humanos , Lipogênese , Saccharomyces cerevisiae
3.
Cell ; 174(6): 1549-1558.e14, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30100189

RESUMO

Engineering microorganisms for production of fuels and chemicals often requires major re-programming of metabolism to ensure high flux toward the product of interest. This is challenging, as millions of years of evolution have resulted in establishment of tight regulation of metabolism for optimal growth in the organism's natural habitat. Here, we show through metabolic engineering that it is possible to alter the metabolism of Saccharomyces cerevisiae from traditional ethanol fermentation to a pure lipogenesis metabolism, resulting in high-level production of free fatty acids. Through metabolic engineering and process design, we altered subcellular metabolic trafficking, fine-tuned NADPH and ATP supply, and decreased carbon flux to biomass, enabling production of 33.4 g/L extracellular free fatty acids. We further demonstrate that lipogenesis metabolism can replace ethanol fermentation by deletion of pyruvate decarboxylase enzymes followed by adaptive laboratory evolution. Genome sequencing of evolved strains showed that pyruvate kinase mutations were essential for this phenotype.


Assuntos
Ácidos Graxos não Esterificados/biossíntese , Engenharia Metabólica , Saccharomyces cerevisiae/metabolismo , Acetilcoenzima A/metabolismo , Glucose/metabolismo , Glicólise , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Lipogênese , NADP/metabolismo , Via de Pentose Fosfato/genética , Piruvato Quinase/genética , Piruvato Quinase/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
4.
Cell ; 175(2): 502-513.e13, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30245009

RESUMO

Acetate is a major nutrient that supports acetyl-coenzyme A (Ac-CoA) metabolism and thus lipogenesis and protein acetylation. However, its source is unclear. Here, we report that pyruvate, the end product of glycolysis and key node in central carbon metabolism, quantitatively generates acetate in mammals. This phenomenon becomes more pronounced in the context of nutritional excess, such as during hyperactive glucose metabolism. Conversion of pyruvate to acetate occurs through two mechanisms: (1) coupling to reactive oxygen species (ROS) and (2) neomorphic enzyme activity from keto acid dehydrogenases that enable function as pyruvate decarboxylases. Further, we demonstrate that de novo acetate production sustains Ac-CoA pools and cell proliferation in limited metabolic environments, such as during mitochondrial dysfunction or ATP citrate lyase (ACLY) deficiency. By virtue of de novo acetate production being coupled to mitochondrial metabolism, there are numerous possible regulatory mechanisms and links to pathophysiology.


Assuntos
Acetatos/metabolismo , Glucose/metabolismo , Ácido Pirúvico/metabolismo , ATP Citrato (pro-S)-Liase/fisiologia , Acetilcoenzima A/biossíntese , Acetilcoenzima A/metabolismo , Acetilação , Animais , Feminino , Glicólise/fisiologia , Lipogênese/fisiologia , Masculino , Mamíferos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Oxirredutases , Piruvato Descarboxilase/fisiologia , Espécies Reativas de Oxigênio/metabolismo
5.
Cell ; 174(4): 831-842.e12, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-30057115

RESUMO

Overnutrition disrupts circadian metabolic rhythms by mechanisms that are not well understood. Here, we show that diet-induced obesity (DIO) causes massive remodeling of circadian enhancer activity in mouse liver, triggering synchronous high-amplitude circadian rhythms of both fatty acid (FA) synthesis and oxidation. SREBP expression was rhythmically induced by DIO, leading to circadian FA synthesis and, surprisingly, FA oxidation (FAO). DIO similarly caused a high-amplitude circadian rhythm of PPARα, which was also required for FAO. Provision of a pharmacological activator of PPARα abrogated the requirement of SREBP for FAO (but not FA synthesis), suggesting that SREBP indirectly controls FAO via production of endogenous PPARα ligands. The high-amplitude rhythm of PPARα imparted time-of-day-dependent responsiveness to lipid-lowering drugs. Thus, acquisition of rhythmicity for non-core clock components PPARα and SREBP1 remodels metabolic gene transcription in response to overnutrition and enables a chronopharmacological approach to metabolic disorders.


Assuntos
Ritmo Circadiano , Dieta/efeitos adversos , Fígado/metabolismo , Obesidade/metabolismo , PPAR alfa/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Regulação da Expressão Gênica , Metabolismo dos Lipídeos , Lipogênese , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/patologia , PPAR alfa/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
6.
Cell ; 175(1): 133-145.e15, 2018 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-30220454

RESUMO

Nonalcoholic fatty liver disease (NAFLD) progresses to nonalcoholic steatohepatitis (NASH) in response to elevated endoplasmic reticulum (ER) stress. Whereas the onset of simple steatosis requires elevated de novo lipogenesis, progression to NASH is triggered by accumulation of hepatocyte-free cholesterol. We now show that caspase-2, whose expression is ER-stress inducible and elevated in human and mouse NASH, controls the buildup of hepatic-free cholesterol and triglycerides by activating sterol regulatory element-binding proteins (SREBP) in a manner refractory to feedback inhibition. Caspase-2 colocalizes with site 1 protease (S1P) and cleaves it to generate a soluble active fragment that initiates SCAP-independent SREBP1/2 activation in the ER. Caspase-2 ablation or pharmacological inhibition prevents diet-induced steatosis and NASH progression in ER-stress-prone mice. Caspase-2 inhibition offers a specific and effective strategy for preventing or treating stress-driven fatty liver diseases, whereas caspase-2-generated S1P proteolytic fragments, which enter the secretory pathway, are potential NASH biomarkers.


Assuntos
Caspase 2/fisiologia , Lipogênese/fisiologia , Pró-Proteína Convertases/fisiologia , Serina Endopeptidases/fisiologia , Animais , Colesterol/metabolismo , Retículo Endoplasmático/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Fígado Gorduroso/fisiopatologia , Células HEK293 , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/metabolismo
7.
Cell ; 171(7): 1545-1558.e18, 2017 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-29153836

RESUMO

mTORC1 is a signal integrator and master regulator of cellular anabolic processes linked to cell growth and survival. Here, we demonstrate that mTORC1 promotes lipid biogenesis via SRPK2, a key regulator of RNA-binding SR proteins. mTORC1-activated S6K1 phosphorylates SRPK2 at Ser494, which primes Ser497 phosphorylation by CK1. These phosphorylation events promote SRPK2 nuclear translocation and phosphorylation of SR proteins. Genome-wide transcriptome analysis reveals that lipid biosynthetic enzymes are among the downstream targets of mTORC1-SRPK2 signaling. Mechanistically, SRPK2 promotes SR protein binding to U1-70K to induce splicing of lipogenic pre-mRNAs. Inhibition of this signaling pathway leads to intron retention of lipogenic genes, which triggers nonsense-mediated mRNA decay. Genetic or pharmacological inhibition of SRPK2 blunts de novo lipid synthesis, thereby suppressing cell growth. These results thus reveal a novel role of mTORC1-SRPK2 signaling in post-transcriptional regulation of lipid metabolism and demonstrate that SRPK2 is a potential therapeutic target for mTORC1-driven metabolic disorders.


Assuntos
Regulação da Expressão Gênica , Lipogênese , Processamento Pós-Transcricional do RNA , Transdução de Sinais , Animais , Núcleo Celular/metabolismo , Colesterol/metabolismo , Ácidos Graxos/metabolismo , Feminino , Xenoenxertos , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo
8.
Cell ; 171(4): 824-835.e18, 2017 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-29056338

RESUMO

Insulin resistance is a hallmark of diabetes and an unmet clinical need. Insulin inhibits hepatic glucose production and promotes lipogenesis by suppressing FOXO1-dependent activation of G6pase and inhibition of glucokinase, respectively. The tight coupling of these events poses a dual conundrum: mechanistically, as the FOXO1 corepressor of glucokinase is unknown, and clinically, as inhibition of glucose production is predicted to increase lipogenesis. Here, we report that SIN3A is the insulin-sensitive FOXO1 corepressor of glucokinase. Genetic ablation of SIN3A abolishes nutrient regulation of glucokinase without affecting other FOXO1 target genes and lowers glycemia without concurrent steatosis. To extend this work, we executed a small-molecule screen and discovered selective inhibitors of FOXO-dependent glucose production devoid of lipogenic activity in hepatocytes. In addition to identifying a novel mode of insulin action, these data raise the possibility of developing selective modulators of unliganded transcription factors to dial out adverse effects of insulin sensitizers.


Assuntos
Proteína Forkhead Box O1/antagonistas & inibidores , Glucose/metabolismo , Hepatócitos/metabolismo , Resistência à Insulina , Acetilação , Animais , Células Cultivadas , Proteína Forkhead Box O1/química , Glucoquinase/genética , Glucoquinase/metabolismo , Glucose-6-Fosfatase/genética , Glucose-6-Fosfatase/metabolismo , Células HEK293 , Hepatócitos/enzimologia , Histona Desacetilases/metabolismo , Humanos , Lipogênese/efeitos dos fármacos , Camundongos , Camundongos Knockout , Fosforilação , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Complexo Correpressor Histona Desacetilase e Sin3
9.
Mol Cell ; 83(16): 3010-3026.e8, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37595559

RESUMO

The mechanistic target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth that stimulates macromolecule synthesis through transcription, RNA processing, and post-translational modification of metabolic enzymes. However, the mechanisms of how mTORC1 orchestrates multiple steps of gene expression programs remain unclear. Here, we identify family with sequence similarity 120A (FAM120A) as a transcription co-activator that couples transcription and splicing of de novo lipid synthesis enzymes downstream of mTORC1-serine/arginine-rich protein kinase 2 (SRPK2) signaling. The mTORC1-activated SRPK2 phosphorylates splicing factor serine/arginine-rich splicing factor 1 (SRSF1), enhancing its binding to FAM120A. FAM120A directly interacts with a lipogenic transcription factor SREBP1 at active promoters, thereby bridging the newly transcribed lipogenic genes from RNA polymerase II to the SRSF1 and U1-70K-containing RNA-splicing machinery. This mTORC1-regulated, multi-protein complex promotes efficient splicing and stability of lipogenic transcripts, resulting in fatty acid synthesis and cancer cell proliferation. These results elucidate FAM120A as a critical transcription co-factor that connects mTORC1-dependent gene regulation programs for anabolic cell growth.


Assuntos
Arginina , Lipogênese , Proteína de Ligação a Elemento Regulador de Esterol 1 , Lipogênese/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Fatores de Processamento de RNA , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Humanos , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo
10.
Cell ; 159(2): 318-32, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25303528

RESUMO

Increased adipose tissue lipogenesis is associated with enhanced insulin sensitivity. Mice overexpressing the Glut4 glucose transporter in adipocytes have elevated lipogenesis and increased glucose tolerance despite being obese with elevated circulating fatty acids. Lipidomic analysis of adipose tissue revealed the existence of branched fatty acid esters of hydroxy fatty acids (FAHFAs) that were elevated 16- to 18-fold in these mice. FAHFA isomers differ by the branched ester position on the hydroxy fatty acid (e.g., palmitic-acid-9-hydroxy-stearic-acid, 9-PAHSA). PAHSAs are synthesized in vivo and regulated by fasting and high-fat feeding. PAHSA levels correlate highly with insulin sensitivity and are reduced in adipose tissue and serum of insulin-resistant humans. PAHSA administration in mice lowers ambient glycemia and improves glucose tolerance while stimulating GLP-1 and insulin secretion. PAHSAs also reduce adipose tissue inflammation. In adipocytes, PAHSAs signal through GPR120 to enhance insulin-stimulated glucose uptake. Thus, FAHFAs are endogenous lipids with the potential to treat type 2 diabetes.


Assuntos
Tecido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ésteres/metabolismo , Ácidos Graxos/metabolismo , Adulto , Animais , Diabetes Mellitus Tipo 2/dietoterapia , Dieta , Ésteres/administração & dosagem , Ésteres/análise , Ácidos Graxos/administração & dosagem , Ácidos Graxos/análise , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Humanos , Inflamação/dietoterapia , Insulina/metabolismo , Resistência à Insulina , Lipogênese , Masculino , Espectrometria de Massas , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Receptores Acoplados a Proteínas G/metabolismo
11.
Mol Cell ; 81(18): 3820-3832.e7, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34233158

RESUMO

A metabolic imbalance between lipid synthesis and degradation can lead to hepatic lipid accumulation, a characteristic of patients with non-alcoholic fatty liver disease (NAFLD). Here, we report that high-fat-diet-induced sterol regulatory element-binding protein (SREBP)-1c, a key transcription factor that regulates lipid biosynthesis, impairs autophagic lipid catabolism via altered H2S signaling. SREBP-1c reduced cystathionine gamma-lyase (CSE) via miR-216a, which in turn decreased hepatic H2S levels and sulfhydration-dependent activation of Unc-51-like autophagy-activating kinase 1 (ULK1). Furthermore, Cys951Ser mutation of ULK1 decreased autolysosome formation and promoted hepatic lipid accumulation in mice, suggesting that the loss of ULK1 sulfhydration was directly associated with the pathogenesis of NAFLD. Moreover, silencing of CSE in SREBP-1c knockout mice increased liver triglycerides, confirming the connection between CSE, autophagy, and SREBP-1c. Overall, our results uncover a 2-fold mechanism for SREBP-1c-driven hepatic lipid accumulation through reciprocal activation and inhibition of hepatic lipid biosynthesis and degradation, respectively.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Fígado Gorduroso/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Autofagia , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/genética , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/fisiologia , Linhagem Celular Tumoral , Dieta Hiperlipídica/efeitos adversos , Fígado Gorduroso/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Lipogênese , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/metabolismo , Transdução de Sinais/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/fisiologia , Triglicerídeos/metabolismo
12.
Mol Cell ; 81(9): 1890-1904.e7, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33657401

RESUMO

O-linked ß-N-acetyl glucosamine (O-GlcNAc) is attached to proteins under glucose-replete conditions; this posttranslational modification results in molecular and physiological changes that affect cell fate. Here we show that posttranslational modification of serine/arginine-rich protein kinase 2 (SRPK2) by O-GlcNAc regulates de novo lipogenesis by regulating pre-mRNA splicing. We found that O-GlcNAc transferase O-GlcNAcylated SRPK2 at a nuclear localization signal (NLS), which triggers binding of SRPK2 to importin α. Consequently, O-GlcNAcylated SRPK2 was imported into the nucleus, where it phosphorylated serine/arginine-rich proteins and promoted splicing of lipogenic pre-mRNAs. We determined that protein nuclear import by O-GlcNAcylation-dependent binding of cargo protein to importin α might be a general mechanism in cells. This work reveals a role of O-GlcNAc in posttranscriptional regulation of de novo lipogenesis, and our findings indicate that importin α is a "reader" of an O-GlcNAcylated NLS.


Assuntos
Neoplasias da Mama/metabolismo , Glucose/metabolismo , Lipogênese , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Neoplasias da Mama/genética , Proliferação de Células , Feminino , Glicosilação , Células HEK293 , Humanos , Células MCF-7 , Camundongos Nus , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Precursores de RNA/genética , Precursores de RNA/metabolismo , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Carga Tumoral , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , beta Carioferinas/genética , beta Carioferinas/metabolismo
13.
Genes Dev ; 35(9-10): 713-728, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33888555

RESUMO

MED1 often serves as a surrogate of the general transcription coactivator complex Mediator for identifying active enhancers. MED1 is required for phenotypic conversion of fibroblasts to adipocytes in vitro, but its role in adipose development and expansion in vivo has not been reported. Here, we show that MED1 is not generally required for transcription during adipogenesis in culture and that MED1 is dispensable for adipose development in mice. Instead, MED1 is required for postnatal adipose expansion and the induction of fatty acid and triglyceride synthesis genes after pups switch diet from high-fat maternal milk to carbohydrate-based chow. During adipogenesis, MED1 is dispensable for induction of lineage-determining transcription factors (TFs) PPARγ and C/EBPα but is required for lipid accumulation in the late phase of differentiation. Mechanistically, MED1 controls the induction of lipogenesis genes by facilitating lipogenic TF ChREBP- and SREBP1a-dependent recruitment of Mediator to active enhancers. Together, our findings identify a cell- and gene-specific regulatory role of MED1 as a lipogenesis coactivator required for postnatal adipose expansion.


Assuntos
Tecido Adiposo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Lipogênese/genética , Subunidade 1 do Complexo Mediador/genética , Subunidade 1 do Complexo Mediador/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo Marrom/embriologia , Animais , Células Cultivadas , Dieta , Camundongos , Ligação Proteica/genética
14.
EMBO J ; 43(7): 1187-1213, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38383863

RESUMO

Histone modifications commonly integrate environmental cues with cellular metabolic outputs by affecting gene expression. However, chromatin modifications such as acetylation do not always correlate with transcription, pointing towards an alternative role of histone modifications in cellular metabolism. Using an approach that integrates mass spectrometry-based histone modification mapping and metabolomics with stable isotope tracers, we demonstrate that elevated lipids in acetyltransferase-depleted hepatocytes result from carbon atoms derived from deacetylation of hyperacetylated histone H4 flowing towards fatty acids. Consistently, enhanced lipid synthesis in acetyltransferase-depleted hepatocytes is dependent on histone deacetylases and acetyl-CoA synthetase ACSS2, but not on the substrate specificity of the acetyltransferases. Furthermore, we show that during diet-induced lipid synthesis the levels of hyperacetylated histone H4 decrease in hepatocytes and in mouse liver. In addition, overexpression of acetyltransferases can reverse diet-induced lipogenesis by blocking lipid droplet accumulation and maintaining the levels of hyperacetylated histone H4. Overall, these findings highlight hyperacetylated histones as a metabolite reservoir that can directly contribute carbon to lipid synthesis, constituting a novel function of chromatin in cellular metabolism.


Assuntos
Carbono , Histonas , Animais , Camundongos , Histonas/metabolismo , Carbono/metabolismo , Lipogênese , Cromatina , Acetiltransferases/metabolismo , Lipídeos , Acetilação , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo
15.
Genes Dev ; 34(9-10): 701-714, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32165409

RESUMO

Metabolism and development must be closely coupled to meet the changing physiological needs of each stage in the life cycle. The molecular mechanisms that link these pathways, however, remain poorly understood. Here we show that the Drosophila estrogen-related receptor (dERR) directs a transcriptional switch in mid-pupae that promotes glucose oxidation and lipogenesis in young adults. dERR mutant adults are viable but display reduced locomotor activity, susceptibility to starvation, elevated glucose, and an almost complete lack of stored triglycerides. Molecular profiling by RNA-seq, ChIP-seq, and metabolomics revealed that glycolytic and pentose phosphate pathway genes are induced by dERR, and their reduced expression in mutants is accompanied by elevated glycolytic intermediates, reduced TCA cycle intermediates, and reduced levels of long chain fatty acids. Unexpectedly, we found that the central pathways of energy metabolism, including glycolysis, the tricarboxylic acid cycle, and electron transport chain, are coordinately induced at the transcriptional level in mid-pupae and maintained into adulthood, and this response is partially dependent on dERR, leading to the metabolic defects observed in mutants. Our data support the model that dERR contributes to a transcriptional switch during pupal development that establishes the metabolic state of the adult fly.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Glicólise/genética , Lipogênese/genética , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Transcrição Gênica/genética , Animais , Drosophila/crescimento & desenvolvimento , Mutação , Pupa , Transcriptoma
16.
Nat Rev Mol Cell Biol ; 16(11): 678-89, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26490400

RESUMO

Fatty acid and fat synthesis in the liver is a highly regulated metabolic pathway that is important for very low-density lipoprotein (VLDL) production and thus energy distribution to other tissues. Having common features at their promoter regions, lipogenic genes are coordinately regulated at the transcriptional level. Transcription factors, such as upstream stimulatory factors (USFs), sterol regulatory element-binding protein 1C (SREBP1C), liver X receptors (LXRs) and carbohydrate-responsive element-binding protein (ChREBP) have crucial roles in this process. Recently, insights have been gained into the signalling pathways that regulate these transcription factors. After feeding, high blood glucose and insulin levels activate lipogenic genes through several pathways, including the DNA-dependent protein kinase (DNA-PK), atypical protein kinase C (aPKC) and AKT-mTOR pathways. These pathways control the post-translational modifications of transcription factors and co-regulators, such as phosphorylation, acetylation or ubiquitylation, that affect their function, stability and/or localization. Dysregulation of lipogenesis can contribute to hepatosteatosis, which is associated with obesity and insulin resistance.


Assuntos
Ácidos Graxos/biossíntese , Lipogênese/genética , Lipoproteínas VLDL/biossíntese , Fígado/metabolismo , Transcrição Gênica/genética , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Proteína Quinase Ativada por DNA/metabolismo , Regulação da Expressão Gênica , Lipogênese/fisiologia , Receptores X do Fígado , Camundongos , Proteínas Nucleares/metabolismo , Receptores Nucleares Órfãos/metabolismo , Proteína Quinase C/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/metabolismo , Fatores Estimuladores Upstream/metabolismo
17.
Mol Cell ; 74(1): 45-58.e7, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30846317

RESUMO

Cells require a constant supply of fatty acids to survive and proliferate. Fatty acids incorporate into membrane and storage glycerolipids through a series of endoplasmic reticulum (ER) enzymes, but how these enzymes are regulated is not well understood. Here, using a combination of CRISPR-based genetic screens and unbiased lipidomics, we identified calcineurin B homologous protein 1 (CHP1) as a major regulator of ER glycerolipid synthesis. Loss of CHP1 severely reduces fatty acid incorporation and storage in mammalian cells and invertebrates. Mechanistically, CHP1 binds and activates GPAT4, which catalyzes the initial rate-limiting step in glycerolipid synthesis. GPAT4 activity requires CHP1 to be N-myristoylated, forming a key molecular interface between the two proteins. Interestingly, upon CHP1 loss, the peroxisomal enzyme, GNPAT, partially compensates for the loss of ER lipid synthesis, enabling cell proliferation. Thus, our work identifies a conserved regulator of glycerolipid metabolism and reveals plasticity in lipid synthesis of proliferating cells.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Retículo Endoplasmático/enzimologia , Glicerídeos/biossíntese , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Lipogênese , Células 3T3 , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proliferação de Células , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/patologia , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Glicerol-3-Fosfato O-Aciltransferase/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Células Jurkat , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Camundongos , Ácido Palmítico/toxicidade , Ligação Proteica
18.
Mol Cell ; 76(5): 811-825.e14, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31628041

RESUMO

Physical contact between organelles is vital to the function of eukaryotic cells. Lipid droplets (LDs) are dynamic organelles specialized in lipid storage that interact physically with mitochondria in several cell types. The mechanisms coupling these organelles are, however, poorly understood, and the cell-biological function of their interaction remains largely unknown. Here, we discover in adipocytes that the outer mitochondrial membrane protein MIGA2 links mitochondria to LDs. We identify an amphipathic LD-targeting motif and reveal that MIGA2 binds to the membrane proteins VAP-A or VAP-B in the endoplasmic reticulum (ER). We find that in adipocytes MIGA2 is involved in promoting triglyceride (TAG) synthesis from non-lipid precursors. Our data indicate that MIGA2 links reactions of de novo lipogenesis in mitochondria to TAG production in the ER, thereby facilitating efficient lipid storage in LDs. Based on its presence in many tissues, MIGA2 is likely critical for lipid and energy homeostasis in a wide spectrum of cell types.


Assuntos
Adipócitos/metabolismo , Lipogênese/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Células 3T3 , Adipócitos/fisiologia , Animais , Células COS , Diferenciação Celular/fisiologia , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Células HEK293 , Humanos , Gotículas Lipídicas/metabolismo , Lipogênese/genética , Proteínas de Membrana/fisiologia , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/fisiologia , Triglicerídeos/biossíntese , Proteínas de Transporte Vesicular/metabolismo
19.
Mol Cell ; 75(2): 357-371.e7, 2019 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-31227231

RESUMO

Carbohydrate response element binding protein (ChREBP) is a key transcriptional regulator of de novo lipogenesis (DNL) in response to carbohydrates and in hepatic steatosis. Mechanisms underlying nutrient modulation of ChREBP are under active investigation. Here we identify host cell factor 1 (HCF-1) as a previously unknown ChREBP-interacting protein that is enriched in liver biopsies of nonalcoholic steatohepatitis (NASH) patients. Biochemical and genetic studies show that HCF-1 is O-GlcNAcylated in response to glucose as a prerequisite for its binding to ChREBP and subsequent recruitment of OGT, ChREBP O-GlcNAcylation, and activation. The HCF-1:ChREBP complex resides at lipogenic gene promoters, where HCF-1 regulates H3K4 trimethylation to prime recruitment of the Jumonji C domain-containing histone demethylase PHF2 for epigenetic activation of these promoters. Overall, these findings define HCF-1's interaction with ChREBP as a previously unappreciated mechanism whereby glucose signals are both relayed to ChREBP and transmitted for epigenetic regulation of lipogenic genes.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Proteínas de Homeodomínio/genética , Fator C1 de Célula Hospedeira/genética , Lipogênese/genética , Hepatopatia Gordurosa não Alcoólica/genética , Animais , Carboidratos/genética , Epigênese Genética , Regulação da Expressão Gênica , Glucose/metabolismo , Hexosaminas/genética , Hexosaminas/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Hepatopatia Gordurosa não Alcoólica/patologia , Regiões Promotoras Genéticas/genética , Mapas de Interação de Proteínas/genética
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA