Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunohorizons ; 6(12): 837-850, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36547387

RESUMEN

Hematopoiesis integrates cytokine signaling, metabolism, and epigenetic modifications to regulate blood cell generation. These processes are linked, as metabolites provide essential substrates for epigenetic marks. In this study, we demonstrate that ATP citrate lyase (Acly), which metabolizes citrate to generate cytosolic acetyl-CoA and is of clinical interest, can regulate chromatin accessibility to limit myeloid differentiation. Acly was tested for a role in murine hematopoiesis by small-molecule inhibition or genetic deletion in lineage-depleted, c-Kit-enriched hematopoietic stem and progenitor cells from Mus musculus. Treatments increased the abundance of cell populations that expressed the myeloid integrin CD11b and other markers of myeloid differentiation. When single-cell RNA sequencing was performed, we found that Acly inhibitor-treated hematopoietic stem and progenitor cells exhibited greater gene expression signatures for macrophages and enrichment of these populations. Similarly, the single-cell assay for transposase-accessible chromatin sequencing showed increased chromatin accessibility at genes associated with myeloid differentiation, including CD11b, CD11c, and IRF8. Mechanistically, Acly deficiency altered chromatin accessibility and expression of multiple C/EBP family transcription factors known to regulate myeloid differentiation and cell metabolism, with increased Cebpe and decreased Cebpa and Cebpb. This effect of Acly deficiency was accompanied by altered mitochondrial metabolism with decreased mitochondrial polarization but increased mitochondrial content and production of reactive oxygen species. The bias to myeloid differentiation appeared due to insufficient generation of acetyl-CoA, as exogenous acetate to support alternate compensatory pathways to produce acetyl-CoA reversed this phenotype. Acly inhibition thus can promote myelopoiesis through deprivation of acetyl-CoA and altered histone acetylome to regulate C/EBP transcription factor family activity for myeloid differentiation.


Asunto(s)
ATP Citrato (pro-S)-Liasa , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Mielopoyesis , Animales , Ratones , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , ATP Citrato (pro-S)-Liasa/deficiencia , ATP Citrato (pro-S)-Liasa/genética , Cromatina/metabolismo , Mielopoyesis/genética
2.
Nat Commun ; 11(1): 6296, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293558

RESUMEN

Macrophages represent a major immune cell population in atherosclerotic plaques and play central role in the progression of this lipid-driven chronic inflammatory disease. Targeting immunometabolism is proposed as a strategy to revert aberrant macrophage activation to improve disease outcome. Here, we show ATP citrate lyase (Acly) to be activated in inflammatory macrophages and human atherosclerotic plaques. We demonstrate that myeloid Acly deficiency induces a stable plaque phenotype characterized by increased collagen deposition and fibrous cap thickness, along with a smaller necrotic core. In-depth functional, lipidomic, and transcriptional characterization indicate deregulated fatty acid and cholesterol biosynthesis and reduced liver X receptor activation within the macrophages in vitro. This results in macrophages that are more prone to undergo apoptosis, whilst maintaining their capacity to phagocytose apoptotic cells. Together, our results indicate that targeting macrophage metabolism improves atherosclerosis outcome and we reveal Acly as a promising therapeutic target to stabilize atherosclerotic plaques.


Asunto(s)
ATP Citrato (pro-S)-Liasa/deficiencia , Macrófagos/metabolismo , Placa Aterosclerótica/inmunología , ATP Citrato (pro-S)-Liasa/antagonistas & inhibidores , ATP Citrato (pro-S)-Liasa/genética , Anciano , Animales , Apoptosis/inmunología , Colesterol/biosíntesis , Colágeno/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Ácidos Grasos/biosíntesis , Femenino , Fibrosis , Perfilación de la Expresión Génica , Humanos , Lipidómica , Lipogénesis/inmunología , Receptores X del Hígado/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Masculino , Ratones Noqueados , Necrosis/inmunología , Necrosis/patología , Fagocitosis , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/patología
3.
PLoS Biol ; 18(11): e3000981, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33253182

RESUMEN

The metabolite acetyl-coenzyme A (acetyl-CoA) serves as an essential element for a wide range of cellular functions including adenosine triphosphate (ATP) production, lipid synthesis, and protein acetylation. Intracellular acetyl-CoA concentrations are associated with nutrient availability, but the mechanisms by which a cell responds to fluctuations in acetyl-CoA levels remain elusive. Here, we generate a cell system to selectively manipulate the nucleo-cytoplasmic levels of acetyl-CoA using clustered regularly interspaced short palindromic repeat (CRISPR)-mediated gene editing and acetate supplementation of the culture media. Using this system and quantitative omics analyses, we demonstrate that acetyl-CoA depletion alters the integrity of the nucleolus, impairing ribosomal RNA synthesis and evoking the ribosomal protein-dependent activation of p53. This nucleolar remodeling appears to be mediated through the class IIa histone deacetylases (HDACs). Our findings highlight acetylation-mediated control of the nucleolus as an important hub linking acetyl-CoA fluctuations to cellular stress responses.


Asunto(s)
Acetilcoenzima A/biosíntesis , Nucléolo Celular/metabolismo , ATP Citrato (pro-S)-Liasa/deficiencia , ATP Citrato (pro-S)-Liasa/genética , ATP Citrato (pro-S)-Liasa/metabolismo , Acetatos/metabolismo , Acetilación , Línea Celular , Nucléolo Celular/ultraestructura , Expresión Génica , Técnicas de Inactivación de Genes , Células HCT116 , Histona Desacetilasas/metabolismo , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Ribosómicas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
4.
Nature ; 579(7800): 586-591, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32214246

RESUMEN

Consumption of fructose has risen markedly in recent decades owing to the use of sucrose and high-fructose corn syrup in beverages and processed foods1, and this has contributed to increasing rates of obesity and non-alcoholic fatty liver disease2-4. Fructose intake triggers de novo lipogenesis in the liver4-6, in which carbon precursors of acetyl-CoA are converted into fatty acids. The ATP citrate lyase (ACLY) enzyme cleaves cytosolic citrate to generate acetyl-CoA, and is upregulated after consumption of carbohydrates7. Clinical trials are currently pursuing the inhibition of ACLY as a treatment for metabolic diseases8. However, the route from dietary fructose to hepatic acetyl-CoA and lipids remains unknown. Here, using in vivo isotope tracing, we show that liver-specific deletion of Acly in mice is unable to suppress fructose-induced lipogenesis. Dietary fructose is converted to acetate by the gut microbiota9, and this supplies lipogenic acetyl-CoA independently of ACLY10. Depletion of the microbiota or silencing of hepatic ACSS2, which generates acetyl-CoA from acetate, potently suppresses the conversion of bolus fructose into hepatic acetyl-CoA and fatty acids. When fructose is consumed more gradually to facilitate its absorption in the small intestine, both citrate cleavage in hepatocytes and microorganism-derived acetate contribute to lipogenesis. By contrast, the lipogenic transcriptional program is activated in response to fructose in a manner that is independent of acetyl-CoA metabolism. These data reveal a two-pronged mechanism that regulates hepatic lipogenesis, in which fructolysis within hepatocytes provides a signal to promote the expression of lipogenic genes, and the generation of microbial acetate feeds lipogenic pools of acetyl-CoA.


Asunto(s)
Acetatos/metabolismo , Azúcares de la Dieta/metabolismo , Fructosa/metabolismo , Microbioma Gastrointestinal/fisiología , Lipogénesis , Hígado/metabolismo , ATP Citrato (pro-S)-Liasa/deficiencia , ATP Citrato (pro-S)-Liasa/genética , ATP Citrato (pro-S)-Liasa/metabolismo , Acetato CoA Ligasa/deficiencia , Acetato CoA Ligasa/genética , Acetato CoA Ligasa/metabolismo , Acetilcoenzima A/metabolismo , Animales , Ácido Cítrico/metabolismo , Azúcares de la Dieta/administración & dosificación , Azúcares de la Dieta/farmacología , Ácidos Grasos/metabolismo , Fructosa/administración & dosificación , Fructosa/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Hepatocitos/metabolismo , Marcaje Isotópico , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Hígado/citología , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratones , Especificidad por Sustrato
5.
Cell Rep ; 17(4): 1037-1052, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27760311

RESUMEN

Mechanisms of metabolic flexibility enable cells to survive under stressful conditions and can thwart therapeutic responses. Acetyl-coenzyme A (CoA) plays central roles in energy production, lipid metabolism, and epigenomic modifications. Here, we show that, upon genetic deletion of Acly, the gene coding for ATP-citrate lyase (ACLY), cells remain viable and proliferate, although at an impaired rate. In the absence of ACLY, cells upregulate ACSS2 and utilize exogenous acetate to provide acetyl-CoA for de novo lipogenesis (DNL) and histone acetylation. A physiological level of acetate is sufficient for cell viability and abundant acetyl-CoA production, although histone acetylation levels remain low in ACLY-deficient cells unless supplemented with high levels of acetate. ACLY-deficient adipocytes accumulate lipid in vivo, exhibit increased acetyl-CoA and malonyl-CoA production from acetate, and display some differences in fatty acid content and synthesis. Together, these data indicate that engagement of acetate metabolism is a crucial, although partial, mechanism of compensation for ACLY deficiency.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Acetatos/metabolismo , Glucosa/metabolismo , ATP Citrato (pro-S)-Liasa/deficiencia , Acetato CoA Ligasa/metabolismo , Acetatos/farmacología , Acetilcoenzima A/metabolismo , Acetilación , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Eliminación de Gen , Histonas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lípidos/biosíntesis , Masculino , Ratones , Regulación hacia Arriba/efectos de los fármacos
6.
Biochim Biophys Acta ; 1851(9): 1107-17, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25959598

RESUMEN

The role of the two key enzymes of fatty acid (FA) synthesis, ATP-citrate lyase (Acl) and malic enzyme (Mae), was analyzed in the oleaginous yeast Yarrowia lipolytica. In most oleaginous yeasts, Acl and Mae are proposed to provide, respectively, acetyl-CoA and NADPH for FA synthesis. Acl was mainly studied at the biochemical level but no strain depleted for this enzyme was analyzed in oleaginous microorganisms. On the other hand the role of Mae in FA synthesis in Y. lipolytica remains unclear since it was proposed to be a mitochondrial NAD(H)-dependent enzyme and not a cytosolic NADP(H)-dependent enzyme. In this study, we analyzed for the first time strains inactivated for corresponding genes. Inactivation of ACL1 decreases FA synthesis by 60 to 80%, confirming its essential role in FA synthesis in Y. lipolytica. Conversely, inactivation of MAE1 has no effects on FA synthesis, except in a FA overaccumulating strain where it improves FA synthesis by 35%. This result definitively excludes Mae as a major key enzyme for FA synthesis in Y. lipolytica. During the analysis of both mutants, we observed a negative correlation between FA and mannitol level. As mannitol and FA pathways may compete for carbon storage, we inactivated YlSDR, encoding a mannitol dehydrogenase converting fructose and NADPH into mannitol and NADP+. The FA content of the resulting mutant was improved by 60% during growth on fructose, demonstrating that mannitol metabolism may modulate FA synthesis in Y. lipolytica.


Asunto(s)
ATP Citrato (pro-S)-Liasa/metabolismo , Ácidos Grasos/metabolismo , Proteínas Fúngicas/metabolismo , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Malato Deshidrogenasa/metabolismo , Yarrowia/metabolismo , ATP Citrato (pro-S)-Liasa/deficiencia , ATP Citrato (pro-S)-Liasa/genética , Acetilcoenzima A/metabolismo , Fructosa/metabolismo , Proteínas Fúngicas/genética , Metabolismo de los Lípidos/genética , Malato Deshidrogenasa/deficiencia , Malato Deshidrogenasa/genética , Manitol/metabolismo , Manitol Deshidrogenasas/deficiencia , Manitol Deshidrogenasas/genética , Manitol Deshidrogenasas/metabolismo , NADP/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Yarrowia/genética
7.
Mol Microbiol ; 86(6): 1404-23, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23078142

RESUMEN

The interaction of Cryptococcus neoformans with phagocytic cells of the innate immune system is a key step in disseminated disease leading to meningoencephalitis in immunocompromised individuals. Transcriptional profiling of cryptococcal cells harvested from cell culture medium or from macrophages found differential expression of metabolic and other functions during fungal adaptation to the intracellular environment. We focused on the ACL1 gene for ATP-citrate lyase, which converts citrate to acetyl-CoA, because this gene showed elevated transcript levels in macrophages and because of the importance of acetyl-CoA as a central metabolite. Mutants lacking ACL1 showed delayed growth on medium containing glucose, reduced cellular levels of acetyl-CoA, defective production of virulence factors, increased susceptibility to the antifungal drug fluconazole and decreased survival within macrophages. Importantly, acl1 mutants were unable to cause disease in a murine inhalation model, a phenotype that was more extreme than other mutants with defects in acetyl-CoA production (e.g. an acetyl-CoA synthetase mutant). Loss of virulence is likely due to perturbation of critical physiological interconnections between virulence factor expression and metabolism in C. neoformans. Phylogenetic analysis and structural modelling of cryptococcal Acl1 identified three indels unique to fungal protein sequences; these differences may provide opportunities for the development of pathogen-specific inhibitors.


Asunto(s)
ATP Citrato (pro-S)-Liasa/deficiencia , Acetilcoenzima A/metabolismo , Cryptococcus neoformans/metabolismo , Cryptococcus neoformans/patogenicidad , Factores de Virulencia/metabolismo , ATP Citrato (pro-S)-Liasa/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Ácido Cítrico/metabolismo , Criptococosis/microbiología , Criptococosis/patología , Cryptococcus neoformans/enzimología , Cryptococcus neoformans/genética , Medios de Cultivo/química , Modelos Animales de Enfermedad , Glucosa/metabolismo , Mutación INDEL , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Viabilidad Microbiana , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Homología de Secuencia de Aminoácido , Virulencia
8.
Biochim Biophys Acta ; 1780(7-8): 966-72, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18439432

RESUMEN

We hypothesized that contrasting leucine with its non-metabolizable analog 2-aminobicyclo[2,2,1]heptane-2-carboxylic acid (BCH) might provide new information about metabolic pathways involved in insulin secretion. Both compounds stimulate insulin secretion by allosterically activating glutamate dehydrogenase, which enhances glutamate metabolism. However, we found that leucine was a stronger secretagogue in rat pancreatic islets and INS-1 cells. This suggested that leucine's metabolism contributed to its insulinotropism. Indeed, we found that leucine increased acetoacetate and was metabolized to CO(2) in pancreatic islets and increased short chain acyl-CoAs (SC-CoAs) in INS-1 cells. We then used the leucine-BCH difference to study the hypothesis that acyl groups derived from secretagogue carbon can be transferred as acetoacetate, in addition to citrate, from mitochondria to the cytosol where they can be converted to SC-CoAs. Since BCH cannot form sufficient acetoacetate from glutamate, transport of any glutamate-derived acyl groups to the cytosol in BCH-stimulated cells must proceed mainly via citrate. In ATP citrate lyase-deficient INS-1 cells, which are unable to convert citrate into cytosolic acetyl-CoA, insulin release by BCH was decreased and adding beta-hydroxybutyrate or alpha-ketoisocaproate, which increases mitochondrial acetoacetate, normalized BCH-induced insulin release. This strengthens the concept that acetoacetate-transferred acyl carbon can be converted to cytosolic SC-CoAs to stimulate insulin secretion.


Asunto(s)
Ácido 3-Hidroxibutírico/metabolismo , ATP Citrato (pro-S)-Liasa/deficiencia , Acetoacetatos/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Leucina/metabolismo , Ácido 3-Hidroxibutírico/farmacología , ATP Citrato (pro-S)-Liasa/genética , ATP Citrato (pro-S)-Liasa/metabolismo , Acetoacetatos/farmacología , Animales , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Glucosa/metabolismo , Glucosa/farmacología , Glutamato Deshidrogenasa/análisis , Glutamina/metabolismo , Glutamina/farmacología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/citología , Ácidos Cetoglutáricos/análisis , Leucina/farmacología , Modelos Biológicos , Ratas , Ratas Sprague-Dawley
9.
J Biol Chem ; 279(10): 9557-64, 2004 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-14662765

RESUMEN

ATP-citrate lyase (Acly) is one of two cytosolic enzymes that synthesize acetyl-coenzyme A (CoA). Because acetyl-CoA is an essential building block for cholesterol and triglycerides, Acly has been considered a therapeutic target for hyperlipidemias and obesity. To define the phenotype of Acly-deficient mice, we created Acly knockout mice in which a beta-galactosidase marker is expressed from Acly regulatory sequences. We also sought to define the cell type-specific expression patterns of Acly to further elucidate the in vivo roles of the enzyme. Homozygous Acly knockout mice died early in development. Heterozygous mice were healthy, fertile, and normolipidemic on both chow and high fat diets, despite expressing half-normal amounts of Acly mRNA and protein. Fibroblasts and hepatocytes from heterozygous Acly mice contained half-normal amounts of Acly mRNA and protein, but this did not perturb triglyceride and cholesterol synthesis or the expression of lipid biosynthetic genes regulated by sterol regulatory element-binding proteins. The expression of acetyl-CoA synthetase 1, another cytosolic enzyme for producing acetyl-CoA, was not up-regulated. As judged by beta-galactosidase staining, Acly was expressed ubiquitously but was expressed particularly highly in tissues with high levels of lipogenesis, such as in the livers of mice fed a high-carbohydrate diet. beta-Galactosidase staining was intense in the developing brain, in keeping with the high levels of de novo lipogenesis of the tissue. In the adult brain, beta-galactosidase staining was in general much lower, consistent with reduced levels of lipogenesis; however, beta-galactosidase expression remained very high in cholinergic neurons, likely reflecting the importance of Acly in generating acetyl-CoA for acetylcholine synthesis. The Acly knockout allele is useful for identifying cell types with a high demand for acetyl-CoA synthesis.


Asunto(s)
ATP Citrato (pro-S)-Liasa/deficiencia , Acetilcoenzima A/biosíntesis , ATP Citrato (pro-S)-Liasa/genética , Animales , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...