Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
J Biol Chem ; 300(1): 105502, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38016515

RESUMEN

Fatty acid handling and complex lipid synthesis are altered in the kidney cortex of diabetic patients. We recently showed that inhibition of the renin-angiotensin system without changes in glycemia can reverse diabetic kidney disease (DKD) and restore the lipid metabolic network in the kidney cortex of diabetic (db/db) mice, raising the possibility that lipid remodeling may play a central role in DKD. However, the roles of specific enzymes involved in lipid remodeling in DKD have not been elucidated. In the present study, we used this diabetic mouse model and a proximal tubule epithelial cell line (HK2) to investigate the potential relationship between long-chain acyl-CoA synthetase 1 (ACSL1) and lipid metabolism in response to fatty acid exposure and inflammatory signals. We found ACSL1 expression was significantly increased in the kidney cortex of db/db mice, and exposure to palmitate or tumor necrosis factor-α significantly increased Acsl1 mRNA expression in HK-2 cells. In addition, palmitate treatment significantly increased the levels of long-chain acylcarnitines and fatty acyl CoAs in HK2 cells, and these increases were abolished in HK2 cell lines with specific deletion of Acsl1(Acsl1KO cells), suggesting a key role for ACSL1 in fatty acid ß-oxidation. In contrast, tumor necrosis factor-α treatment significantly increased the levels of short-chain acylcarnitines and long-chain fatty acyl CoAs in HK2 cells but not in Acsl1KO cells, consistent with fatty acid channeling to complex lipids. Taken together, our data demonstrate a key role for ACSL1 in regulating lipid metabolism, fatty acid partitioning, and inflammation.


Asunto(s)
Coenzima A Ligasas , Nefropatías Diabéticas , Ácidos Grasos , Animales , Humanos , Ratones , Coenzima A Ligasas/metabolismo , Diabetes Mellitus/patología , Nefropatías Diabéticas/metabolismo , Ácidos Grasos/metabolismo , Ligasas , Palmitatos , Factor de Necrosis Tumoral alfa
2.
Mol Carcinog ; 62(8): 1119-1135, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37144835

RESUMEN

Acute myeloid leukemia (AML) is a hematological malignancy with an alarming mortality rate. The development of novel therapeutic targets or drugs for AML is urgently needed. Ferroptosis is a form of regulated cell death driven by iron-dependent lipid peroxidation. Recently, ferroptosis has emerged as a novel method for targeting cancer, including AML. Epigenetic dysregulation is a hallmark of AML, and a growing body of evidence suggests that ferroptosis is subject to epigenetic regulation. Here, we identified protein arginine methyltransferase 1 (PRMT1) as a ferroptosis regulator in AML. The type I PRMT inhibitor GSK3368715 promoted ferroptosis sensitivity in vitro and in vivo. Moreover, PRMT1-knockout cells exhibited significantly increased sensitivity to ferroptosis, suggesting that PRMT1 is the primary target of GSK3368715 in AML. Mechanistically, both GSK3368715 and PRMT1 knockout upregulated acyl-CoA synthetase long-chain family member 1 (ACSL1), which acts as a ferroptosis promoter by increasing lipid peroxidation. Knockout ACSL1 reduced the ferroptosis sensitivity of AML cells following GSK3368715 treatment. Additionally, the GSK3368715 treatment reduced the abundance of H4R3me2a, the main histone methylation modification mediated by PRMT1, in both genome-wide and ACSL1 promoter regions. Overall, our results demonstrated a previously unknown role of the PRMT1/ACSL1 axis in ferroptosis and suggested the potential value and applications of the combination of PRMT1 inhibitor and ferroptosis inducers in AML treatment.


Asunto(s)
Ferroptosis , Leucemia Mieloide Aguda , Humanos , Ferroptosis/genética , Regulación hacia Arriba , Epigénesis Genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Inhibidores Enzimáticos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas Represoras/metabolismo , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo
3.
Med Mycol ; 61(8)2023 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-37573133

RESUMEN

Vulvovaginal candidiasis (VVC) is an inflammatory disease primarily infected by Candida albicans. The condition has good short-term treatment effects, high recurrence, and seriously affects the quality of life of women. Metabolomics has been applied to research a variety of inflammatory diseases. In the present study, the vaginal metabolic profiles of VVC patients and healthy populations (Cnotrol (CTL)) were explored by a non-targeted metabolomics approach. In total, 211 differential metabolites were identified, with the VVC group having 128 over-expressed and 83 under-expressed metabolites compared with healthy individuals. Functional analysis showed that these metabolites were mainly involved in amino acid metabolism and lipid metabolism. In addition, network software analysis indicated that the differential metabolites were associated with mitogen-activated protein kinase (MAPK) signaling and NF-κB signaling. Further molecular docking suggested that linoleic acid can bind to the acyl-CoA synthetase 1 (ACSL1) protein, which has been shown to be associated with multiple inflammatory diseases and is an upstream regulator of the MAPK and NF-κB signaling pathways that mediate inflammation. Therefore, our preliminary analysis results suggest that VVC has a unique metabolic profile. Linoleic acid, a significantly elevated unsaturated fatty acid in the VVC group, may promote VVC development through the ACSL1/MAPK and ACSL1/NF-κB signaling pathways. This study's findings contribute to further exploring the mechanism of VVC infection and providing new perspectives for the treatment of Candida albicans vaginal infection.


Candida albicans is the main pathogen that causes VVC. Through non-targeted metabolomics, this study shows that VVC caused by C. albicans has unique vaginal metabolic characteristics, the changed metabolites might provide useful diagnostic and therapeutic methods for VVC.


Asunto(s)
Candidiasis Vulvovaginal , Candidiasis , Femenino , Animales , Candidiasis Vulvovaginal/tratamiento farmacológico , Candidiasis Vulvovaginal/veterinaria , FN-kappa B , Ácido Linoleico , Simulación del Acoplamiento Molecular , Calidad de Vida , Candida albicans , Candidiasis/tratamiento farmacológico , Candidiasis/veterinaria , Metabolómica , Homeostasis , Antifúngicos/farmacología , Antifúngicos/uso terapéutico
4.
Biochem Biophys Res Commun ; 620: 35-41, 2022 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-35777132

RESUMEN

BACKGROUND: Circular RNAs (circRNAs) have been proved could regulate many cancers, including prostate cancer (PCa). In this paper, we reconnoitered the roles of circRNA pyruvate dehydrogenase complex component X (circPDHX) in PCa. METHODS: The circPDHX, microRNA (miR)-497-5p and acyl-CoA synthetase long chain family member 1 (ACSL1) contents were detected by quantitative real-time PCR and Western blot analysis. Cell proliferation was measured by cell counting kit-8 assay, 5-Ethynyl-2'-deoxyuridine assay, and colony formation assay. Cell migration was examined by wound healing assay. The apoptosis was detected by flow cytometry assay. The ELISA kits were applied to quantify the fatty acid metabolites. Furthermore, the interplay between miR-497-5p and circPDHX or ACSL1 was detected by dual-luciferase reporter assay and RIP assay. The role of circPDHX in PCa was supplementary substantiated in vivo. RESULTS: CircPDHX and ACSL1 contents were upregulated, and the miR-497-5p level was downregulated in PCa. CircPDHX deficiency attenuated PCa cell proliferation, migration, and fatty acid metabolites, while intensified cell apoptosis. CircPDHX bound to miR-497-5p to adjust ACSL1. Moreover, miR-497-5p inhibited the PCa progression by regulating ACSL1. In the meantime, circPDHX deficiency repressed PCa tumor growth in vivo. CONCLUSION: CircPDHX stimulated PCa development via miR-497-5p/ACSL1, which presented a new thought for PCa treatment.


Asunto(s)
MicroARNs , Neoplasias de la Próstata , Proliferación Celular/genética , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Ácidos Grasos , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , MicroARNs/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Circular/genética
5.
Genomics ; 113(6): 3967-3977, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34601049

RESUMEN

Long-chain acyl-CoA synthetase 1 (ACSL1) is a member of the acyl-CoA synthetase family that plays a vital role in lipid metabolism. We have previously shown that the ACSL1 gene regulates the composition of unsaturated fatty acids (UFAs) in bovine skeletal muscle, which in turn regulates the fatty acid synthesis and the generation of lipid droplets. Here, we used RNA-Seq to screen circRNAs that regulated the expression of ACSL1 gene and other UFA synthesis-related genes by RNA interference and noninterference in bovine adipocytes. The results of KEGG pathway analysis showed that the parental genes of differentially expressed (DE)-circRNAs were primarily enriched in the adipocytokine signaling pathway. The prediction results showed that novel_circ_0004855, novel_circ_0001507, novel_circ_0001731, novel_circ_0005276, novel_circ_0002060, novel_circ_0005405 and novel_circ_0004254 regulated UFA synthesis-related genes by interacting with the related miRNAs. These results could help expand our knowledge of the molecular mechanisms of circRNAs in the regulation of UFA synthesis in bovine adipocytes.


Asunto(s)
MicroARNs , ARN Circular , Adipocitos/metabolismo , Animales , Bovinos , Ácidos Grasos Insaturados/genética , Ácidos Grasos Insaturados/metabolismo , Perfilación de la Expresión Génica , Metabolismo de los Lípidos , MicroARNs/genética , MicroARNs/metabolismo , Transcriptoma
6.
Bioorg Med Chem Lett ; 33: 127722, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33285268

RESUMEN

Long-chain acyl-CoA synthetase-1 (ACSL1), an enzyme that catalyzes the synthesis of long-chain acyl-CoA from the corresponding fatty acids, is believed to play essential roles in lipid metabolism. Structure activity relationship studies based on HTS hit compound 1 delivered the benzimidazole series as the first selective and highly potent ACSL1 inhibitors. Representative compound 13 exhibited not only remarkable inhibitory activity against ACSL1 (IC50 = 0.042 µM) but also excellent selectivity for the other ACSL isoforms. In addition, compound 13 demonstrated an in vivo suppression effect against the production of long-chain acyl-CoAs in mouse.


Asunto(s)
Bencimidazoles/farmacología , Coenzima A Ligasas/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Animales , Bencimidazoles/síntesis química , Bencimidazoles/química , Coenzima A Ligasas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Ratones , Ratones Noqueados , Estructura Molecular , Relación Estructura-Actividad
7.
Int J Med Sci ; 18(3): 615-625, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33437196

RESUMEN

Normally, there are multiple microRNAs involved in the pathogenesis of liver fibrosis. In our work, we aimed at identifying the role of miR-34c in the hepatic stellate cell (HSC) activation and liver fibrosis and its potential mechanism. Our results have shown that during natural activation of HSC, the level of miR-34c was increased significantly whereas acyl-CoA synthetase long-chain family member-1(ACSL1), which is a key enzyme can affect fatty acid(FA) synthesis, was decreased. A double fluorescence reporter assay further confirmed that ACSL1 is a direct target gene of miR-34c. Moreover, the inhibition of miR-34C can attenuate the synthesis of collagen in HSC-T6. In our rescue assay, ACSL1 expression was 1.49-fold higher compared to normal control cells which were transfected with the miR-34c inhibitor in a stable low expression ACSL1 cell line. While at the same time, α-SMA and Col1α expression decreased by 18.22% and 2.58%, respectively. Moreover, we performed an in vivo model using dimethylnitrosamine (DMN) in conjunction with the miR-34c agomir, combined with the treatment of DMN and the miR-34c agomir can increase liver fibrosis. Meanwhile, the degree of hepatic fibrosis was increased and lipid droplets reduced dramatically in rats and HSC-T6 cell treated with miR-34c mimics alone compared to untreated groups. Our results indicate that miR-34c plays an essential role in liver fibrosis by targeting ACSL1 closely associated with lipid droplets, and it might be used as a potential therapeutic target.


Asunto(s)
Coenzima A Ligasas/genética , Células Estrelladas Hepáticas/patología , Cirrosis Hepática Experimental/genética , Hígado/patología , MicroARNs/metabolismo , Animales , Coenzima A Ligasas/metabolismo , Colágeno/biosíntesis , Dimetilnitrosamina/administración & dosificación , Dimetilnitrosamina/toxicidad , Células Estrelladas Hepáticas/efectos de los fármacos , Humanos , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/genética , Hígado/citología , Hígado/efectos de los fármacos , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/patología , MicroARNs/agonistas , MicroARNs/antagonistas & inhibidores , Ratas
8.
J Biol Chem ; 294(22): 8819-8833, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-30975900

RESUMEN

Loss of long-chain acyl-CoA synthetase isoform-1 (ACSL1) in mouse skeletal muscle (Acsl1M-/-) severely reduces acyl-CoA synthetase activity and fatty acid oxidation. However, the effects of decreased fatty acid oxidation on skeletal muscle function, histology, use of alternative fuels, and mitochondrial function and morphology are unclear. We observed that Acsl1M-/- mice have impaired voluntary running capacity and muscle grip strength and that their gastrocnemius muscle contains myocytes with central nuclei, indicating muscle regeneration. We also found that plasma creatine kinase and aspartate aminotransferase levels in Acsl1M-/- mice are 3.4- and 1.5-fold greater, respectively, than in control mice (Acsl1flox/flox ), indicating muscle damage, even without exercise, in the Acsl1M-/- mice. Moreover, caspase-3 protein expression exclusively in Acsl1M-/- skeletal muscle and the presence of cleaved caspase-3 suggested myocyte apoptosis. Mitochondria in Acsl1M-/- skeletal muscle were swollen with abnormal cristae, and mitochondrial biogenesis was increased. Glucose uptake did not increase in Acsl1M-/- skeletal muscle, and pyruvate oxidation was similar in gastrocnemius homogenates from Acsl1M-/- and control mice. The rate of protein synthesis in Acsl1M-/- glycolytic muscle was 2.1-fold greater 30 min after exercise than in the controls, suggesting resynthesis of proteins catabolized for fuel during the exercise. At this time, mTOR complex 1 was activated, and autophagy was blocked. These results suggest that fatty acid oxidation is critical for normal skeletal muscle homeostasis during both rest and exercise. We conclude that ACSL1 deficiency produces an overall defect in muscle fuel metabolism that increases protein catabolism, resulting in exercise intolerance, muscle weakness, and myocyte apoptosis.


Asunto(s)
Aminoácidos/metabolismo , Coenzima A Ligasas/genética , Ácidos Grasos/metabolismo , Músculo Esquelético/metabolismo , Animales , Apoptosis , Aspartato Aminotransferasas/metabolismo , Caspasa 3/metabolismo , Coenzima A Ligasas/deficiencia , Creatina Quinasa/metabolismo , Metabolismo de los Lípidos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Músculo Esquelético/patología , Oxidación-Reducción , Condicionamiento Físico Animal , Regulación hacia Arriba
9.
J Cell Biochem ; 121(10): 4034-4042, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31961004

RESUMEN

Thyroid cancer (TC) has been characterized as the most common malignant malady of the endocrine system. Small nucleolar RNA host gene 7 (SNHG7) has been reported to serve as a key regulator in a large number of human cancer types, but its role in TC and the underlying regulatory mechanism have never been evaluated yet. The present study indicated that the expression of SNHG7 was markedly higher in TC cell lines. Knockdown of SNHG7 led to a suppression of TC cell progression and migration. Acyl-CoA synthetase long-chain family member 1 (ACSL1) has also been demonstrated as an oncogene in many cancers. Herein an inhibition of ACSL1 after SNHG7 knockdown was captured. Further, the suppressing effects of SNHG7 knockdown on TC cell processes were counteracted by ACSL1 overexpression. Data from online bioinformatics analysis, RNA immunoprecipitation, and luciferase reporter assays validated the interaction between microRNA-449a (miR-449a) and SNHG7 or ACSL1. It was also verified that SNHG7 sequestered miR-449a and therefore elevated ACSL1 expression levels. To conclude, the current study indicated that SNHG7 promoted proliferation and migration of TC cells by sponging miR-449a and therefore upregulating ACSL1. The present study may provide more explorations about the molecular regulation mechanism of long noncoding RNAs in TC progression.


Asunto(s)
Coenzima A Ligasas/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal/genética , Neoplasias de la Tiroides/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Coenzima A Ligasas/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , MicroARNs/genética , Oncogenes , ARN Largo no Codificante/genética , Neoplasias de la Tiroides/patología , Transfección , Regulación hacia Arriba/genética
10.
J Hepatol ; 73(3): 603-615, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32593682

RESUMEN

BACKGROUND & AIMS: Obesity is an independent risk factor for malignancies, including hepatocellular carcinoma (HCC). However, it remains unknown whether maternal obesity affects the incidence of HCC in offspring. Thus, we aimed to investigate this association and its underlying mechanisms. METHODS: Diethylnitrosamine (DEN) was used to induce HCC in a high-fat diet (HFD)-induced multigenerational obesity model. RNA-sequencing was performed to identify the genes and microRNAs (miRNAs) that were altered over generations. The role of the miR-27a-3p-Acsl1/Aldh2 axis in HCC was evaluated in cell lines and HCC-bearing nude mice, and its intergenerational impact was studied in pregnant mice and their offspring. RESULTS: Under HFD stress, maternal obesity caused susceptibility of offspring to DEN-induced HCC, and such susceptibility was cumulative over generations. We identified that Acsl1 and Aldh2, direct targets of miR-27a-3p, were gradually changed over generations. Under hyperlipidemic conditions, downregulation of Acsl1 and Aldh2 increased cell proliferation (in vitro) or tumor growth (in vivo) in synergy. Intratumor injection of an miR-27a-3p agomir exacerbated tumor growth by downregulating Acsl1 and Aldh2; while intratumor injection of an miR-27a-3p antagomir had the opposite effect. Moreover, serum miR-27a-3p levels gradually increased in the HFD-fed maternal lineage over generations. Injecting pregnant mice with an miR-27a-3p agomir not only upregulated hepatic miR-27a-3p and downregulated Acsl1/Aldh2 in offspring (fetus, young and adult stages), but also exacerbated HCC development in DEN-treated offspring. In human HCC, upregulated miR-27a-3p and downregulated Acsl1/Aldh2 were negatively correlated with survival on TCGA analysis; while, hepatic miR-27a-3p was negatively correlated with Acsl1/Aldh2 expression in tumor/non-tumor tissues from fatty/non-fatty livers. CONCLUSIONS: Maternal obesity plays a role in regulating cumulative susceptibility to HCC development in offspring over multiple generations through the miR-27a-3p-Acsl1/Aldh2 axis. LAY SUMMARY: It is not currently known how maternal obesity affects the incidence of liver cancer in offspring. In this study, we identified a microRNA (miR-27a-3p) that was upregulated in obese mothers and could be passed on to their offspring. This microRNA enhanced the risk of liver cancer in offspring by regulating 2 genes (Acsl1 and Aldh2). This mechanism could be a future therapeutic target.


Asunto(s)
Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/epidemiología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/epidemiología , MicroARNs/metabolismo , Obesidad Materna/complicaciones , Obesidad Materna/metabolismo , Aldehído Deshidrogenasa Mitocondrial/genética , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Animales , Carcinoma Hepatocelular/patología , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Dieta Alta en Grasa/efectos adversos , Dietilnitrosamina/efectos adversos , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Incidencia , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , MicroARNs/genética , Obesidad Materna/etiología , Embarazo , Transfección , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Arch Biochem Biophys ; 695: 108648, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33098867

RESUMEN

There exists a positive correlation between the unsaturated fatty acids (UFA) content in the bovine species and their taste and nutritional significance. Long-chain acyl-CoA synthetase 1 (ACSL1) is known to be involved in lipid synthesis as well as fatty acid transport and degradation. This gene has been identified as the key candidate gene for regulating lipid composition in the bovine skeletal muscle; however, its mechanism of action in regulating UFA synthesis in bovine adipocytes is unclear. In this study, we used a recombinant adenovirus vector (Ad-ACSL1) to overexpress the ACSL1 gene using Ad-NC (recombinant adenovirus of green fluorescent protein) as the control. Quantitative real-time PCR (qRT-PCR) was done to examine the gene expression associated with the synthesis of UFA, followed by the analysis of the fatty acid composition. Oil red O staining was done to examine the aggregation of lipid droplets. We found that ACSL1 overexpression was associated with an upregulated expression of PPARγ, FABP3, ACLY, SCD1, and FASN, and downregulated expression of CPT1A. Additionally, ACSL1 overexpression resulted in elevated saturated fatty acid content, especially C16:0 and C18:0, than the control group (Ad-NC cells) (p < 0.05). Furthermore, the overexpression of ACSL1 enhanced the proportion of eicosapentaenoic acid (EPA), decreased the proportion of C22:4, and significantly upregulated polyunsaturated fatty acid (PUFA) content. These results were supported by oil red O staining, which revealed an increase in the lipid droplets in bovine adipocytes after the overexpression of the ACSL1 gene. Thus, the results of this study indicated that ACSL1 positively regulated PUFA synthesis in bovine adipocytes.


Asunto(s)
Adipocitos/metabolismo , Coenzima A Ligasas/biosíntesis , Ácidos Grasos Insaturados/biosíntesis , Regulación Enzimológica de la Expresión Génica , Adenoviridae , Animales , Bovinos , Coenzima A Ligasas/genética , Ácidos Grasos Insaturados/genética , Vectores Genéticos , Transducción Genética
12.
Int J Med Sci ; 17(6): 720-727, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32218693

RESUMEN

In clinical cohort studies, high expression of long-chain acyl-coenzyme A synthetases 1 (ACSL1 gene) in peripheral white blood cells of patients with acute myocardial infarction (AMI) has been utilized as molecular markers of myocardial infarction diagnosis. The plasma triglyceride level of AMI patients is significantly higher than that of healthy individuals. We hypothesized that the high expression of ACSL1 increases the level of triglyceride, which is one of the pathogenesis of AMI promoted by ACSL1. In this report, cell culture based methods were adopted to test the hypothesis and further investigate the effect and mechanism of ACSL1 on lipid metabolism. In this study, liver cells of healthy individuals were cultured, the overexpression and the knockdown vectors of ACSL1 were constructed and transfected into liver cells. The transfection was verified at the mRNA and protein level. Intracellular triglyceride content was quantitatively analyzed using ELISA. Changes of genes related to lipid metabolism were subsequently measured through PCR array. Overexpression of ACSL1 led to higher gene expression and protein levels compared to control and the triglyceride content was significantly increased in overexpressing cells. The expression level of fatty acid oxidation pathway PPARγ was significantly down-regulated compared with the control group, as were genes associated with fatty acid synthesis pathways: SREBP1, ACC, FAS, and SCD1. ACSL1 knockdown decreased the content of triglyceride whereas PPARγ was up-regulated and SREBP1, ACC, FAS, and SCD1 were down-regulated compared with the control group. In summary, high expression of ACSL1 reduced fatty acid ß-oxidation through the PPARγ pathway, thereby increasing triglyceride levels.


Asunto(s)
Coenzima A Ligasas/sangre , Infarto del Miocardio/sangre , PPAR gamma/sangre , Triglicéridos/sangre , Acetil-CoA Carboxilasa/genética , Biomarcadores/sangre , Coenzima A Ligasas/genética , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Humanos , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/patología , PPAR gamma/genética , Cultivo Primario de Células , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Transfección
13.
Int J Mol Sci ; 21(6)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32192050

RESUMEN

Long-chain fatty acyl-CoA synthetase (ACSLs) is an essential enzyme for the synthesis of fatty acyl-CoA. ACSL1 plays a key role in the synthesis of triglycerides, phospholipids, and cholesterol esters. BACKGROUND: In the current study, triglyceride content did not increase after overexpression of the ACSL1 gene. METHODS: RNA-seq and lipid metabolome profiling were performed to determine why triglyceride levels did not change with ACSL1 overexpression. RESULTS: Fatty acyl-CoA produced by ACSL1 was determined to be involved in the diglyceride synthesis pathway, and diglyceride content significantly increased when ACSL1 was overexpressed. Moreover, the arachidonic acid (AA) content in sheep adipocytes significantly increased, and the level of cyclooxygenase 2 (COX2) expression, the downstream metabolic gene, was significantly downregulated. Knocking down the ACSL1 gene was associated with an increase in COX2 mRNA expression, as well as an increase in prostaglandin content, which is the downstream metabolite of AA. CONCLUSIONS: The overexpression of the ACSL1 gene promotes the production of AA via downregulation of COX2 gene expression.


Asunto(s)
Adipocitos/metabolismo , Ácido Araquidónico/metabolismo , Coenzima A Ligasas/metabolismo , Diglicéridos/biosíntesis , Ácidos Grasos/metabolismo , Metabolismo de los Lípidos , Animales , Coenzima A Ligasas/genética , Ciclooxigenasa 2/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Redes y Vías Metabólicas , Plásmidos/genética , Análisis de Secuencia de ARN , Ovinos
14.
Molecules ; 25(20)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066575

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a monomeric glycoprotein that has been implicated in the tumor growth and progression of different types of cancer. GM-CSF is produced by various non-immune cells including MDA-MB-231 in response to various stimuli. However, the role of lipopolysaccharide (LPS) in the regulation of GM-CSF in MDA-MB-231 breast cancer cells so far remains unclear. Herein, we asked whether LPS could induce GM-CSF production in MDA-MB-231 cells, and if so, which signaling pathway was involved. MDA-MB-231 cells were treated with LPS or tumor necrosis factor alpha (TNF-α; positive control), and GM-CSF expression levels were determined by qRT-PCR, ELISA, and confocal microscopy. Phosphorylation of the mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-kB) signaling proteins were evaluated by flow cytometry. Our results show that LPS induces GM-CSF expression at both mRNA and protein levels in MDA-MBA-231 cells. Inhibition of acyl-CoA synthetase 1 (ACSL1) activity in the cells with triacsin C significantly reduces the secretion of GM-CSF. Furthermore, the inhibition of ACSL1 activity significantly blocks the LPS-mediated phosphorylation of p38 MAPK, MEK1/2, extracellular signal-regulated kinase (ERK)1/2, c-Jun NH2-terminal kinase (JNK), and nuclear factor-κB (NF-kB) in the cells. These findings provide the first evidence that LPS induces ACSL1-dependent GM-CSF gene expression in MDA-MB-231 breast cancer cells, which requires the activation of p38 MAPK, MEK1/2, ERK1/2, JNK, and NF-kB.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Coenzima A Ligasas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Lipopolisacáridos/farmacología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , MAP Quinasa Quinasa 4/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
15.
Cell Physiol Biochem ; 52(3): 397-407, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30845379

RESUMEN

BACKGROUND/AIMS: TNF-α-mediated pro-inflammatory phenotypic change in monocytes is known to be implicated in the pathogenesis of metabolic inflammation and insulin resistance. However, the mechanism by which TNF-α-induces inflammatory phenotypic shift in monocytes is poorly understood. Since long-chain acyl-CoA synthetase 1 (ACSL1) is associated with inflammatory monocytes/macrophages, we investigated the role of ACSL1 in the TNF-α-driven inflammatory phenotypic shift in the monocytes. METHODS: Monocytes (Human monocytic THP-1 cells) were stimulated with TNF-α. Inflammatory phenotypic markers (CD16, CD11b, CD11c and HLA-DR) expression was determined with real time RTPCR and flow cytometry. IL-1ß and MCP-1 were determined by ELISA. Signaling pathways were identified by using ACSL1 inhibitor, ACSL1 siRNA and NF-κB reporter monocytic cells. Phosphorylation of NF-κB was analyzed by western blotting and flow cytometry. RESULTS: Our data show that TNF-α induced significant increase in the expression of CD16, CD11b, CD11c and HLA-DR. Inhibition of ACSL1 activity in the cells with triacsin C significantly suppressed the expression of these inflammatory markers. Using ACSL-1 siRNA, we further demonstrate that TNF-α-induced inflammatory markers expression in monocytic cells requires ACSL1. In addition, IL-1b and MCP-1 production by TNF-α activated monocytic cells was significantly blocked by the inhibition of ACSL-1 activity. Interestingly, elevated NF-κB activity resulting from TNF-α stimulation was attenuated in ACSL1 deficient cells. CONCLUSION: Our findings provide an evidence that TNF-α-associated inflammatory polarization in monocytes is an ACSL1 dependent process, which indicates its central role in TNF-α-driven metabolic inflammation.


Asunto(s)
Coenzima A Ligasas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Factor de Necrosis Tumoral alfa/farmacología , Línea Celular , Quimiocina CCL2/análisis , Coenzima A Ligasas/antagonistas & inhibidores , Coenzima A Ligasas/genética , Antígenos HLA-DR/genética , Antígenos HLA-DR/metabolismo , Humanos , Inflamación/metabolismo , Interleucina-1beta/análisis , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Triazenos/química , Triazenos/metabolismo
16.
Ren Fail ; 41(1): 821-831, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31488013

RESUMEN

Background: Obesity has become a worldwide epidemic, and the incidence of obesity is increasing year by year. Obesity-related nephropathy (ORN) is a common kidney complication of obesity. Long-chain acyl-CoA synthetases-1, (ACSL1), is a key enzyme in the oxidative metabolism of fatty acids in mitochondria and ACSL1 may play a direct role in renal lipid deposition and promote the progress of ORN. In this study, we focus on the renoprotective role of ACSL1 in ORN. Methods: Electron microscopy, immunohistochemical (IHC) staining, Western blot, and real-time PCR were used to detect the expression of ACSL1and Nrf2 in ORN patients, ob/ob mice and palmitic acid (PA)-treated HK-2 cells. Oil red staining and Elisa Kit were used to detect the intracellular FFA and TG contents in ob/ob mice and PA-treated HK-2 cells. Dihydroethidium (DHE) staining and the MDA/SOD measurement were used to detect the ROS production. In order to demonstrate the role of ACSL1 and the interaction between ACSL1 and Nrf2 in ORN, related siRNA and plasmid were transfected into HK-2 cells. Results: More ROS production and renal lipid deposition have been found in ORN patients, ob/ob mice and PA-treated HK-2 cells. Compared with control, all the expression of ACSL1and Nrf2 were down-regulated in ORN patients, ob/ob mice and PA-treated HK-2 cells. The Nrf2 could regulate the expression of ACSL1 and the ACSL1 played the direct role in renal lipid deposition. Conclusions: The Nrf2 is inhibited in ORN, resulting more ROS production and oxidative stress. Increased oxidative stress will suppress the expression of ACSL1, which could increase the intracellular FFA and TG contents, ultimately leading to renal lipid deposition in renal tubulars and accelerating the development of ORN.


Asunto(s)
Coenzima A Ligasas/metabolismo , Enfermedades Renales/patología , Factor 2 Relacionado con NF-E2/metabolismo , Obesidad/complicaciones , Adulto , Animales , Biopsia , Línea Celular , Coenzima A Ligasas/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ácidos Grasos no Esterificados/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Enfermedades Renales/etiología , Túbulos Renales/patología , Túbulos Renales/ultraestructura , Masculino , Ratones , Microscopía Electrónica , Factor 2 Relacionado con NF-E2/genética , Obesidad/genética , Estrés Oxidativo , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Triglicéridos/metabolismo
17.
Int J Mol Sci ; 20(18)2019 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-31500376

RESUMEN

Accumulating evidence has shown that miR-34a serves as a posttranscriptional regulatory molecule of lipid metabolism in mammals. However, little studies about miR-34a on lipid metabolism in poultry have been reported until now. To gain insight into the biological functions and action mechanisms of miR-34a on hepatic lipid metabolism in poultry, we firstly investigated the expression pattern of miR-34a-5p, a member of miR-34a family, in liver of chicken, and determined its function in hepatocyte lipid metabolism by miR-34a-5p overexpression and inhibition, respectively. We then validated the interaction between miR-34a-5p and its target using dual-luciferase reporter assay, and explored the action mechanism of miR-34a-5p on its target by qPCR and Western blotting. Additionally, we looked into the function of the target gene on hepatocyte lipid metabolism by gain- and loss-of-function experiments. Our results indicated that miR-34a-5p showed a significantly higher expression level in livers in peak-laying hens than that in pre-laying hens. miR-34a-5p could increase the intracellular levels of triglycerides and total cholesterol in hepatocyte. Furthermore, miR-34a-5p functioned by inhibiting the translation of its target gene, long-chain acyl-CoA synthetase 1 (ACSL1), which negatively regulates hepatocyte lipid content. In conclusion, miR-34a-5p could increase intracellular lipid content by reducing the protein level, without influencing mRNA stability of the ACSL1 gene in chickens.


Asunto(s)
Pollos/genética , Pollos/metabolismo , Colesterol/metabolismo , Coenzima A Ligasas/genética , Hígado/metabolismo , MicroARNs/genética , Triglicéridos/metabolismo , Animales , Secuencia de Bases , Línea Celular , Coenzima A Ligasas/metabolismo , Expresión Génica , Metabolismo de los Lípidos , MicroARNs/química
18.
Int J Mol Sci ; 20(15)2019 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-31344914

RESUMEN

The deregulation of cancer cell metabolic networks is now recognized as one of the hallmarks of cancer. Abnormal lipid synthesis and extracellular lipid uptake are advantageous modifications fueling the needs of uncontrolled cancer cell proliferation. Fatty acids are placed at the crossroads of anabolic and catabolic pathways, as they are implicated in the synthesis of phospholipids and triacylglycerols, or they can undergo ß-oxidation. Key players to these decisions are the long-chain acyl-CoA synthetases, which are enzymes that catalyze the activation of long-chain fatty acids of 12-22 carbons. Importantly, the long-chain acyl-CoA synthetases are deregulated in many types of tumors, providing a rationale for anti-tumor therapeutic opportunities. The purpose of this review is to summarize the last up-to-date findings regarding their role in cancer, and to discuss the related emerging tumor targeting opportunities.


Asunto(s)
Coenzima A Ligasas/metabolismo , Lípidos/genética , Neoplasias/genética , Animales , Coenzima A Ligasas/genética , Humanos , Lípidos/biosíntesis , Neoplasias/enzimología , Neoplasias/patología , Fosfolípidos/biosíntesis , Fosfolípidos/metabolismo , Triglicéridos/biosíntesis , Triglicéridos/metabolismo
19.
J Biol Chem ; 291(10): 5373-84, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26728456

RESUMEN

Long-chain acyl-CoA synthetase 1 (ACSL1) plays a key role in fatty acid metabolism. To identify novel transcriptional modulators of ACSL1, we examined ACSL1 expression in liver tissues of hamsters fed a normal diet, a high fat diet, or a high cholesterol and high fat diet (HCHFD). Feeding hamsters HCHFD markedly reduced hepatic Acsl1 mRNA and protein levels as well as acyl-CoA synthetase activity. Decreases in Acsl1 expression strongly correlated with reductions in hepatic Srebp2 mRNA level and mature Srebp2 protein abundance. Conversely, administration of rosuvastatin (RSV) to hamsters increased hepatic Acsl1 expression. These new findings were reproduced in mice treated with RSV or fed the HCHFD. Furthermore, the RSV induction of acyl-CoA activity in mouse liver resulted in increases in plasma and hepatic cholesterol ester concentrations and reductions in free cholesterol amounts. Investigations on different ACSL1 transcript variants in HepG2 cells revealed that the mRNA expression of C-ACSL1 was specifically regulated by the sterol regulatory element (SRE)-binding protein (SREBP) pathway, and RSV treatment increased the C-ACSL1 abundance from a minor mRNA species to an abundant transcript. We analyzed 5'-flanking sequence of exon 1C of the human ACSL1 gene and identified one putative SRE site. By performing a promoter activity assay and DNA binding assays, we firmly demonstrated the key role of this SRE motif in SREBP2-mediated activation of C-ACSL1 gene transcription. Finally, we demonstrated that knockdown of endogenous SREBP2 in HepG2 cells lowered ACSL1 mRNA and protein levels. Altogether, this work discovered an unprecedented link between ACSL1 and SREBP2 via the specific regulation of the C-ACSL1 transcript.


Asunto(s)
Coenzima A Ligasas/metabolismo , Hígado/metabolismo , Elementos de Respuesta , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Animales , Sitios de Unión , Colesterol/sangre , Colesterol/metabolismo , Coenzima A Ligasas/química , Coenzima A Ligasas/genética , Cricetinae , Dieta Alta en Grasa , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Biochem Biophys Res Commun ; 484(3): 565-571, 2017 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-28132807

RESUMEN

The oncoprotein hepatitis B X-interacting protein (HBXIP) results in the dysregulation of lipid metabolism to enhance the development of breast cancer. Acyl-CoA synthetase long-chain family member 1 (ACSL1) is required for thioesterification of long-chain fatty acids into their acyl-CoA derivatives. In this study, we present a hypothesis that HBXIP might be involved in the regulation of ACSL1 in breast cancer. Interestingly, we found that the overexpression of HBXIP was able to up-regulate ACSL1 at the levels of mRNA and protein in a dose-dependent manner in breast cancer cells. Conversely, silencing of HBXIP led to the opposite results. Mechanistically, HBXIP as a coactivator interacted with transcriptional factor Sp1 through binding to the promoter of ACSL1 by ChIP assays analysis, leading to the transcription of ACSL1 in breast cancer cells. Immunohistochemistry staining revealed that the positive rate of ACSL1 was 71.4% (35/49) in clinical breast cancer tissues, HBXIP 79.6% (39/49), in which the positive rate of ACSL1 was 76.9% (30/39) in the HBXIP-positive specimens. But, few positive rate of ACSL1 10% (1/10) was observed in normal breast tissues. The mRNA levels of ACSL1 were significantly higher in clinical breast cancer tissues than those in their corresponding peritumor tissues. The mRNA levels of ACSL1 were positively associated with those of HBXIP in clinical breast cancer tissues. Thus, we conclude that the oncoprotein HBXIP is able to up-regulate ACSL1 through activating the transcriptional factor Sp1 in breast cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Coenzima A Ligasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción Sp1/metabolismo , Línea Celular Tumoral , Humanos , Transducción de Señal , Activación Transcripcional , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda