Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 293
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Pharmacol Res ; 201: 107105, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38367917

RESUMO

Chronic interstitial fibrosis presents a significant challenge to the long-term survival of transplanted kidneys. Our research has shown that reduced expression of acyl-coenzyme A oxidase 1 (ACOX1), which is the rate-limiting enzyme in the peroxisomal fatty acid ß-oxidation pathway, contributes to the development of fibrosis in renal allografts. ACOX1 deficiency leads to lipid accumulation and excessive oxidation of polyunsaturated fatty acids (PUFAs), which mediate epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) reorganization respectively, thus causing fibrosis in renal allografts. Furthermore, activation of Toll-like receptor 4 (TLR4)-nuclear factor kappa-B (NF-κB) signaling induced ACOX1 downregulation in a DNA methyltransferase 1 (DNMT1)-dependent manner. Overconsumption of PUFA resulted in endoplasmic reticulum (ER) stress, which played a vital role in facilitating ECM reorganization. Supplementation with PUFAs contributed to delayed fibrosis in a rat model of renal transplantation. The study provides a novel therapeutic approach that can delay chronic interstitial fibrosis in renal allografts by targeting the disorder of lipid metabolism.


Assuntos
Acil-CoA Oxidase , Transplante de Rim , Rim , Doenças Metabólicas , Animais , Ratos , Acil-CoA Oxidase/metabolismo , Aloenxertos , Fibrose , Rim/patologia , Lipídeos
2.
Exp Cell Res ; 430(1): 113700, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37380010

RESUMO

Growing evidence suggests a strong association between decreased lipid catabolism and the development of cancer. Solute carrier family 9 member A5 (SLC9A5) plays a regulatory role in colorectal function. However, the specific involvement of SLC9A5 in colorectal cancer (CRC) remains unclear, as well as its potential connection to lipid catabolism. We found that SLC9A5 exhibited significantly higher expression in CRC tumor tissues compared to adjacent paratumor tissues, as confirmed through analysis of the TCGA database and validation on a CRC tissue chip using IHC. Furthermore, in vitro experiments showed that knockdown of SLC9A5 resulted in suppressed cell proliferation, migration, and invasion. Then we performed bioinformatics analysis and found that SLC9A5 was significantly enriched in peroxisomal fatty acid oxidation (FAO) pathway and negatively correlated with its first rate-limiting enzyme acyl-CoA oxidases (ACOX). Interestingly, the expression of ACOX1, as well as FAO process indicated by changes in very long chain fatty acid levels, were enhanced upon SLC9A5 knockdown in CRC cells. Moreover, the attenuated tumor growth, migration, invasion, and increased FAO observed after SLC9A5 knockdown could be reversed by simultaneous knockdown of both SLC9A5 and ACOX1. In summary, these findings reveal the oncogenic role of SLC9A5 in CRC, particularly in relation to ACOX1-mediated peroxidation, and might serve as a promising therapeutic target for inhibiting the progression of colorectal cancer.


Assuntos
Neoplasias Colorretais , Ácidos Graxos , Humanos , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Lipídeos , Neoplasias Colorretais/patologia , Movimento Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética
3.
Mol Genet Genomics ; 298(6): 1247-1260, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37555868

RESUMO

ß-oxidation of fatty acids is an important metabolic pathway and is a shared function between mitochondria and peroxisomes in mammalian cells. On the other hand, peroxisomes are the sole site for the degradation of fatty acids in yeast. The first reaction of this pathway is catalyzed by the enzyme acyl CoA oxidase housed in the matrix of peroxisomes. Studies in various model organisms have reported the conserved function of the protein in fatty acid oxidation. The importance of this enzyme is highlighted by the lethal conditions caused in humans due to its altered function. In this review, we discuss various aspects ranging from gene expression, structure, folding, and import of the protein in both yeast and human cells. Further, we highlight recent findings on the role of the protein in human health and aging, and discuss the identified mutations in the protein associated with debilitating conditions in patients.


Assuntos
Peroxissomos , Saccharomyces cerevisiae , Animais , Humanos , Acil-CoA Oxidase/metabolismo , Saccharomyces cerevisiae/metabolismo , Peroxissomos/genética , Peroxissomos/metabolismo , Oxirredução , Ácidos Graxos/metabolismo , Mamíferos
4.
PLoS Pathog ; 17(7): e1009767, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34270617

RESUMO

Hypobiosis (facultative developmental arrest) is the most important life-cycle adaptation ensuring survival of parasitic nematodes under adverse conditions. Little is known about such survival mechanisms, although ascarosides (ascarylose with fatty acid-derived side chains) have been reported to mediate the formation of dauer larvae in the free-living nematode Caenorhabditis elegans. Here, we investigated the role of a key gene acox-1, in the larval development of Haemonchus contortus, one of the most important parasitic nematodes that employ hypobiosis as a routine survival mechanism. In this parasite, acox-1 encodes three proteins (ACOXs) that all show a fatty acid oxidation activity in vitro and in vivo, and interact with a peroxin PEX-5 in peroxisomes. In particular, a peroxisomal targeting signal type1 (PTS1) sequence is required for ACOX-1 to be recognised by PEX-5. Analyses on developmental transcription and tissue expression show that acox-1 is predominantly expressed in the intestine and hypodermis of H. contortus, particularly in the early larval stages in the environment and the arrested fourth larval stage within host animals. Knockdown of acox-1 and pex-5 in parasitic H. contortus shows that these genes play essential roles in the post-embryonic larval development and likely in the facultative arrest of this species. A comprehensive understanding of these genes and the associated ß-oxidation cycle of fatty acids should provide novel insights into the developmental regulation of parasitic nematodes, and into the discovery of novel interventions for species of socioeconomic importance.


Assuntos
Acil-CoA Oxidase/metabolismo , Haemonchus/metabolismo , Proteínas de Helminto/metabolismo , Larva/metabolismo , Receptor 1 de Sinal de Orientação para Peroxissomos/metabolismo , Animais , Hemoncose/metabolismo , Haemonchus/crescimento & desenvolvimento , Larva/crescimento & desenvolvimento , Coelhos , Ovinos
5.
Biochem Biophys Res Commun ; 613: 47-52, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35526488

RESUMO

Peroxisome proliferator-activated receptor α (PPARα) regulates fatty acid oxidation (FAO). Usually, very-long chain fatty acids are first activated by acyl-CoA synthetase (ACS) to generate acyl-CoA for oxidation by acyl-CoA oxidase (ACOX) in peroxisomes, and the resultant shorter chain fatty acids will be further oxidized in mitochondria. ACS long-chain family member 4 (ACSL4) preferentially uses arachidonic acid (AA) as substrates to synthesize arachidonoyl-CoA. Arachidonoyl-CoA is usually esterified into phospholipids. When AA is released by phospholipase A2 (PLA2) from phospholipids, it will be used for prostaglandin synthesis by cyclooxygenases (COX). In this study, when PPARα agonist WY-14,643 was mixed in liquid Lieber-DeCarli ethanol or control diets and fed to mice, liver PLA2, COX-2, and ACOX1 were induced but ACSL4 was inhibited, suggesting that AA released by PLA2 from phospholipid will be metabolized to prostaglandin via COX-2 instead of being synthesized into acyl-CoA by ACSL4. However, liver prostaglandin E2 (PGE2), a major component of prostaglandin, was not increased with the induced COX-2 but decreased by WY-14,643. ACOX1 specific inhibitor mixed in the liquid diets restored both the WY-14,643-suppressed liver TG and PGE2, but COX-2 specific inhibitor celecoxib mixed in the liquid diets reversed the WY-14,643-suppressed liver TG but not liver PGE2 contents. These results suggest that induction of PLA2, COX-2 and ACOX1 orchestrates to increase oxidation of AA/PGE2, which constitutes one new mechanism by which PPARα induces peroxisomal FAO and inhibits ethanol-induced liver fat accumulation.


Assuntos
Acil-CoA Oxidase , Ciclo-Oxigenase 2 , Fígado Gorduroso Alcoólico , PPAR alfa , Fosfolipases A2 , Pirimidinas , Acil-CoA Oxidase/metabolismo , Animais , Coenzima A/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ácidos Graxos/metabolismo , Fígado Gorduroso Alcoólico/tratamento farmacológico , Fígado Gorduroso Alcoólico/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/metabolismo , Camundongos , PPAR alfa/agonistas , PPAR alfa/metabolismo , Peroxissomos/efeitos dos fármacos , Peroxissomos/metabolismo , Fosfolipases A2/metabolismo , Fosfolipídeos/metabolismo , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
6.
Mar Drugs ; 20(8)2022 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-35892940

RESUMO

Filter-feeding bivalves can accumulate paralytic shellfish toxins (PST) produced by toxic microalgae, which may induce oxidative stress and lipid peroxidation. Peroxisomal acyl-coenzyme A oxidases (ACOXs) are key enzymes functioning in maintaining redox and lipid homeostasis, but their roles in PST response in bivalves are less understood. Herein, a total of six and six ACOXs were identified in the Chlamys farreri and Patinopecten yessoensis genome, respectively, and the expansion of ACOX1s was observed. Gene expression analysis revealed an organ/tissue-specific expression pattern in both scallops, with all ACOXs being predominantly expressed in the two most toxic organs, digestive glands and kidneys. The regulation patterns of scallop ACOXs after exposure to different PST-producing algaes Alexandrium catenella (ACDH) and A. minutum (AM-1) were revealed. After ACDH exposure, more differentially expressed genes (DEGs) were identified in C. farreri digestive glands (three) and kidneys (five) than that in P. yessoensis (two), but the up-regulated DEGs showed similar expression patterns in both species. In C. farreri, three DEGs were found in both digestive glands and kidneys after AM-1 exposure, with two same CfACOX1s being acutely and chronically induced, respectively. Notably, these two CfACOX1s also showed different expression patterns in kidneys between ACDH (acute response) and AM-1 (chronic response) exposure. Moreover, inductive expression of CfACOXs after AM-1 exposure was observed in gills and mantles, and all DEGs in both tissues were up-regulated and their common DEGs exhibited both acute and chronic induction. These results indicate the involvement of scallop ACOXs in PST response, and their plasticity expression patterns between scallop species, among tissues, and between the exposure of different PST analogs.


Assuntos
Bivalves , Dinoflagellida , Pectinidae , Toxinas Biológicas , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Animais , Bivalves/metabolismo , Coenzima A/metabolismo , Dinoflagellida/genética , Dinoflagellida/metabolismo , Oxirredução , Pectinidae/genética
7.
Proc Natl Acad Sci U S A ; 116(21): 10576-10585, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31064880

RESUMO

Epigenetic markers, such as histone acetylation and DNA methylation, determine chromatin organization. In eukaryotic cells, metabolites from organelles or the cytosol affect epigenetic modifications. However, the relationships between metabolites and epigenetic modifications are not well understood in plants. We found that peroxisomal acyl-CoA oxidase 4 (ACX4), an enzyme in the fatty acid ß-oxidation pathway, is required for suppressing the silencing of some endogenous loci, as well as Pro35S:NPTII in the ProRD29A:LUC/C24 transgenic line. The acx4 mutation reduces nuclear histone acetylation and increases DNA methylation at the NOS terminator of Pro35S:NPTII and at some endogenous genomic loci, which are also targeted by the demethylation enzyme REPRESSOR OF SILENCING 1 (ROS1). Furthermore, mutations in multifunctional protein 2 (MFP2) and 3-ketoacyl-CoA thiolase-2 (KAT2/PED1/PKT3), two enzymes in the last two steps of the ß-oxidation pathway, lead to similar patterns of DNA hypermethylation as in acx4 Thus, metabolites from fatty acid ß-oxidation in peroxisomes are closely linked to nuclear epigenetic modifications, which may affect diverse cellular processes in plants.


Assuntos
Arabidopsis/metabolismo , Metilação de DNA , Epigênese Genética , Ácidos Graxos/metabolismo , Peroxissomos/metabolismo , ATP Citrato (pro-S)-Liase/metabolismo , Acetilação , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Regulação da Expressão Gênica de Plantas , Histonas/metabolismo , Oxirredução , Plantas Geneticamente Modificadas , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo
8.
Plant Foods Hum Nutr ; 77(4): 529-537, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35986845

RESUMO

The aim of the present study was to investigate the browning effects mechanism of Smilax china L. polyphenols (SCLP) and its monomer. In this study, polyphenols (SCLP, engeletin, quercetin and caffeic acid) markedly suppressed lipid accumulation. Polyphenols significantly up-graded the expression of protein kinase A (PKA), adipose triglyceride lipase (ATGL), peroxisome proliferators-activated receptors alpha (PPARα), carnitine palmitoyl transferase (CPT) and acyl-CoA oxidase (ACO) to promote lipolysis and ß-oxidation. Moreover, polyphenols greatly enhanced mitochondrial biogenesis in adipocytes, as demonstrated by the expression of Nrf1 and Tfam were up-regulated. Furthermore, polyphenols treatment greatly up-regulated the browning program in adipocytes by increased brown-specific genes and proteins uncoupling protein 1 (UCP-1), peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and PR domain containing 16 (PRDM16), as well as beige-specific genes (Tmem26, Tbx1, CD137, Cited1), especially engeletin. Further research found that the brown-specific markers were decreased by antagonist treatment of AMPK or ß3-AR, but polyphenols treatment reversed the effect of antagonists and improved the expression of UCP-1, PRDM16 and PGC-1α. In conclusion, these results indicated that polyphenols stimulate browning in adipocytes via activation of the ß3-AR/AMPK signaling pathway, and SCLP and its monomer may be worth investigating to prevent obesity.


Assuntos
Polifenóis , Smilax , Animais , Camundongos , Células 3T3-L1 , Acil-CoA Oxidase/metabolismo , Adipócitos Marrons/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Carnitina/metabolismo , China , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Lipase/metabolismo , Lipídeos , Polifenóis/farmacologia , PPAR alfa/metabolismo , PPAR gama/metabolismo , Quercetina/farmacologia , Transdução de Sinais , Smilax/metabolismo , Fatores de Transcrição/metabolismo , Proteína Desacopladora 1/metabolismo
9.
Traffic ; 20(7): 504-515, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31042004

RESUMO

Most soluble proteins targeted to the peroxisomal matrix contain a C-terminal peroxisome targeting signal type 1 (PTS1) or an N-terminal PTS2 that is recognized by the receptors Pex5p and Pex7p, respectively. These receptors cycle between the cytosol and peroxisome and back again for multiple rounds of cargo delivery to the peroxisome. A small number of peroxisomal matrix proteins, including all six isozymes of peroxisomal fatty acyl-CoA oxidase (Aox) of the yeast Yarrowia lipolytica, contain neither a PTS1 nor a PTS2. Pex20p has been shown to function as a co-receptor for Pex7p in the import of PTS2 cargo into peroxisomes. Here we show that cells of Y. lipolytica deleted for the PEX20 gene fail to import not only the PTS2-containing protein 3-ketoacyl-CoA thiolase (Pot1p) but also the non-PTS1/non-PTS2 Aox isozymes. Pex20p binds directly to Aox isozymes Aox3p and Aox5p, which requires the C-terminal Wxxx(F/Y) motif of Pex20p. A W411G mutation in the C-terminal Wxxx(F/Y) motif causes Aox isozymes to be mislocalized to the cytosol. Pex20p interacts physically with members of the peroxisomal import docking complex, Pex13p and Pex14p. Our results are consistent with a role for Pex20p as the receptor for import of the non-PTS1/non-PTS2 Aox isozymes into peroxisomes.


Assuntos
Acil-CoA Oxidase/metabolismo , Proteínas Fúngicas/metabolismo , Peroxissomos/metabolismo , Sítios de Ligação , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Ligação Proteica , Transporte Proteico , Proteínas de Ligação a Telômeros/metabolismo , Yarrowia/genética , Yarrowia/metabolismo
10.
J Cell Physiol ; 236(1): 625-640, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32583421

RESUMO

Lipid deposition caused by the disorder of renal lipid metabolism is involved in diabetic nephropathy (DN). Carbohydrate response element-binding protein (ChREBP) is a key transcription factor in high glucose-induced cellular fat synthesis. At present, the regulation and mechanism of ChREBP on fat metabolism in diabetic kidneys are still unclear. In this study, we showed that lack of ChREBP significantly improved renal injury, inhibited oxidative stress, lipid deposition, fatty acid synthase (FASN), acetyl-CoA carboxylase (ACC) and thioredoxin-interacting protein (TXNIP) expression, as well as the activity of mammalian target of rapamycin complex 1 (mTORC1) in diabetic kidneys. Meanwhile, ChREBP deficiency upregulated the expression of peroxisome proliferator-activated receptor-α (PPARα), carnitine palmitoyltransferaser 1A (CPT1A) and acyl-coenzyme A oxidase 1 (ACOX1) in diabetic kidneys. In vitro, knockdown of ChREBP attenuated lipid deposition, mTORC1 activation, and expression of FASN and ACC, increased PPARα, CPT1A, and ACOX1 expression in HK-2 cells and podocytes under high glucose (HG) conditions. Moreover, HG-induced lipid deposition, increased expression of FASN and ACC and decreased expression of PPARα, CPT1A, and ACOX1 were reversed by rapamycin, a specific inhibitor of mTORC1, in HK-2 cells. These results indicate that ChREBP deficiency alleviates diabetes-associated renal lipid accumulation by inhibiting mTORC1 activity and suggest that reduction of ChREBP is a potential therapeutic strategy to treat DN.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Nefropatias Diabéticas/metabolismo , Metabolismo dos Lipídeos/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Acil-CoA Oxidase/metabolismo , Animais , Carnitina O-Palmitoiltransferase/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Diabetes Mellitus/metabolismo , Ácido Graxo Sintases/metabolismo , Regulação da Expressão Gênica/fisiologia , Glucose/metabolismo , Humanos , Rim/metabolismo , Camundongos Knockout , PPAR alfa/metabolismo , Podócitos/metabolismo , Regulação para Cima/fisiologia
11.
Proc Natl Acad Sci U S A ; 115(30): E7015-E7022, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29987047

RESUMO

Robust molecular tool kits in model and industrial microalgae are key to efficient targeted manipulation of endogenous and foreign genes in the nuclear genome for basic research and, as importantly, for the development of algal strains to produce renewable products such as biofuels. While Cas9-mediated gene knockout has been demonstrated in a small number of algal species with varying efficiency, the ability to stack traits or generate knockout mutations in two or more loci are often severely limited by selectable agent availability. This poses a critical hurdle in developing production strains, which require stacking of multiple traits, or in probing functionally redundant gene families. Here, we combine Cas9 genome editing with an inducible Cre recombinase in the industrial alga Nannochloropsis gaditana to generate a strain, NgCas9+Cre+, in which the potentially unlimited stacking of knockouts and addition of new genes is readily achievable. Cre-mediated marker recycling is first demonstrated in the removal of the selectable marker and GFP reporter transgenes associated with the Cas9/Cre construct in NgCas9+Cre+ Next, we show the proof-of-concept generation of a markerless knockout in a gene encoding an acyl-CoA oxidase (Aco1), as well as the markerless recapitulation of a 2-kb insert in the ZnCys gene 5'-UTR, which results in a doubling of wild-type lipid productivity. Finally, through an industrially oriented process, we generate mutants that exhibit up to ∼50% reduction in photosynthetic antennae size by markerless knockout of seven genes in the large light-harvesting complex gene family.


Assuntos
Acil-CoA Oxidase , Sistemas CRISPR-Cas , Edição de Genes , Lipídeos , Característica Quantitativa Herdável , Estramenópilas , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Complexos de Proteínas Captadores de Luz/genética , Complexos de Proteínas Captadores de Luz/metabolismo , Lipídeos/biossíntese , Lipídeos/genética , Estramenópilas/genética , Estramenópilas/metabolismo
12.
Proc Natl Acad Sci U S A ; 115(6): 1262-1267, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29358378

RESUMO

Bio-based production of fatty acids and fatty acid-derived products can enable sustainable substitution of petroleum-derived fuels and chemicals. However, developing new microbial cell factories for producing high levels of fatty acids requires extensive engineering of lipid metabolism, a complex and tightly regulated metabolic network. Here we generated a Saccharomyces cerevisiae platform strain with a simplified lipid metabolism network with high-level production of free fatty acids (FFAs) due to redirected fatty acid metabolism and reduced feedback regulation. Deletion of the main fatty acid activation genes (the first step in ß-oxidation), main storage lipid formation genes, and phosphatidate phosphatase genes resulted in a constrained lipid metabolic network in which fatty acid flux was directed to a large extent toward phospholipids. This resulted in simultaneous increases of phospholipids by up to 2.8-fold and of FFAs by up to 40-fold compared with wild-type levels. Further deletion of phospholipase genes PLB1 and PLB2 resulted in a 46% decrease in FFA levels and 105% increase in phospholipid levels, suggesting that phospholipid hydrolysis plays an important role in FFA production when phospholipid levels are increased. The multiple deletion mutant generated allowed for a study of fatty acid dynamics in lipid metabolism and represents a platform strain with interesting properties that provide insight into the future development of lipid-related cell factories.


Assuntos
Ácidos Graxos/metabolismo , Metabolismo dos Lipídeos , Saccharomyces cerevisiae/metabolismo , Acil Coenzima A/genética , Acil Coenzima A/metabolismo , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Deleção de Genes , Regulação Fúngica da Expressão Gênica , Metabolismo dos Lipídeos/genética , Lisofosfolipase/genética , Lisofosfolipase/metabolismo , Lipídeos de Membrana/biossíntese , Lipídeos de Membrana/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfatidato Fosfatase/genética , Fosfatidato Fosfatase/metabolismo , Fosfolipídeos/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
13.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34445672

RESUMO

In mammalian cells, two cellular organelles, mitochondria and peroxisomes, share the ability to degrade fatty acid chains. Although each organelle harbors its own fatty acid ß-oxidation pathway, a distinct mitochondrial system feeds the oxidative phosphorylation pathway for ATP synthesis. At the same time, the peroxisomal ß-oxidation pathway participates in cellular thermogenesis. A scientific milestone in 1965 helped discover the hepatomegaly effect in rat liver by clofibrate, subsequently identified as a peroxisome proliferator in rodents and an activator of the peroxisomal fatty acid ß-oxidation pathway. These peroxisome proliferators were later identified as activating ligands of Peroxisome Proliferator-Activated Receptor α (PPARα), cloned in 1990. The ligand-activated heterodimer PPARα/RXRα recognizes a DNA sequence, called PPRE (Peroxisome Proliferator Response Element), corresponding to two half-consensus hexanucleotide motifs, AGGTCA, separated by one nucleotide. Accordingly, the assembled complex containing PPRE/PPARα/RXRα/ligands/Coregulators controls the expression of the genes involved in liver peroxisomal fatty acid ß-oxidation. This review mobilizes a considerable number of findings that discuss miscellaneous axes, covering the detailed expression pattern of PPARα in species and tissues, the lessons from several PPARα KO mouse models and the modulation of PPARα function by dietary micronutrients.


Assuntos
Ácidos Graxos/metabolismo , PPAR alfa/metabolismo , Peroxissomos/metabolismo , Acil-CoA Oxidase/metabolismo , Animais , Humanos , Fígado/metabolismo , Oxirredução , Oxirredutases/metabolismo , PPAR alfa/fisiologia , Proliferadores de Peroxissomos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Elementos de Resposta/genética , Receptores X de Retinoides/metabolismo , Ativação Transcricional/genética
14.
EMBO Rep ; 19(5)2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29491006

RESUMO

Peroxisomes account for ~35% of total H2O2 generation in mammalian tissues. Peroxisomal ACOX1 (acyl-CoA oxidase 1) is the first and rate-limiting enzyme in fatty acid ß-oxidation and a major producer of H2O2 ACOX1 dysfunction is linked to peroxisomal disorders and hepatocarcinogenesis. Here, we show that the deacetylase sirtuin 5 (SIRT5) is present in peroxisomes and that ACOX1 is a physiological substrate of SIRT5. Mechanistically, SIRT5-mediated desuccinylation inhibits ACOX1 activity by suppressing its active dimer formation in both cultured cells and mouse livers. Deletion of SIRT5 increases H2O2 production and oxidative DNA damage, which can be alleviated by ACOX1 knockdown. We show that SIRT5 downregulation is associated with increased succinylation and activity of ACOX1 and oxidative DNA damage response in hepatocellular carcinoma (HCC). Our study reveals a novel role of SIRT5 in inhibiting peroxisome-induced oxidative stress, in liver protection, and in suppressing HCC development.


Assuntos
Acil-CoA Oxidase/antagonistas & inibidores , Acil-CoA Oxidase/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Estresse Oxidativo , Sirtuínas/metabolismo , Acil-CoA Oxidase/genética , Animais , Dano ao DNA , Regulação para Baixo , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Peróxido de Hidrogênio , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Oxirredução , Peroxissomos/química , Prognóstico , Sirtuínas/genética
15.
Biol Pharm Bull ; 43(9): 1382-1392, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32879213

RESUMO

The effects of different dietary fats on hepatic fatty acid oxidation were compared in male ICR mice and Sprague-Dawley rats. Animals were fed diets containing 100 g/kg of either palm oil (saturated fat), safflower oil (rich in linoleic acid), an oil of evening primrose origin (γ-linolenic acid, GLA oil), perilla oil (α-linolenic acid) or fish oil (eicosapentaenoic and doxosahexaenoic acids) for 21 d. GLA, perilla and fish oils, compared with palm and safflower oils, increased the activity of fatty acid oxidation enzymes in both mice and rats, with some exceptions. In mice, GLA and fish oils greatly increased the peroxisomal palmitoyl-CoA oxidation rate, and the activity of acyl-CoA oxidase and enoyl-CoA hydratase to the same degree. The effects were much smaller with perilla oil. In rats, enhancing effects were more notable with fish oil than with GLA and perilla oils, excluding the activity of enoyl-CoA hydratase, and were comparable between GLA and perilla oils. In mice, strong enhancing effects of GLA oil, which were greater than with perilla oil and comparable to those of fish oil, were confirmed on mRNA levels of peroxisomal but not mitochondrial fatty acid oxidation enzymes. In rats, the effects of GLA and perilla oils on mRNA levels of peroxisomal and mitochondrial enzymes were indistinguishable, and lower than those observed with fish oil. Therefore, considerable diversity in the response to dietary polyunsaturated fats, especially the oil rich in γ-linolenic acid and fish oil, of hepatic fatty acid oxidation pathway exists between mice and rats.


Assuntos
Gorduras na Dieta/administração & dosagem , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Ácido gama-Linolênico/administração & dosagem , Acil-CoA Oxidase/metabolismo , Ração Animal , Animais , Enoil-CoA Hidratase/metabolismo , Óleos de Peixe/administração & dosagem , Óleos de Peixe/química , Fígado/citologia , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Oxirredução/efeitos dos fármacos , Peroxissomos/efeitos dos fármacos , Peroxissomos/enzimologia , Óleos de Plantas/administração & dosagem , Óleos de Plantas/química , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
16.
Fish Physiol Biochem ; 46(5): 1645-1652, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32601856

RESUMO

Acyl-coenzyme A oxidases 1 (ACOX1) is the first rate-limiting enzyme responsible for peroxisomal ß-oxidation. In the present study, two mRNA variants, ACOX1a and ACOX1b, transcribed from a single gene, were for the first time isolated and characterized from grass carp Ctenopharyngodon idella, both encoding putative peptides of 660 amino acids. Analysis of the exon-intron structures clarified that grass carp ACOX1a and ACOX1b comprise 14 coding exons and correspond to 3a and 3b isoforms of exon 3 splicing variants. Both ACOX1a and ACOX1b mRNAs were expressed in a wide range of tissues, but the abundance of each ACOX1 mRNA showed the tissue-dependent expression patterns. Time-course analysis of ACOX1 expressions indicated that the level of ACOX1a mRNA reached an almost maximal level at day 2, while that of ACOX1b mRNA reached an almost maximal level at day 8 during grass carp primary preadipocyte differentiation. In fasting-induced adipocyte lipolysis, only ACOX1a showed a significant increase in adipocyte, indicating that two ACOX1 isoforms may serve somewhat different roles in the peroxisomal ß-oxidation. These results suggested that grass carp ACOX1a and ACOX1b were differently modulated by fasting in adipocyte. In addition, we found that mitochondrial ß-oxidation might dominate at the early stage of fasting in adipocytes, indicating that mitochondria and peroxisomes might possess different capacities in fasting-induced adipocytes fatty acid oxidation.


Assuntos
Acil-CoA Oxidase/metabolismo , Adipócitos/metabolismo , Carpas/metabolismo , Privação de Alimentos , Regulação Enzimológica da Expressão Gênica/fisiologia , Metabolismo dos Lipídeos/fisiologia , Acil-CoA Oxidase/genética , Processamento Alternativo , Animais , Clonagem Molecular , Isoenzimas
17.
Lipids Health Dis ; 18(1): 11, 2019 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-30621686

RESUMO

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is closely linked to obesity, type 2 diabetes and other metabolic disorders worldwide. Crocin is a carotenoid compound possessing various pharmacological activities. In the present study, we aimed to investigate the effect on fatty liver under diabetic and obese condition and to examine the possible role of AMP-activated protein kinase (AMPK) signaling. METHODS: db/db mice were administrated with crocin and injected with LV-shAMPK or its negative control lentivirus. Metabolic dysfunction, lipogenesis and fatty acid-oxidation in liver were evaluated. RESULTS: In db/db mice, we found that oral administration of crocin significantly upregulated the phosphorylation of AMPK and downregulated the phosphorylation of mTOR in liver. Crocin reduced liver weight, serum levels of alanine aminotransferase, alanine aminotransferase, and liver triglyceride content, and attenuated morphological injury of liver in db/db mice. Crocin inhibited the mRNA expression of lipogenesis-associated genes, including sterol regulatory element binding protein-1c, peroxisome proliferator-activated receptor γ, fatty acid synthase, stearoyl-CoA desaturase 1, and diacylglycerol acyltransferase 1, and increased the mRNA expression of genes involved in the regulation of ß-oxidation of fatty acids, including PPARα, acyl-CoA oxidase 1, carnitine palmitoyltransferase 1, and 3-hydroxy-3-methylglutaryl-CoA synthase 2. Moreover, treatment of crocin resulted in a amelioration of general metabolic disorder, as evidenced by decreased fasting blood glucose, reduced serum levels of insulin, triglyceride, total cholesterol, and non-esterified fatty acid, and improved glucose intolerance. Crocin-induced protective effects against fatty liver and metabolic disorder were significantly blocked by lentivirus-mediated downregulation of AMPK. CONCLUSIONS: The results suggest that crocin can inhibit lipogenesis and promote ß-oxidation of fatty acids through activation of AMPK, leading to improvement of fatty liver and metabolic dysfunction. Therefore, crocin may be a potential promising option for the clinical treatment for NAFLD and associated metabolic diseases.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Fármacos Antiobesidade/farmacologia , Carotenoides/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/farmacologia , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Proteínas Quinases Ativadas por AMP/metabolismo , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Alanina Transaminase/sangue , Alanina Transaminase/genética , Animais , Aspartato Aminotransferases/sangue , Aspartato Aminotransferases/genética , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Diacilglicerol O-Aciltransferase/antagonistas & inibidores , Diacilglicerol O-Aciltransferase/genética , Diacilglicerol O-Aciltransferase/metabolismo , Modelos Animais de Doenças , Ácido Graxo Sintases/antagonistas & inibidores , Ácido Graxo Sintases/genética , Ácido Graxo Sintases/metabolismo , Regulação da Expressão Gênica , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , PPAR alfa/agonistas , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gama/antagonistas & inibidores , PPAR gama/genética , PPAR gama/metabolismo , Transdução de Sinais , Estearoil-CoA Dessaturase/antagonistas & inibidores , Estearoil-CoA Dessaturase/genética , Estearoil-CoA Dessaturase/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/antagonistas & inibidores , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Resultado do Tratamento , Triglicerídeos/sangue
18.
Proc Natl Acad Sci U S A ; 113(36): 10055-60, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27551084

RESUMO

Caenorhabditis elegans secretes ascarosides as pheromones to communicate with other worms and to coordinate the development and behavior of the population. Peroxisomal ß-oxidation cycles shorten the side chains of ascaroside precursors to produce the short-chain ascaroside pheromones. Acyl-CoA oxidases, which catalyze the first step in these ß-oxidation cycles, have different side chain-length specificities and enable C. elegans to regulate the production of specific ascaroside pheromones. Here, we determine the crystal structure of the acyl-CoA oxidase 1 (ACOX-1) homodimer and the ACOX-2 homodimer bound to its substrate. Our results provide a molecular basis for the substrate specificities of the acyl-CoA oxidases and reveal why some of these enzymes have a very broad substrate range, whereas others are quite specific. Our results also enable predictions to be made for the roles of uncharacterized acyl-CoA oxidases in C. elegans and in other nematode species. Remarkably, we show that most of the C. elegans acyl-CoA oxidases that participate in ascaroside biosynthesis contain a conserved ATP-binding pocket that lies at the dimer interface, and we identify key residues in this binding pocket. ATP binding induces a structural change that is associated with tighter binding of the FAD cofactor. Mutations that disrupt ATP binding reduce FAD binding and reduce enzyme activity. Thus, ATP may serve as a regulator of acyl-CoA oxidase activity, thereby directly linking ascaroside biosynthesis to ATP concentration and metabolic state.


Assuntos
Acil-CoA Oxidase/química , Proteínas de Caenorhabditis elegans/química , Caenorhabditis elegans/química , Feromônios/química , Acil-CoA Oxidase/genética , Acil-CoA Oxidase/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Caenorhabditis elegans/enzimologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Cristalografia por Raios X , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Expressão Gênica , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Modelos Moleculares , Mutação , Oxirredução , Feromônios/biossíntese , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Especificidade por Substrato
19.
Hepatobiliary Pancreat Dis Int ; 18(4): 360-365, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31126802

RESUMO

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent chronic liver diseases. However, the exact pathogenesis of NAFLD remains to be elucidated. Despite the association with tumors and cardiovascular diseases, the role of miR-222 in NAFLD remains unclear. The present study was to investigate the role of miR-222 in NAFLD. METHODS: Wild-type C57BL/6 mice were fed a high-fat diet for 12 weeks to induce NAFLD. Normal human liver cell line (L02) was cultured with free fatty acid (FFA)-containing medium to stimulate cell steatosis. The mRNA levels of miR-222 and acyl Coenzyme A xidase 1 (ACOX1) were detected by quantitative-PCR (Q-PCR). The prediction of ACOX1 as the target gene for miR-222 was conducted via TargetScan. The overexpression or inhibition of miR-222 was mediated by miR-222 mimics or antagomir, and intracellular triglyceride levels were measured using a triglyceride kit. Luciferase reporter assays verified ACOX1 as the target gene for miR-222. RESULTS: miR-222 was significantly elevated in both the in vivo and in vitro NAFLD models. Overexpression of miR-222 significantly increased triglyceride content in the L02 cells, while inhibition of miR-222 expression restricted the accumulation of triglyceride. Overexpression of miR-222 significantly inhibited ACOX1 expression. Transient transfection assays verified that ACOX1 3'-UTR luciferase reporter activity could be inhibited by miR-222 overexpression. CONCLUSIONS: The present study suggested that miR-222 promotes the accumulation of triglycerides by inhibiting ACOX1.


Assuntos
Acil-CoA Oxidase/metabolismo , Hepatócitos/enzimologia , MicroRNAs/metabolismo , Hepatopatia Gordurosa não Alcoólica/enzimologia , Oxirredutases/metabolismo , Triglicerídeos/metabolismo , Regiões 3' não Traduzidas , Acil-CoA Oxidase/genética , Animais , Sítios de Ligação , Linhagem Celular , Dieta Hiperlipídica , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Hepatócitos/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Oxirredutases/genética , Regulação para Cima
20.
J Biol Chem ; 292(9): 3800-3809, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28077576

RESUMO

A chronic high fat diet results in hepatic mitochondrial dysfunction and induction of peroxisomal fatty acid oxidation (FAO); whether specific inhibition of peroxisomal FAO benefits mitochondrial FAO and reactive oxygen species (ROS) metabolism remains unclear. In this study a specific inhibitor for the rate-limiting enzyme involved in peroxisomal FAO, acyl-CoA oxidase-1 (ACOX1) was developed and used for the investigation of peroxisomal FAO inhibition upon mitochondrial FAO and ROS metabolism. Specific inhibition of ACOX1 by 10,12-tricosadiynoic acid increased hepatic mitochondrial FAO via activation of the SIRT1-AMPK (adenosine 5'-monophosphate-activated protein kinase) pathway and proliferator activator receptor α and reduced hydrogen peroxide accumulation in high fat diet-fed rats, which significantly decreased hepatic lipid and ROS contents, reduced body weight gain, and decreased serum triglyceride and insulin levels. Inhibition of ACOX1 is a novel and effective approach for the treatment of high fat diet- or obesity-induced metabolic diseases by improving mitochondrial lipid and ROS metabolism.


Assuntos
Acil-CoA Oxidase/metabolismo , Ácidos Graxos Insaturados/farmacologia , Lipídeos/química , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Acil-CoA Oxidase/antagonistas & inibidores , Animais , Peso Corporal , Dieta Hiperlipídica , Insulina/metabolismo , Fígado/metabolismo , Masculino , Mitocôndrias/metabolismo , Oxigênio/química , Peroxissomos/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/metabolismo , Sirtuína 1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA