Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.875
Filtrar
1.
Nat Commun ; 15(1): 7746, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39232011

RESUMEN

Beige fat activation involves a fuel switch to fatty acid oxidation following chronic cold adaptation. Mitochondrial acyl-CoA synthetase long-chain family member 1 (ACSL1) localizes in the mitochondria and plays a key role in fatty acid oxidation; however, the regulatory mechanism of the subcellular localization remains poorly understood. Here, we identify an endosomal trafficking component sortilin (encoded by Sort1) in adipose tissues that shows dynamic expression during beige fat activation and facilitates the translocation of ACSL1 from the mitochondria to the endolysosomal pathway for degradation. Depletion of sortilin in adipocytes results in an increase of mitochondrial ACSL1 and the activation of AMPK/PGC1α signaling, thereby activating beige fat and preventing high-fat diet (HFD)-induced obesity and insulin resistance. Collectively, our findings indicate that sortilin controls adipose tissue fatty acid oxidation by substrate fuel selection during beige fat activation and provides a potential targeted approach for the treatment of metabolic diseases.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Coenzima A Ligasas , Dieta Alta en Grasa , Metabolismo Energético , Mitocondrias , Termogénesis , Animales , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Masculino , Mitocondrias/metabolismo , Ratones , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Ratones Endogámicos C57BL , Obesidad/metabolismo , Obesidad/genética , Adipocitos/metabolismo , Tejido Adiposo Beige/metabolismo , Resistencia a la Insulina , Ácidos Grasos/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Transporte de Proteínas , Oxidación-Reducción , Transducción de Señal , Células 3T3-L1 , Ratones Noqueados
2.
Biol Direct ; 19(1): 68, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39160584

RESUMEN

BACKGROUND: Cancer-associated fibroblasts (CAFs) have been reported that can affect cancer cell proliferation, metastasis, ferroptosis, and immune escape. METTL3-mediated N6-methyladenine (m6A) modification is involved in the tumorigenesis of colorectal cancer (CRC). Herein, we investigated whether METTL3-dependent m6A in CAFs-derived exosomes (exo) affected CRC progression. METHODS: qRT-PCR and western blotting analyses detected levels of mRNAs and proteins. Cell proliferation and metastasis were evaluated using MTT, colony formation, transwell, and wound healing assays, respectively. Cell ferroptosis was assessed by detecting cell viability and the levels of Fe+, reactive oxygen species, and glutathione after erastin treatment. Exosomes were isolated from CAFs by ultracentrifugation. The m6A modification profile was determined by methylated RNA immunoprecipitation assay and the interaction between METTL3 and ACSL3 (acyl-CoA synthetase 3) was verified using dual-luciferase reporter assay. Animal models were established for in vivo analysis. RESULTS: CAFs promoted CRC cell proliferation and metastasis, and suppressed cell ferroptosis. METTL3 was enriched in CAFs and was packaged into exosomes. The m6A modification and METTL3 expression were increased in CRC samples. Knockdown of METTL3 in CAFs-exo suppressed CRC cell proliferation and metastasis, and induced cell ferroptosis. Mechanistically, METTL3 induced ACSL3 m6A modification and stabilized its expression. The anticancer effects mediated by METTL3-silenced CAFs-exo could be rescued by ACSL3 overexpression. Moreover, in vivo assay also showed that CAFs-exo with decreased METTL3 could hinder CRC growth and metastasis in mice models. CONCLUSION: CAFs promoted the proliferation and metastasis, and restrained the ferroptosis in CRC by exosomal METTL3-elicited ACSL3 m6A modification.


Asunto(s)
Fibroblastos Asociados al Cáncer , Proliferación Celular , Coenzima A Ligasas , Neoplasias Colorrectales , Exosomas , Ferroptosis , Metiltransferasas , Ferroptosis/genética , Metiltransferasas/metabolismo , Metiltransferasas/genética , Exosomas/metabolismo , Exosomas/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Ratones , Fibroblastos Asociados al Cáncer/metabolismo , Animales , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Metástasis de la Neoplasia , Línea Celular Tumoral , Ratones Desnudos , Masculino
3.
ACS Synth Biol ; 13(8): 2611-2620, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39092606

RESUMEN

3-Hydroxypropionic acid (3-HP) is a highly sought-after platform chemical serving as a precursor to a variety of high value-added chemical products. In this study, we designed and constructed a novel light-powered in vitro synthetic enzymatic biosystem comprising acetyl-CoA ligase, acetyl-CoA carboxylase, malonyl-CoA reductase, and phosphotransferase to efficiently produce 3-HP through CO2 fixation from acetate, a cost-effective and readily available substrate. The system employed natural thylakoid membranes (TMs) for the regeneration of adenosine triphosphate and nicotinamide adenine dinucleotide phosphate. Comprehensive investigations were conducted on the effects of buffer solutions, substrate concentrations, enzyme loading levels, and TMs loading levels to optimize the yield of 3-HP. Following optimization, a production of 0.46 mM 3-HP was achieved within 6 h from an initial 0.5 mM acetate, with a yield nearing 92%. This work underscores the simplicity of 3-HP production via an in vitro biomanufacturing platform and highlights the potential for incorporating TMs as a sustainable and environmentally friendly approach in biomanufacturing processes.


Asunto(s)
Acetil-CoA Carboxilasa , Dióxido de Carbono , Ácido Láctico , Dióxido de Carbono/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Ácido Láctico/metabolismo , Ácido Láctico/análogos & derivados , Luz , Tilacoides/metabolismo , Adenosina Trifosfato/metabolismo , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Acetatos/metabolismo , Acetatos/química , Oxidorreductasas
4.
Commun Biol ; 7(1): 1027, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39169121

RESUMEN

The retina is light-sensitive neuronal tissue in the back of the eye. The phospholipid composition of the retina is unique and highly enriched in polyunsaturated fatty acids, including docosahexaenoic fatty acid (DHA). While it is generally accepted that a high DHA content is important for vision, surprisingly little is known about the mechanisms of DHA enrichment in the retina. Furthermore, the biological processes controlled by DHA in the eye remain poorly defined as well. Here, we combined genetic manipulations with lipidomic analysis in mice to demonstrate that acyl-CoA synthetase 6 (Acsl6) serves as a regulator of the unique composition of retinal membranes. Inactivation of Acsl6 reduced the levels of DHA-containing phospholipids, led to progressive loss of light-sensitive rod photoreceptor neurons, attenuated the light responses of these cells, and evoked distinct transcriptional response in the retina involving the Srebf1/2 (sterol regulatory element binding transcription factors 1/2) pathway. This study identifies one of the major enzymes responsible for DHA enrichment in the retinal membranes and introduces a model allowing an evaluation of rod functioning and pathology caused by impaired DHA incorporation/retention in the retina.


Asunto(s)
Coenzima A Ligasas , Fosfolípidos , Células Fotorreceptoras Retinianas Bastones , Animales , Células Fotorreceptoras Retinianas Bastones/metabolismo , Ratones , Fosfolípidos/metabolismo , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Retina/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Ratones Noqueados , Ratones Endogámicos C57BL
5.
Sci Rep ; 14(1): 19591, 2024 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-39179766

RESUMEN

Liver cancer is the 4th most lethal form of cancer with a poor prognosis for patients worldwide. Dysregulation of lipid metabolism is related to FA oxidation alternation which can be modified by peroxisome proliferator-activated receptor-α (PPARα). Therefore, it is important to identify the lipid metabolism-related genes regulated by PPARα in liver cancer. Hub genes related to the lipid metabolism pathway of HCC samples treated with PPARα agonist (WY-14,643) were identified through a weighted gene co-expression network analysis (WGCNA). Gene expression and clinical information were obtained from the Gene Expression Omnibus (GEO) database. The network of top main hub genes was visualized by the Cytoscape software using MCODE and CytoHubba plugins. Finally, the expression and clinical association of each hub gene were evaluated using enrichment analysis, TCGA data, GEPIA, GSCA, and q-PCR. Based on our results, the top 5 co-expressed genes including (CPT2, ACSL1, ACSL3, ACOX1, and SLC27A2) were selected as the main hub genes participating in fatty acid metabolism, fatty acid beta-oxidation, and PPAR signaling pathway. All association of higher ACSL3 expression with lower outcomes and survival rates was detected in HCC patients. Therefore, lipid metabolism-related Hub genes regulated by PPARα are potential biomarkers, and they may offer a therapeutical foundation for targeted therapy directed against the HCC antitumor strategy.


Asunto(s)
Carcinoma Hepatocelular , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Metabolismo de los Lípidos , Neoplasias Hepáticas , PPAR alfa , Humanos , PPAR alfa/agonistas , PPAR alfa/genética , PPAR alfa/metabolismo , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/efectos de los fármacos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Perfilación de la Expresión Génica/métodos , Pronóstico , Biología Computacional/métodos
6.
PLoS One ; 19(8): e0307802, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39178212

RESUMEN

Prolonged consumption of diet rich in fats is regarded as the major factor leading to the insulin resistance (IR) and type 2 diabetes (T2D). Emerging evidence link excessive accumulation of bioactive lipids such as diacylglycerol (DAG) and ceramide (Cer), with impairment of insulin signaling in skeletal muscle. Until recently, little has been known about the involvement of long-chain acyl-CoAs synthetases in the above mechanism. To examine possible role of long-chain acyl-coenzyme A synthetase 1 (Acsl1) (a major muscular ACSL isoform) in mediating HFD-induced IR we locally silenced Acsl1 in gastrocnemius of high-fat diet (HFD)-fed C57BL/6J mice through electroporation-delivered shRNA and compared it to non-silenced tissue within the same animal. Acsl1 down-regulation decreased the content of muscular long-chain acyl-CoA (LCACoA) and both the Cer (C18:1-Cer and C24:1-Cer) and DAG (C16:0/18:0-DAG, C16:0/18:2-DAG, C18:0/18:0-DAG) and simultaneously improved insulin sensitivity and glucose uptake as compared with non-silenced tissue. Acsl1 down-regulation decreased expression of mitochondrial ß-oxidation enzymes, and the content of both the short-chain acylcarnitine (SCA-Car) and short-chain acyl-CoA (SCACoA) in muscle, pointing towards reduction of mitochondrial FA oxidation. The results indicate, that beneficial effects of Acsl1 partial ablation on muscular insulin sensitivity are connected with inhibition of Cer and DAG accumulation, and outweigh detrimental impact of decreased mitochondrial fatty acids metabolism in skeletal muscle of obese HFD-fed mice.


Asunto(s)
Coenzima A Ligasas , Dieta Alta en Grasa , Diglicéridos , Regulación hacia Abajo , Resistencia a la Insulina , Ratones Endogámicos C57BL , Músculo Esquelético , Obesidad , ARN Interferente Pequeño , Animales , Músculo Esquelético/metabolismo , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Ratones , Dieta Alta en Grasa/efectos adversos , Obesidad/metabolismo , Obesidad/genética , Diglicéridos/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Masculino , Ceramidas/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Insulina/metabolismo , Ratones Obesos
7.
Cell Death Dis ; 15(8): 611, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39174525

RESUMEN

Diffuse large B-cell lymphoma (DLBCL), an invasive lymphoma with substantial heterogeneity, can be mainly categorised into germinal centre B-cell-like (GCB) and non-GCB subtypes. DLBCL cells are highly susceptible to ferroptosis, which offers an effective avenue for treating recurrent and refractory DLBCL. Moreover, various heat shock proteins are involved in regulating the sensitivity of tumour cells to ferroptosis. Among these proteins, tailless complex polypeptide 1 (TCP1), a subunit of chaperonin-containing T-complex protein-1 (CCT), plays a role in tumour proliferation and survival. Therefore, we explored the role of TCP1 in different DLBCL subtypes, the sensitivity of GCB and non-GCB subtypes to the ferroptosis inducer RAS-selective lethal small molecule 3 (RSL3), and the underlying molecular mechanism. In GCB cells, TCP1 promoted RSL3-induced ferroptosis. Notably, TCP1 could bind with acyl-CoA synthetase long-chain family member 4 (ACSL4), a key enzyme regulating lipid composition and facilitating ferroptosis, to reduce its ubiquitination and degradation. This interaction activated the ACSL4/LPCAT3 signalling pathway and promoted ferroptosis in the GCB subtype. However, in the non-GCB subtype, TCP1 did not act as a positive regulator but served as a predictor of an unfavourable prognosis in patients with non-GCB. In conclusion, our results suggest that in DLBCL, high TCP1 expression enhances the sensitivity of GCB tumour cells to ferroptosis and serves as a marker of poor prognosis in patients with non-GCB DLBCL.


Asunto(s)
Chaperonina con TCP-1 , Coenzima A Ligasas , Ferroptosis , Linfoma de Células B Grandes Difuso , Ferroptosis/genética , Humanos , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/genética , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Pronóstico , Línea Celular Tumoral , Chaperonina con TCP-1/metabolismo , Chaperonina con TCP-1/genética , Animales , Ratones , Femenino , Regulación Neoplásica de la Expresión Génica , Masculino , Carbolinas
8.
Appl Microbiol Biotechnol ; 108(1): 435, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39126431

RESUMEN

Naringenin is a plant polyphenol, widely explored due to its interesting biological activities, namely anticancer, antioxidant, and anti-inflammatory. Due to its potential applications and attempt to overcome the industrial demand, there has been an increased interest in its heterologous production. The microbial biosynthetic pathway to produce naringenin is composed of tyrosine ammonia-lyase (TAL), 4-coumarate-CoA ligase (4CL), chalcone synthase (CHS), and chalcone isomerase (CHI). Herein, we targeted the efficient de novo production of naringenin in Escherichia coli by performing a step-by-step validation and optimization of the pathway. For that purpose, we first started by expressing two TAL genes from different sources in three different E. coli strains. The highest p-coumaric acid production (2.54 g/L) was obtained in the tyrosine-overproducing M-PAR-121 strain carrying TAL from Flavobacterium johnsoniae (FjTAL). Afterwards, this platform strain was used to express different combinations of 4CL and CHS genes from different sources. The highest naringenin chalcone production (560.2 mg/L) was achieved by expressing FjTAL combined with 4CL from Arabidopsis thaliana (At4CL) and CHS from Cucurbita maxima (CmCHS). Finally, different CHIs were tested and validated, and 765.9 mg/L of naringenin was produced by expressing CHI from Medicago sativa (MsCHI) combined with the other previously chosen genes. To our knowledge, this titer corresponds to the highest de novo production of naringenin reported so far in E. coli. KEY POINTS: • Best enzyme and strain combination were selected for de novo naringenin production. • After genetic and operational optimizations, 765.9 mg/L of naringenin was produced. • This de novo production is the highest reported so far in E. coli.


Asunto(s)
Aciltransferasas , Amoníaco-Liasas , Vías Biosintéticas , Coenzima A Ligasas , Escherichia coli , Flavanonas , Flavanonas/biosíntesis , Flavanonas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Vías Biosintéticas/genética , Aciltransferasas/genética , Aciltransferasas/metabolismo , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Amoníaco-Liasas/genética , Amoníaco-Liasas/metabolismo , Ingeniería Metabólica/métodos , Ácidos Cumáricos/metabolismo , Liasas Intramoleculares/genética , Liasas Intramoleculares/metabolismo , Tirosina/metabolismo
9.
Genes (Basel) ; 15(8)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39202344

RESUMEN

Long-chain acyl-CoA synthetases (LACSs) are essential enzymes that activate free fatty acids to fatty acyl-CoA thioesters, playing key roles in fatty acid (FA) catabolism, lipid synthesis and storage, epidermal wax synthesis, and stress tolerance. Despite their importance, comprehensive information about LACS genes in maize, a primary food crop, remains scarce. In the present work, eleven maize LACS genes were identified and mapped across five chromosomes. Three pairs of segmentally duplicated genes were detected in the maize LACS gene family, which underwent significant purifying selection (Ka/Ks < 1). Subsequently, phylogenetic analysis indicated that ZmLACS genes were divided into four subclasses, as supported by highly conserved motifs and gene structures. On the basis of the PlantCARE database, analysis of the ZmLACS promoter regions revealed various cis-regulatory elements related to tissue-specific expression, hormonal regulation, and abiotic stress response. RT-qPCR analysis showed that ZmLACS genes exhibit tissue-specific expression patterns and respond to diverse abiotic stresses including drought and salt, as well as phytohormone abscisic acid. Furthermore, using the STRING database, several proteins involved in fatty acid and complex lipid synthesis were identified to be the potential interaction partners of ZmLACS proteins, which was also confirmed by the yeast two-hybrid (Y2H) assay, enhancing our understanding of wax biosynthesis and regulatory mechanisms in response to abiotic stresses in maize. These findings provide a comprehensive understanding of ZmLACS genes and offer a theoretical foundation for future research on the biological functions of LACS genes in maize environmental adaptability.


Asunto(s)
Coenzima A Ligasas , Regulación de la Expresión Génica de las Plantas , Filogenia , Proteínas de Plantas , Estrés Fisiológico , Zea mays , Zea mays/genética , Zea mays/metabolismo , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Estrés Fisiológico/genética , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Genoma de Planta , Cromosomas de las Plantas/genética , Sequías
10.
Cell Biochem Funct ; 42(7): e4114, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39210707

RESUMEN

Long-chain acyl-CoA synthetases (ACSLs) are pivotal enzymes in fatty acid metabolism, essential for maintaining cellular homeostasis and energy production. Recent research has uncovered their significant involvement in the pathophysiology of various kidney diseases, including acute kidney injury (AKI), chronic kidney disease (CKD), diabetic kidney disease (DKD), and renal cell carcinoma (RCC). While ACSL1, ACSL3, ACSL4, and ACSL5 have been extensively studied for their roles in processes such as ferroptosis, lipid peroxidation, renal fibrosis, epithelial-mesenchymal transition, and tumor progression, the role of ACSL6 in kidney diseases remain largely unexplored. Notably, these isoenzymes exhibit distinct functions in different kidney diseases. Therefore, to provide a comprehensive understanding of their involvement, this review highlights the molecular pathways influenced by ACSLs and their roles in modulating cell death, inflammation, and fibrosis during kidney disease progression. By examining these mechanisms in detail, this review underscores the potential of ACSLs as biomarkers and therapeutic targets, advocating for further research to elucidate the precise roles of individual ACSL isoenzymes in kidney disease progression. Understanding these mechanisms opens new avenues for developing targeted interventions and improving therapeutic outcomes for patients with kidney diseases.


Asunto(s)
Coenzima A Ligasas , Isoenzimas , Enfermedades Renales , Humanos , Coenzima A Ligasas/metabolismo , Isoenzimas/metabolismo , Enfermedades Renales/metabolismo , Enfermedades Renales/enzimología , Enfermedades Renales/patología , Animales , Fibrosis/metabolismo
11.
Int J Biol Macromol ; 277(Pt 3): 134438, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39098676

RESUMEN

Long-chain acyl-CoA synthetases (LACSs), belonging to the acyl-activating enzyme superfamily, play crucial roles in lipid biosynthesis and fatty acid catabolism. Here, we identified 11 LACS genes in the tomato reference genome, and these genes were clustered into six subfamilies. Gene structure and conserved motif analyses indicated that LACSs from the same subfamily shared conserved gene and protein structures. Expression analysis revealed that SlLACS1 was highly expressed in the outer epidermis of tomato fruits and leaves. Subcellular localization assay results showed that SlLACS1 was located in the endoplasmic reticulum. Compared with wild-type plants, the wax content on leaves and fruits decreased by 22.5-34.2 % in SlLACS1 knockout lines, confirming that SlLACS1 was involved in wax biosynthesis in both leaves and fruits. Water loss, chlorophyll extraction, water-deficit, and toluidine blue assays suggested that cuticle permeability was elevated in SlLACS1 knockout lines, resulting in reduction in both drought stress resistance and fruit shelf-life. Overall, our analysis of the LACSs in tomato, coupled with investigations of SlLACS1 function, yielded a deeper understanding of the evolutionary patterns of LACS members and revealed the involvement of SlLACS1 in wax accumulation contribute to drought resistance and extended fruit shelf-life in tomato.


Asunto(s)
Coenzima A Ligasas , Regulación de la Expresión Génica de las Plantas , Familia de Multigenes , Filogenia , Solanum lycopersicum , Ceras , Solanum lycopersicum/genética , Solanum lycopersicum/metabolismo , Solanum lycopersicum/enzimología , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Ceras/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Proteínas de Plantas/química , Frutas/genética , Frutas/metabolismo , Hojas de la Planta/genética , Hojas de la Planta/metabolismo , Epidermis de la Planta/metabolismo , Epidermis de la Planta/genética
12.
Biochemistry ; 63(17): 2153-2165, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39152907

RESUMEN

Per and polyfluoroalkyl substances (PFAS) are a large family of anthropogenic fluorinated chemicals of increasing environmental concern. Over recent years, numerous microbial communities have been found to be capable of metabolizing some polyfluoroalkyl substances, generating a range of low-molecular-weight PFAS metabolites. One proposed pathway for the microbial breakdown of fluorinated carboxylates includes ß-oxidation, this pathway is initiated by the formation of a CoA adduct. However, until recently no PFAS-CoA adducts had been reported. In a previous study, we were able to use a bacterial medium-chain acyl-CoA synthetase (mACS) to form CoA adducts of fluorinated adducts of propanoic acid and pentanoic acid but were not able to detect any products of fluorinated hexanoic acid analogues. Herein, we expressed and purified a long-chain acyl-CoA synthetase (lACS) and a A461K variant of mACS from the soil bacterium Gordonia sp. strain NB4-1Y and performed an analysis of substrate scope and enzyme kinetics using fluorinated and nonfluorinated carboxylates. We determined that lACS can catalyze the formation of CoA adducts of 1:5 fluorotelomer carboxylic acid (FTCA), 2:4 FTCA and 3:3 FTCA, albeit with generally low turnover rates (<0.02 s-1) compared with the nonfluorinated hexanoic acid (5.39 s-1). In addition, the A461K variant was found to have an 8-fold increase in selectivity toward hexanoic acid compared with wild-type mACS, suggesting that Ala-461 has a mechanistic role in selectivity toward substrate chain length. This provides further evidence to validate the proposed activation step involving the formation of CoA adducts in the enzymatic breakdown of PFAS.


Asunto(s)
Caproatos , Coenzima A Ligasas , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Coenzima A Ligasas/química , Caproatos/metabolismo , Caproatos/química , Bacteria Gordonia/metabolismo , Bacteria Gordonia/enzimología , Bacteria Gordonia/genética , Halogenación , Coenzima A/metabolismo , Coenzima A/química , Cinética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Acilcoenzima A/metabolismo , Acilcoenzima A/química , Especificidad por Sustrato
13.
Redox Biol ; 75: 103301, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39116527

RESUMEN

The dysfunction of TAR DNA-binding protein 43 (TDP-43) is implicated in various neurodegenerative diseases, though the specific contributions of its toxic gain-of-function versus loss-of-function effects remain unclear. This study investigates the impact of TARDBP loss on cellular metabolism and viability using human-induced pluripotent stem cell-derived motor neurons and HeLa cells. TARDBP silencing led to reduced metabolic activity and cell growth, accompanied by neurite degeneration and decreased oxygen consumption rates in both cell types. Notably, TARDBP depletion induced a metabolic shift, impairing ATP production, increasing metabolic inflexibility, and elevating free radical production, indicating a critical role for TDP-43 in maintaining cellular bioenergetics. Furthermore, TARDBP loss triggered non-apoptotic cell death, increased ACSL4 expression, and reprogrammed lipid metabolism towards lipid droplet accumulation, while paradoxically enhancing resilience to ferroptosis inducers. Overall, our findings highlight those essential cellular traits such as ATP production, metabolic activity, oxygen consumption, and cell survival are highly dependent on TARDBP function.


Asunto(s)
Adenosina Trifosfato , Proteínas de Unión al ADN , Metabolismo Energético , Metabolismo de los Lípidos , Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Células HeLa , Adenosina Trifosfato/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Supervivencia Celular , Consumo de Oxígeno , Ferroptosis
14.
Int Immunopharmacol ; 140: 112800, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39096875

RESUMEN

Oltipraz (OPZ) is a synthetic dithiolethione and is considered a novel activator of nuclear factor E2-related factor 2 (Nrf2). Increasing evidence indicates that Nrf2 protects against cerebral ischemia/reperfusion (I/R) injury by antagonizing ferroptosis and lipid peroxidation. However, the protective effects of OPZ on cerebral I/R injury remain to be elucidated. We investigated the in vitro and in vivo neuroprotective effects of OPZ. Mice were subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) to construct an in vivo model and PC12 cells were exposed to oxygen and glucose deprivation/reoxygenation (OGD/R) to establish an in vitro model. OPZ administration reduced the infarct volume and brain water content, and alleviated the neurological deficit of MCAO/R mice. Moreover, OPZ ameliorated MCAO/R-induced oxidative stress by decreasing the levels of 4-HNE and MDA and increasing the activities of SOD and GSH. We also found that OPZ ameliorated MCAO/R-induced ferroptosis by increasing SLC7A11 and GPX4 protein expression and downregulating ACSL4 protein expression. Similarly, the in vitro results revealed that OGD/R-induced oxidative stress and ferroptosis. Finally, mechanistic analysis revealed that OPZ significantly upregulated the Nrf2 expression and Nrf2 knockout (Nrf2 KO) abolished the OPZ-mediated protective effects. Taken together, these findings demonstrate that OPZ ameliorates cerebral I/R injury by suppressing the oxidative stress and ferroptosis.


Asunto(s)
Ferroptosis , Infarto de la Arteria Cerebral Media , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2 , Fármacos Neuroprotectores , Estrés Oxidativo , Daño por Reperfusión , Tionas , Tiofenos , Animales , Ferroptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Tionas/farmacología , Tionas/uso terapéutico , Células PC12 , Factor 2 Relacionado con NF-E2/metabolismo , Ratones , Masculino , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Tiofenos/farmacología , Tiofenos/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Ratas , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Modelos Animales de Enfermedad , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/metabolismo , Pirazinas
15.
Biol Direct ; 19(1): 57, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39039611

RESUMEN

Laryngeal carcinoma (LC) is a common cancer of the respiratory tract. This study aims to investigate the role of RNA-binding motif protein 15 (RBM15) in the cisplatin (DDP) resistance of LC cells. LC-DDP-resistant cells were constructed. RBM15, lysine-specific demethylase 5B (KDM5B), lncRNA Fer-1 like family member 4 (FER1L4), lncRNA KCNQ1 overlapping transcript 1 (KCNQ1OT1), glutathione peroxidase 4 (GPX4), and Acyl-CoA synthetase long-chain family (ACSL4) was examined. Cell viability, IC50, and proliferation were assessed after RBM15 downregulation. The enrichment of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) and N6-methyladenosine (m6A) on KDM5B was analyzed. KDM5B mRNA stability was measured after actinomycin D treatment. A tumor xenograft assay was conducted to verify the role of RBM15 in LC. Results showed that RBM15 was upregulated in LC and its knockdown decreased IC50, cell viability, proliferation, glutathione, and upregulated iron ion content, ROS, malondialdehyde, ACSL4, and ferroptosis. Mechanistically, RBM15 improved KDM5B stability in an IGF2BP3-dependent manner, resulting in FER1L4 downregulation and GPX4 upregulation. KDM5B increased KCNQ1OT1 and inhibited ACSL4. KDM5B/KCNQ1OT1 overexpression or FER1L4 knockdown promoted DDP resistance in LC by inhibiting ferroptosis. In conclusion, RBM15 promoted KDM5B expression, and KDM5B upregulation inhibited ferroptosis and promoted DDP resistance in LC by downregulating FER1L4 and upregulating GPX4, as well as by upregulating KCNQ1OT1 and inhibiting ACSL4. Silencing RBM15 inhibited tumor growth in vivo.


Asunto(s)
Cisplatino , Resistencia a Antineoplásicos , Epigénesis Genética , Ferroptosis , Neoplasias Laríngeas , Proteínas de Unión al ARN , Ferroptosis/genética , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Línea Celular Tumoral , Ratones , Animales , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica , Proliferación Celular/efectos de los fármacos , Antineoplásicos/farmacología , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo
16.
FASEB J ; 38(13): e23788, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38963329

RESUMEN

Intermittent hypoxia (IH) is an independent risk factor for metabolic dysfunction-associated fatty liver disease (MAFLD). Copper deficiency can disrupt redox homeostasis, iron, and lipid metabolism. Here, we investigated whether hepatic copper deficiency plays a role in IH-associated MAFLD and explored the underlying mechanism(s). Male C57BL/6 mice were fed a western-type diet with adequate copper (CuA) or marginally deficient copper (CuD) and were exposed separately to room air (RA) or IH. Hepatic histology, plasma biomarkers, copper-iron status, and oxidative stress were assessed. An in vitro HepG2 cell lipotoxicity model and proteomic analysis were used to elucidate the specific targets involved. We observed that there were no differences in hepatic phenotypes between CuA-fed and CuD-fed mice under RA. However, in IH exposure, CuD-fed mice showed more pronounced hepatic steatosis, liver injury, and oxidative stress than CuA-fed mice. IH induced copper accumulation in the brain and heart and exacerbated hepatic copper deficiency and secondary iron deposition. In vitro, CuD-treated cells with IH exposure showed elevated levels of lipid accumulation, oxidative stress, and ferroptosis susceptibility. Proteomic analysis identified 360 upregulated and 359 downregulated differentially expressed proteins between CuA and CuD groups under IH; these proteins were mainly enriched in citrate cycle, oxidative phosphorylation, fatty acid metabolism, the peroxisome proliferator-activated receptor (PPAR)α pathway, and ferroptosis. In IH exposure, CuD significantly upregulated the ferroptosis-promoting factor arachidonyl-CoA synthetase long chain family member (ACSL)4. ACSL4 knockdown markedly eliminated CuD-induced ferroptosis and lipid accumulation in IH exposure. In conculsion, IH can lead to reduced hepatic copper reserves and secondary iron deposition, thereby inducing ferroptosis and subsequent MAFLD progression. Insufficient dietary copper may worsen IH-associated MAFLD.


Asunto(s)
Cobre , Ferroptosis , Hipoxia , Ratones Endogámicos C57BL , Animales , Cobre/metabolismo , Cobre/deficiencia , Masculino , Ratones , Hipoxia/metabolismo , Humanos , Células Hep G2 , Hígado/metabolismo , Hígado/patología , Estrés Oxidativo , Metabolismo de los Lípidos , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/etiología , Hierro/metabolismo , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , PPAR alfa/metabolismo , PPAR alfa/genética
17.
Cell Death Dis ; 15(7): 545, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085201

RESUMEN

Chemotherapeutic efficacy is seriously impeded by chemoresistance in more than half of hepatocellular carcinoma (HCC) patients. However, the mechanisms involved in chemotherapy-induced upregulation of chemoresistant genes are not fully understood. Here, this study unravels a novel mechanism controlling nuclear acetyl-CoA production to activate the transcription of chemoresistant genes in HCC. NAT10 is upregulated in HCC tissues and its upregulation is correlated with poor prognosis of HCC patients. NAT10 is also upregulated in chemoresistant HCC cells. Targeting NAT10 increases the cytotoxicity of chemotherapy in HCC cells and mouse xenografts. Upon chemotherapy, NAT10 translocates from the nucleolus to the nucleus to activate the transcription of CYP2C9 and PIK3R1. Additionally, nuclear acetyl-CoA is specifically upregulated by NAT10. Mechanistically, NAT10 binds with ACLY in the nucleus and acetylates ACLY at K468 to counteract the SQSTM1-mediated degradation upon chemotherapy. ACLY K468-Ac specifically accumulates in the nucleus and increases nuclear acetyl-CoA production to activate the transcription of CYP2C9 and PIK3R1 through enhancing H3K27ac. Importantly, K468 is required for nuclear localization of ACLY. Significantly, ACLY K468-Ac is upregulated in HCC tissues, and ablation of ACLY K468-Ac sensitizes HCC cells and mouse xenografts to chemotherapy. Collectively, these findings identify NAT10 as a novel chemoresistant driver and the blockage of NAT10-mediated ACLY K468-Ac possesses the potential to attenuate HCC chemoresistance.


Asunto(s)
Acetilcoenzima A , Carcinoma Hepatocelular , Núcleo Celular , Resistencia a Antineoplásicos , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Acetilcoenzima A/metabolismo , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Animales , Acetilación , Ratones , Núcleo Celular/metabolismo , Línea Celular Tumoral , Ratones Desnudos , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Acetiltransferasas N-Terminal/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ratones Endogámicos BALB C , Masculino
18.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167354, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39004378

RESUMEN

Acute lung injury (ALI) is a serious disorder characterized by the release of pro-inflammatory cytokines and cascade activation of macrophages. Ferroptosis, a form of iron-dependent cell death triggered by intracellular phospholipid peroxidation, has been implicated as an internal mechanism underlying ALI. In this study, we investigated the effects of m6A demethylase fat mass and obesity-associated protein (FTO) on the inhibition of macrophage ferroptosis in ALI. Using a mouse model of lipopolysaccharide (LPS)-induced ALI, we observed the induction of ferroptosis and its co-localization with the macrophage marker F4/80, suggesting that ferroptosis might be induced in macrophages. Ferroptosis was promoted during LPS-induced inflammation in macrophages in vitro, and the inflammation was counteracted by the ferroptosis inhibitor ferrostatin-1 (fer-1). Given that FTO showed lower expression levels in the lung tissue of mice with ALI and inflammatory macrophages, we further dissected the regulatory capacity of FTO in ferroptosis. The results demonstrated that FTO alleviated macrophage inflammation by inhibiting ferroptosis. Mechanistically, FTO decreased the stability of ACSL4 mRNA via YTHDF1, subsequently inhibiting ferroptosis and inflammation by interrupting polyunsaturated fatty acid consumption. Moreover, FTO downregulated the synthesis and secretion of prostaglandin E2, thereby reducing ferroptosis and inflammation. In vivo, the FTO inhibitor FB23-2 aggravated lung injury, the inflammatory response, and ferroptosis in mice with ALI; however, fer-1 therapy mitigated these effects. Overall, our findings revealed that FTO may function as an inhibitor of the inflammatory response driven by ferroptosis, emphasizing its potential as a target for ALI treatment.


Asunto(s)
Lesión Pulmonar Aguda , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Coenzima A Ligasas , Ferroptosis , Inflamación , Macrófagos , Animales , Masculino , Ratones , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Ciclohexilaminas , Modelos Animales de Enfermedad , Ferroptosis/efectos de los fármacos , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Lipopolisacáridos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones Endogámicos C57BL , Fenilendiaminas/farmacología , Células RAW 264.7
19.
Sci Rep ; 14(1): 15968, 2024 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987531

RESUMEN

To analyze the mechanism of how interfering with the cytokeratin 19 (CK19) pathway via the ferroptosis pathway affects tumor biological behaviors in the process of oral squamous cell carcinoma (OSCC) development. TCGA was used to analyze the expression of CK19 in pan-cancer and head and neck squamous cell carcinoma (HNSC) and to explore the ferroptosis-related genes related to HNSC. The effect of silencing CK19 on the migration ability of HSC-4 cells was verified by wound healing and migration assay. HSC-4 cells with silencing of CK19 and tumor-bearing nude mouse model were constructed. RT-qPCR, immunofluorescence and western blot were used to analyze the expression of ferroptosis-related genes. CK19 is highly expressed in human OSCC and nude mice. The migration ability of cells in the CK19-silenced group was lower than that of the control group. In vivo and in vitro, CK19 was negatively correlated with the expression of ACSL4 and positively correlated with the expression of GPX4. Compared with the control group, GPX4 expression was down-regulated and ACSL4 expression was up-regulated in the CK19-silenced group. Silencing CK19 also increased intracellular Fe2+ content and MDA content. Silencing CK19 can affect the expression of GPX4 and ACSL4 to regulate ferroptosis and at the same time increase the content of MDA, Fe2+ and ROS levels, thereby activating the regulation of ferroptosis pathway in the development of OSCC.


Asunto(s)
Coenzima A Ligasas , Ferroptosis , Regulación Neoplásica de la Expresión Génica , Queratina-19 , Ratones Desnudos , Neoplasias de la Boca , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Animales , Humanos , Ratones , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Ferroptosis/genética , Silenciador del Gen , Queratina-19/metabolismo , Queratina-19/genética , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética
20.
Cancer Biol Med ; 21(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38953696

RESUMEN

OBJECTIVE: Mitochondrial fatty acid oxidation is a metabolic pathway whose dysregulation is recognized as a critical factor in various cancers, because it sustains cancer cell survival, proliferation, and metastasis. The acyl-CoA synthetase long-chain (ACSL) family is known to activate long-chain fatty acids, yet the specific role of ACSL3 in breast cancer has not been determined. METHODS: We assessed the prognostic value of ACSL3 in breast cancer by using data from tumor samples. Gain-of-function and loss-of-function assays were also conducted to determine the roles and downstream regulatory mechanisms of ACSL3 in vitro and in vivo. RESULTS: ACSL3 expression was notably downregulated in breast cancer tissues compared with normal tissues, and this phenotype correlated with improved survival outcomes. Functional experiments revealed that ACSL3 knockdown in breast cancer cells promoted cell proliferation, migration, and epithelial-mesenchymal transition. Mechanistically, ACSL3 was found to inhibit ß-oxidation and the formation of associated byproducts, thereby suppressing malignant behavior in breast cancer. Importantly, ACSL3 was found to interact with YES proto-oncogene 1, a member of the Src family of tyrosine kinases, and to suppress its activation through phosphorylation at Tyr419. The decrease in activated YES1 consequently inhibited YAP1 nuclear colocalization and transcriptional complex formation, and the expression of its downstream genes in breast cancer cell nuclei. CONCLUSIONS: ACSL3 suppresses breast cancer progression by impeding lipid metabolism reprogramming, and inhibiting malignant behaviors through phospho-YES1 mediated inhibition of YAP1 and its downstream pathways. These findings suggest that ACSL3 may serve as a potential biomarker and target for comprehensive therapeutic strategies for breast cancer.


Asunto(s)
Neoplasias de la Mama , Proliferación Celular , Coenzima A Ligasas , Progresión de la Enfermedad , Metabolismo de los Lípidos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-yes , Factores de Transcripción , Proteínas Señalizadoras YAP , Humanos , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Femenino , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Proteínas Señalizadoras YAP/metabolismo , Animales , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Proto-Oncogénicas c-yes/metabolismo , Proteínas Proto-Oncogénicas c-yes/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Transición Epitelial-Mesenquimal , Ratones Desnudos , Pronóstico , Movimiento Celular , Transducción de Señal , Reprogramación Metabólica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA