Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 363
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Cell Dev Biol ; 36: 115-139, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33021827

RESUMO

Lipid droplets (LDs) are endoplasmic reticulum-derived organelles that consist of a core of neutral lipids encircled by a phospholipid monolayer decorated with proteins. As hubs of cellular lipid and energy metabolism, LDs are inherently involved in the etiology of prevalent metabolic diseases such as obesity and nonalcoholic fatty liver disease. The functions of LDs are regulated by a unique set of associated proteins, the LD proteome, which includes integral membrane and peripheral proteins. These proteins control key activities of LDs such as triacylglycerol synthesis and breakdown, nutrient sensing and signal integration, and interactions with other organelles. Here we review the mechanisms that regulate the composition of the LD proteome, such as pathways that mediate selective and bulk LD protein degradation and potential connections between LDs and cellular protein quality control.


Assuntos
Gotículas Lipídicas/metabolismo , Proteínas/metabolismo , Animais , Autofagia , Humanos , Proteólise , Proteoma/metabolismo , Ubiquitina/metabolismo
2.
Mol Cell ; 79(1): 30-42.e4, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32473093

RESUMO

Autophagy is activated by prolonged fasting but cannot overcome the ensuing hepatic lipid overload, resulting in fatty liver. Here, we describe a peroxisome-lysosome metabolic link that restricts autophagic degradation of lipids. Acyl-CoA oxidase 1 (Acox1), the enzyme that catalyzes the first step in peroxisomal ß-oxidation, is enriched in liver and further increases with fasting or high-fat diet (HFD). Liver-specific Acox1 knockout (Acox1-LKO) protected mice against hepatic steatosis caused by starvation or HFD due to induction of autophagic degradation of lipid droplets. Hepatic Acox1 deficiency markedly lowered total cytosolic acetyl-CoA levels, which led to decreased Raptor acetylation and reduced lysosomal localization of mTOR, resulting in impaired activation of mTORC1, a central regulator of autophagy. Dichloroacetic acid treatment elevated acetyl-CoA levels, restored mTORC1 activation, inhibited autophagy, and increased hepatic triglycerides in Acox1-LKO mice. These results identify peroxisome-derived acetyl-CoA as a key metabolic regulator of autophagy that controls hepatic lipid homeostasis.


Assuntos
Acetilcoenzima A/metabolismo , Acil-CoA Oxidase/fisiologia , Autofagia , Ácidos Graxos/química , Fígado Gorduroso/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Peroxissomos/química , Acetilação , Animais , Proteína 5 Relacionada à Autofagia/fisiologia , Dieta Hiperlipídica/efeitos adversos , Jejum , Fígado Gorduroso/etiologia , Fígado Gorduroso/metabolismo , Feminino , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Oxirredução , Peroxissomos/metabolismo , Proteína Regulatória Associada a mTOR/genética , Proteína Regulatória Associada a mTOR/metabolismo
3.
Immunity ; 47(3): 466-480.e5, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28916263

RESUMO

Neutrophils are critical and short-lived mediators of innate immunity that require constant replenishment. Their differentiation in the bone marrow requires extensive cytoplasmic and nuclear remodeling, but the processes governing these energy-consuming changes are unknown. While previous studies show that autophagy is required for differentiation of other blood cell lineages, its function during granulopoiesis has remained elusive. Here, we have shown that metabolism and autophagy are developmentally programmed and essential for neutrophil differentiation in vivo. Atg7-deficient neutrophil precursors had increased glycolytic activity but impaired mitochondrial respiration, decreased ATP production, and accumulated lipid droplets. Inhibiting autophagy-mediated lipid degradation or fatty acid oxidation alone was sufficient to cause defective differentiation, while administration of fatty acids or pyruvate for mitochondrial respiration rescued differentiation in autophagy-deficient neutrophil precursors. Together, we show that autophagy-mediated lipolysis provides free fatty acids to support a mitochondrial respiration pathway essential to neutrophil differentiation.


Assuntos
Autofagia , Diferenciação Celular , Ácidos Graxos não Esterificados/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Adaptação Biológica , Animais , Análise por Conglomerados , Metabolismo Energético , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Glucose/metabolismo , Metabolismo dos Lipídeos , Lipólise , Mielopoese , Neutrófilos/ultraestrutura , Oxirredução , Ácido Pirúvico/metabolismo
4.
J Biol Chem ; 300(8): 107549, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39002673

RESUMO

Porcine epidemic diarrhea virus (PEDV) belongs to the Alphacoronavirus genus within the Coronavirus family, causing severe watery diarrhea in piglets and resulting in significant economic losses. Medium-chain acyl-CoA dehydrogenase (ACADM) is an enzyme participating in lipid metabolism associated with metabolic diseases and pathogen infections. Nonetheless, the precise role of ACADM in regulating PEDV replication remains uncertain. In this study, we identified ACADM as the host binding partner of NSP4 via immunoprecipitation-mass spectrometry analysis. The interaction between ACADM and NSP4 was subsequently corroborated through coimmunoprecipitation and laser confocal microscopy. Following this, a notable upsurge in ACADM expression was observed during PEDV infection. ACADM overexpression effectively inhibited virus replication, whereas ACADM knockdown facilitated virus replication, suggesting ACADM has negative regulation effect on PEDV infection. Furthermore, we demonstrated fatty acid ß-oxidation affected PEDV replication for the first time, inhibition of fatty acid ß-oxidation reduced PEDV replication. ACADM decreased PEDV-induced ß-oxidation to suppress PEDV replication. Mechanistically, ACADM reduced cellular free fatty acid levels and subsequent ß-oxidation by hindering AMPK-mediated lipophagy. In summary, our results reveal that ACADM plays a negative regulatory role in PEDV replication by regulating lipid metabolism. The present study introduces a novel approach for the prevention and control of PEDV infection.


Assuntos
Proteínas Quinases Ativadas por AMP , Oxirredução , Vírus da Diarreia Epidêmica Suína , Replicação Viral , Vírus da Diarreia Epidêmica Suína/fisiologia , Animais , Chlorocebus aethiops , Células Vero , Proteínas Quinases Ativadas por AMP/metabolismo , Suínos , Humanos , Acil-CoA Desidrogenase/metabolismo , Acil-CoA Desidrogenase/genética , Metabolismo dos Lipídeos , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/genética , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Ácidos Graxos/metabolismo , Células HEK293 , Ativação Enzimática
5.
J Biol Chem ; 299(8): 104946, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37348559

RESUMO

Dysregulated bile acid (BA)/lipid metabolism and gut bacteria dysbiosis are tightly associated with the development of obesity and non-alcoholic fatty liver disease (NAFLD). The orphan nuclear receptor, Small Heterodimer Partner (SHP/NR0B2), is a key regulator of BA/lipid metabolism, and its gene-regulating function is markedly enhanced by phosphorylation at Thr-58 mediated by a gut hormone, fibroblast growth factor-15/19 (FGF15/19). To investigate the role of this phosphorylation in whole-body energy metabolism, we generated transgenic SHP-T58A knock-in mice. Compared with wild-type (WT) mice, the phosphorylation-defective SHP-T58A mice gained weight more rapidly with decreased energy expenditure and increased lipid/BA levels. This obesity-prone phenotype was associated with the upregulation of lipid/BA synthesis genes and downregulation of lipophagy/ß-oxidation genes. Mechanistically, defective SHP phosphorylation selectively impaired its interaction with LRH-1, resulting in de-repression of SHP/LRH-1 target BA/lipid synthesis genes. Remarkably, BA composition and selective gut bacteria which are known to impact obesity, were also altered in these mice. Upon feeding a high-fat diet, fatty liver developed more severely in SHP-T58A mice compared to WT mice. Treatment with antibiotics substantially improved the fatty liver phenotypes in both groups but had greater effects in the T58A mice so that the difference between the groups was largely eliminated. These results demonstrate that defective phosphorylation at a single nuclear receptor residue can impact whole-body energy metabolism by altering BA/lipid metabolism and gut bacteria, promoting complex metabolic disorders like NAFLD. Since posttranslational modifications generally act in gene- and context-specific manners, the FGF15/19-SHP phosphorylation axis may allow more targeted therapy for NAFLD.


Assuntos
Ácidos e Sais Biliares , Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Ácidos e Sais Biliares/análise , Ácidos e Sais Biliares/genética , Lipídeos/sangue , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/microbiologia , Obesidade/microbiologia , Fosforilação , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Masculino , Antibacterianos/farmacologia
6.
Pflugers Arch ; 476(1): 59-74, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37910205

RESUMO

The formation of foam cells, lipid-loaded macrophages, is the hallmark event of atherosclerosis. Since cigarette smoking is a risk factor for developing atherosclerosis, the current study investigated the effects of cigarette smoke extract (CSE) on different events like expressions of genes involved in lipid influx and efflux, lipophagy, etc., that play vital roles in foam cell formation. The accumulation of lipids after CSE treatment U937 macrophage cells was examined by staining lipids with specific dyes: Oil red O and BODIPY493/503. Results showed an accumulation of lipids in CSE-treated cells, confirming foam cell formation by CSE treatment. To decipher the mechanism, the levels of CD36, an ox-LDL receptor, and ABCA1, an exporter of lipids, were examined in CSE-treated and -untreated U937 cells by real-time PCR and immunofluorescence analysis. Consistent with lipid accumulation, an increased level of CD36 and a reduction in ABCA1 were observed in CSE-treated cells. Moreover, CSE treatment caused inhibition of lipophagy-mediated lipid degradation by blocking lipid droplets (LDs)-lysosome fusion and increasing the lysosomal pH. CSE also impaired mitochondrial lipid oxidation. Thus, the present study demonstrates that CSE treatment affects lipid homeostasis by altering its influx and efflux, lysosomal degradation, and mitochondrial utilization, leading to the formation of lipid-loaded foam cells. Moreover, the current study also showed that the leucine supplement caused a significant reduction of CSE-induced foam cell formation in vitro. Thus, the current study provides insight into CS-induced atherosclerosis and an agent to combat the disease.


Assuntos
Aterosclerose , Fumar Cigarros , Humanos , Células Espumosas/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Gotículas Lipídicas/metabolismo , Células U937 , Aterosclerose/metabolismo
7.
EMBO J ; 39(3): e102525, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31919869

RESUMO

Extracellular vesicles are emerging key actors in adipocyte communication. Notably, small extracellular vesicles shed by adipocytes stimulate fatty acid oxidation and migration in melanoma cells and these effects are enhanced in obesity. However, the vesicular actors and cellular processes involved remain largely unknown. Here, we elucidate the mechanisms linking adipocyte extracellular vesicles to metabolic remodeling and cell migration. We show that adipocyte vesicles stimulate melanoma fatty acid oxidation by providing both enzymes and substrates. In obesity, the heightened effect of extracellular vesicles depends on increased transport of fatty acids, not fatty acid oxidation-related enzymes. These fatty acids, stored within lipid droplets in cancer cells, drive fatty acid oxidation upon being released by lipophagy. This increase in mitochondrial activity redistributes mitochondria to membrane protrusions of migrating cells, which is necessary to increase cell migration in the presence of adipocyte vesicles. Our results provide key insights into the role of extracellular vesicles in the metabolic cooperation that takes place between adipocytes and tumors with particular relevance to obesity.


Assuntos
Adipócitos/citologia , Vesículas Extracelulares/metabolismo , Ácidos Graxos/metabolismo , Melanoma/metabolismo , Obesidade/complicações , Células 3T3 , Adipócitos/metabolismo , Animais , Autofagia , Linhagem Celular Tumoral , Movimento Celular , Humanos , Metabolismo dos Lipídeos , Masculino , Camundongos , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Obesidade/metabolismo , Oxirredução
8.
J Cell Sci ; 135(5)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35260889

RESUMO

Lipophagy is a central cellular process for providing the cell with a readily utilized, high energy source of neutral lipids. Since its discovery over a decade ago, we are just starting to understand the molecular components that drive lipophagy, how it is activated in response to nutrient availability, and its potential as a therapeutic target in disease. In this Cell Science at a Glance article and the accompanying poster, we first provide a brief overview of the different structural and enzymatic proteins that comprise the lipid droplet (LD) proteome and reside within the limiting phospholipid monolayer of this complex organelle. We then highlight key players in the catabolic breakdown of LDs during the functionally linked lipolysis and lipophagy processes. Finally, we discuss what is currently known about macro- and micro-lipophagy based on findings in yeast, mammalian and other model systems, and how impairment of these important functions can lead to disease states.


Assuntos
Gotículas Lipídicas , Lipólise , Animais , Autofagia/fisiologia , Gotículas Lipídicas/metabolismo , Metabolismo dos Lipídeos/fisiologia , Mamíferos/metabolismo , Fosfolipídeos/metabolismo , Proteínas/metabolismo , Saccharomyces cerevisiae/metabolismo
9.
Small ; 20(27): e2308148, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38290809

RESUMO

Hexagonal boron nitride (hBN) is an emerging 2D material attracting significant attention due to its superior electrical, chemical, and therapeutic properties. However, inhalation toxicity mechanisms of hBN in human lung cells are poorly understood. Here, cellular interaction and effects of hBN nanosheets is investigated in alveolar epithelial cells cultured on porous inserts and exposed under air-liquid interface conditions for 24 h. hBN is taken up by the cells as determined in a label-free manner via RAMAN-confocal microscopy, ICP-MS, TEM, and SEM-EDX. No significant (p > 0.05) effects are observed on cell membrane integrity (LDH release), epithelial barrier integrity (TEER), interleukin-8 cytokine production or reactive oxygen production at tested dose ranges (1, 5, and 10 µg cm-2). However, it is observed that an enhanced accumulation of lipid granules in cells indicating the effect of hBN on lipid metabolism. In addition, it is observed that a significant (p < 0.05) and dose-dependent (5 and 10 µg cm-2) induction of autophagy in cells after exposure to hBN, potentially associated with the downstream processing and breakdown of excess lipid granules to maintain lipid homeostasis. Indeed, lysosomal co-localization of lipid granules supporting this argument is observed. Overall, the results suggest that the continuous presence of excess intracellular lipids may provoke adverse outcomes in the lungs.


Assuntos
Células Epiteliais Alveolares , Autofagia , Compostos de Boro , Humanos , Compostos de Boro/química , Compostos de Boro/farmacologia , Autofagia/efeitos dos fármacos , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/efeitos dos fármacos , Nanoestruturas/química , Metabolismo dos Lipídeos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
10.
New Phytol ; 243(1): 284-298, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38730535

RESUMO

Autophagy is a central degradative pathway highly conserved among eukaryotes, including microalgae, which remains unexplored in extremophilic organisms. In this study, we described and characterized autophagy in the newly identified extremophilic green microalga Chlamydomonas urium, which was isolated from an acidic environment. The nuclear genome of C. urium was sequenced, assembled and annotated in order to identify autophagy-related genes. Transmission electron microscopy, immunoblotting, metabolomic and photosynthetic analyses were performed to investigate autophagy in this extremophilic microalga. The analysis of the C. urium genome revealed the conservation of core autophagy-related genes. We investigated the role of autophagy in C. urium by blocking autophagic flux with the vacuolar ATPase inhibitor concanamycin A. Our results indicated that inhibition of autophagic flux in this microalga resulted in a pronounced accumulation of triacylglycerols and lipid droplets (LDs). Metabolomic and photosynthetic analyses indicated that C. urium cells with impaired vacuolar function maintained an active metabolism. Such effects were not observed in the neutrophilic microalga Chlamydomonas reinhardtii. Inhibition of autophagic flux in C. urium uncovered an active recycling of LDs through lipophagy, a selective autophagy pathway for lipid turnover. This study provided the metabolic basis by which extremophilic algae are able to catabolize lipids in the vacuole.


Assuntos
Autofagia , Chlamydomonas , Metabolismo dos Lipídeos , Fotossíntese , Chlamydomonas/metabolismo , Fotossíntese/efeitos dos fármacos , Extremófilos/metabolismo , Gotículas Lipídicas/metabolismo , Vacúolos/metabolismo , Filogenia , Triglicerídeos/metabolismo , Macrolídeos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA