Skip to main content
World Journal of Gastroenterology logoLink to World Journal of Gastroenterology
. 2015 Mar 28;21(12):3492–3498. doi: 10.3748/wjg.v21.i12.3492

Long-chain acyl-CoA synthetase in fatty acid metabolism involved in liver and other diseases: An update

Sheng Yan 1,2,3, Xue-Feng Yang 1,2,3, Hao-Lei Liu 1,2,3, Nian Fu 1,2,3, Yan Ouyang 1,2,3, Kai Qing 1,2,3
PMCID: PMC4375570  PMID: 25834313

Abstract

Long-chain acyl-CoA synthetase (ACSL) family members include five different ACSL isoforms, each encoded by a separate gene and have multiple spliced variants. ACSLs on endoplasmic reticulum and mitochondrial outer membrance catalyze fatty acids with chain lengths from 12 to 20 carbon atoms to form acyl-CoAs, which are lipid metabolic intermediates and involved in fatty acid metabolism, membrane modifications and various physiological processes. Gain- or loss-of-function studies have shown that the expression of individual ACSL isoforms can alter the distribution and amount of intracellular fatty acids. Changes in the types and amounts of fatty acids, in turn, can alter the expression of intracellular ACSLs. ACSL family members affect not only the proliferation of normal cells, but the proliferation of malignant tumor cells. They also regulate cell apoptosis through different signaling pathways and molecular mechanisms. ACSL members have individual functions in fatty acid metabolism in different types of cells depending on substrate preferences, subcellular location and tissue specificity, thus contributing to liver diseases and metabolic diseases, such as fatty liver disease, obesity, atherosclerosis and diabetes. They are also linked to neurological disorders and other diseases. However, the mechanisms are unclear. This review addresses new findings in the classification and properties of ACSLs and the fatty acid metabolism-associated effects of ACSLs in diseases.

Keywords: Long-chain acyl-CoA synthetase, Fatty acid, Proliferation, Apoptosis, Liver diseases, Metabolic diseases, Pathways


Core tip: Recent research has shown that long-chain acyl-CoA synthetase (ACSL) family members have individual functions in fatty acid metabolism in different types of cells depending on substrate preferences, subcellular location and tissue specificity, thus contributing to several diseases. These enzymes also regulate cell proliferation and apoptosis through different mechanisms. This review addresses new findings in the fatty acid metabolism-associated effects of ACSLs in diseases.

INTRODUCTION

Fatty acids are a major source of energy in mammals. Given a sufficient oxygen supply, fatty acids can be degraded to CO2 and H2O in the body and large quantities of energy are released in the form of adenosine triphosphate (ATP), which can then be used by the organism. Exogenous or endogenous fatty acids are chemically quite inert and need to be activated to form acyl-CoA in the cell, outside the mitochondrion, before they enter a metabolic pathway. Acyl-CoA synthetase on the endoplasmic reticulum (ER) and mitochondrial outer membrane catalyzes the conversion of fatty acids to acyl-CoA in the presence of ATP, CoA and Mg2+.

The acyl-CoA synthases can be classified, according to the carbon chain length of the fatty acid they catalyze, into very long-chain acyl-CoA synthases (ACSVL), long-chain acyl-CoA synthases (ACSL), medium-chain acyl-CoA synthases (ACSM) and short-chain acyl-CoA synthases (ACSS). ACSLs mainly catalyze fatty acids with chain lengths of 12-20 carbons[1]. ACSL1 was initially reported in 1953 and ACSL3 to 6 were subsequently discovered in succession[2]. There are five different ACSL isoenzymes in mammals, and each isoenzyme has multiple spliced variants. Mouse ACSL4 variants 1 and 3 are equivalent to the human variants 2 and 1, respectively[1]. ACSL1, ACSL5 and ACSL6 (formerly ACSL2) show a 60% amino acid sequence homology. ACSL3 and ACSL4 show a 68% homology with each other[3,4]. There are many differences in the five ACSLs with regard to regulating fatty acid metabolism, promoting cell proliferation and apoptosis and causing diseases.

EFFECTS OF ACSL ON FATTY ACID METABOLISM

ACSLs are characterized by substrate and tissue specificity. The expression of ACSL family members varies in different tissues and in its subcellular location. For example, ACSL6 has been reported to be mainly expressed in neural cells and the brain, and ACSL3 protein is predominant in brain and testis tissue. However, it is undetectable in the heart[5,6]. ACSL5 is localized in the outer mitochondrial membrane and microsomes[7].

ACSL1

ACSL1 is the predominant isoform in the liver. The activity of ACSL1 accounts for 50% of total hepatic ACSL activity[8]. ACSL1 over-expression increases the proportion of oleic acid in diacylglycerol (DAG) and phospholipids (PLs), but reduces the amount of oleic acid in cholesterol[9]. ACSL1 and ACSL5 are thought to play an important role in partitioning fatty acids toward triglyceride (TG) synthesis[10]. Deficiency of ACSL1 causes reduced synthesis of TG, but facilitates β-oxidation and the synthesis of PL species in hepatic cells[8]. Over-expression of ACSL1 results in the generation of large amounts of TG and its accumulation in hepatoma cells[11]. ACSL1 is able to activate membrane transport to effectively import free fatty acids into cells[12]. Compared with the normal control group, ACSL1 protein expression was observed to increase 2.468-5.418-fold 48 h after hepatocytes were treated with various concentrations of free long-chain fatty acids. In the presence of excessive free fatty acids, hepatocytes increase their uptake of free fatty acids and deposit them in the form of TG[13].

ACSL3

ACSL3 is mainly expressed during fetal development, and the level of ACSL3 in adult cerebrum is only 10% of its maximum on 15 d after birth[14]. ACSL3 is localized in the ER and cytosolic lipid droplets (LDs)[15]. LDs are the main organelles for the storage of neutral lipids, and contribute to the maintenance of lipid homeostasis. The physiological function of ACSL3 is to promote the synthesis of lecithin and the formation of LDs, which is not seen in other isoforms of ACSL[16]. Lecithin is the main PL on the surface of very low density lipoprotein (VLDL). Secretion of VLDL is suppressed in case of ACSL3 insufficiency. ACSL3 has an influence on the secretion of VLDL through the promotion of lecithin synthesis provided by the ACSL3-mediated synthesis of DAG[17]. The N-terminal region of ACSL3 is required for fatty acid uptake, due to its effect on enzymatic activity[18]. Experimental findings have shown that ACSL3 is able to regulate lipogenesis by facilitating the gene activity of several lipogenic transcription factors, including peroxisome proliferator-activated receptor-γ (PPAR-γ), carbohydrate-responsive element-binding protein (ChREB) and sterol regulatory element-binding protein-1c (SREBP-1c)[17,19,20].

ACSL4

ACSL4 is mainly expressed in adrenal glands and other steroid producing organs. ACSL4 expression is related to steroid hormones and growth factor receptors[21]. Silencing ACSL4 can inhibit the generation of hormone-related steroid[22]. ACSL4 has a substrate preference for arachidonic acid (AA). The catalytic activity of ACSL4 for AA is 5 to 6 times higher than that for linoleic acid[4]. ACSL4 expression decreases the conversion of free AA into leukotrienes while increasing the conversion of free AA into prostaglandins[21,23]. ACSL4 over-expression also significantly increases the synthesis of eicosanoid-CoA and promotes conversion of AA into phosphatidyl ethanolamine (PE), phosphatidyl inositol (PI) and TG[23,24].

ACSL5

It is well known that the absorption of dietary long chain fatty acids largely occurs in the jejunum and ACSL5 is mainly expressed in the small intestine. This suggests that ACSL5 plays a crucial role in the absorption of dietary long-chain fatty acids. Studies on ACSL5 have shown that it can catalyze the metabolism of exogenous, but not de novo synthesized fatty acids[7,25]. ACSL5-knockout mice did not show changes in the absorption of long-chain fatty acids or weight gain after a high-fat diet[26]. ACSL5 is also expressed in the liver and brown adipose tissue characterized by high levels of TG synthetase, and is involved in the synthesis of TG. ACSL5 over-expression promotes the synthesis of DAG and TG from fatty acids, which may be attributed to accelerated re-acylation by ACSL5. In contrast, ACSL5-knockout resulted in decreased synthesis of TG[11,27]. In addition, ACSL5 is also the only ACSL isoform localized in mitochondria. It is commonly believed to be related to β-oxidation. However, some data have indicated that ACSL5 over-expression does not have an impact on the β-oxidation of fatty acids[7].

ACSL6

ACSL6 also catalyzes very long-chain fatty acids (C18 to C26) to form acyl-CoAs. Following the supplementation of polyunsaturated fatty acids to cells over-expressing ACSL6, both polyunsaturated fatty acids and saturated fatty acids increased in the cells, but to different degrees. Compared with oleic acid and AA, over-expression of ACSL6 in PC12 cells preferentially promotes docosahexaenoic acid (DHA) to form DHA-CoA and to further synthesize PLs and TGs. ACSL6 over-expression increases the level of PLs, but does not alter the distribution of fatty acids among the major PL species[28].

EFFECTS OF ACSL ON CELL PROLIFERATION

ACSLs affect the proliferation of normal cells. ACSL5 contributes to cell proliferation along the intestinal crypt-villus axis (CVA)[29]. Schoonjans et al[30] induced liver cell proliferation through partial liver resection, and ACSL mRNAs in the liver cells almost completely disappeared within 24 h after the surgery; then restored to 40% of the preoperative level 48 h later; and to 70% of the preoperative level 72 h later. These results suggest a substantial connection between ACSL and the proliferation of normal cells.

ACSL is also associated with the proliferation of malignant tumor cells. Tumor cells may over-express ACSL to utilize fatty acids as an energy source for cell proliferation. Studies have shown that ACSL4 expression is induced in MCF-7 and SKBr3 breast cancer cells and this promotes the proliferation of tumor cells[21,31]. In addition, ACSL4 is also involved in cell proliferation in liver cancer and colon cancer[32,33]. ACSL6 was found to be related to tumor cell proliferation in experiments on neuroblastoma cells and pheochromocytoma.

EFFECTS OF ACSL ON CELL APOPTOSIS

ACSLs not only affect cell proliferation, but also play a role in cell apoptosis. Studies have shown that ACSL6 expression is elevated in animal models of non-alcoholic fatty liver disease (NAFLD), which may promote liver cell apoptosis[34]. In apoptotic intestinal cells, ACSL5 down-regulates cellular Fas associated death domain-like interleukin-1β converting enzyme inhibitory protein (cFLIP), which has been proven to be anti-apoptotic, and up-regulates tumor necrosis factor-related apoptosis inducing ligand receptor 1 (TRAIL-R1), which is the membrane receptor of the tumor necrosis factor/c-Jun N-terminal kinase (TNF/JNK) apoptosis pathway. A study has also shown that ACSL5 induces synthesis of ceramide, an important second messenger of the apoptosis pathway[35] (Figure 1). A study on an animal model of systemic lupus erythematosus showed that transcription of ACSL5 was significantly elevated in the disease model compared with normal controls and silencing ACSL5 with siRNA reduced apoptosis[36].

Figure 1.

Figure 1

Long-chain acyl-CoA synthetases induce apoptotic cell death via the c-Jun N-terminal kinase pathway. JNK can be activated via phosphorylation by increased ceramide which can be induced by ACSLs that subsequently activate caspase 3, leading to apoptotic cell death. This shows that ACSLs may induce apoptotic cell death via the JNK pathway. In addition, anti-apoptotic proteins such as cFLIP, which is downregulated by ACSLs, may inhibit the activation of JNK. ACSL: Long-chain acyl-CoA synthetase; cFLIP: Cellular Fas associated death domain-like interleukin-1β converting enzyme inhibitory protein; JNK: c-Jun N-terminal kinase; TRAIL: Tumor necrosis factor-related apoptosis inducing ligand; TRAIL-R1: Tumor necrosis factor-related apoptosis inducing ligand receptor 1.

Different fatty acids have different effects on cell apoptosis. Saturated fatty acids such as palmitic acid and stearic acid can activate caspase 3 (Figure 1), which leads to cell apoptosis, while unsaturated fatty acids have little effect on cell apoptosis. Studies have shown that ACSL1 over-expression can further enhance palmitoyl lipid-induced apoptosis, while treatment with triacsin C, an ACSL inhibitor, reverses the pro-apoptotic effect of saturated fatty acids[37].

ACSL AND LIVER DISEASES

With the exception of ACSL6, all ACSL isoforms are expressed in the liver and the major subtype of ACSL in the liver is ACSL1, which is also a target gene of peroxisome proliferator-activated receptor-α (PPAR-α). PPAR-α is involved in fatty acid metabolism[38]. Deficient or repressed PPAR-α expression in the liver leads to decreased activity of ACSL1 and some other key enzymes involved in fatty acid metabolism, and consequently fat deposition and inflammation which promote liver fibrosis[39]. Another subtype of ACSL, ACSL3, plays an important role in the pathogenesis of fatty liver in addition to ACSL5[25,40]. Silencing ACSL3 inhibits the release of HCV particles from infected liver cells into plasma. HCV proliferation in liver cells can cause structural and functional changes or interfere with protein synthesis, and ultimately degeneration and necrosis of liver cells[17]. ACSL4 is upregulated in patients with NAFLD who have undergone bariatric surgery[41]. The regulation of ACSL4 may be through both the 3’-5’-cyclic adenosine monophosphate (cAMP) and p38 mitogen-activated protein kinase (MAPK) pathways in liver cancer[32] (Table 1).

Table 1.

Long-chain acyl-CoA synthetase expression and pathways involved in liver diseases

Liver diseases mRNA expression Pathways Ref.
ACSL1 HF PPAR/NF-κB/p65 Xin et al[39] and Pyper et al[42]
NAFLD TGF Uto et al[43]
ACSL3 NAFLD LXR/RXR Dong et al[44]
ACSL4 HCV -- Yao et al[17]
HCC P38/cAMP Liang et al[32]
NAFLD -- Stepanova et al[41]
ACSL5 NAFLD Caspase Reinartz et al[40]

ACSL: Long-chain acyl-CoA synthetase; HF: Hepatic fibrosis; NAFLD: Non-alcoholic fatty liver disease; HCV: Hepatitis C virus; HCC: Hepatocellular carcinoma; PPAR: Peroxisome proliferator-activated receptor; NF-κB: Nuclear factor of kappa light polypeptide gene enhancer in B-cells; TGF: Transforming growth factor; LXR: Liver X receptor; RXR: Retinoid X receptor; cAMP: 3’-5’-cyclic adenosine monophosphate.

ACSL AND METABOLIC DISEASES

Fatty acid metabolism disorder is involved in the occurrence of many metabolic diseases, such as obesity, diabetes, cardiovascular disease and atherosclerosis. ACSLs play an important role in fatty acid metabolism, and dysfunction of these enzymes often leads to fatty acid metabolism disorders. The rs9997745 mutant of ACSL1 increases the risk of metabolic syndrome, related to type 2 diabetes[45,46]. In addition, ACSL1 expression is increased in mononuclear cells in type 1 diabetic patients[47], suggesting that ACSL1 may be involved in the pathogenesis of diabetes.

ACSL AND NERVOUS SYSTEM DISEASES

Abnormal lipid metabolism can also cause neurological disease. ACSL6 predominates in nerve cells and ACSL6 insufficiency can lead to neuronal degeneration[48]. Studies have shown that ACSL6 over-expression leads to neurite outgrowth in rats, while silencing ACSL6 inhibits axon outgrowth of mouse neural cells[28,48]. ACSL6-induced activation of acetylcholinesterase may be involved in this process, as acetylcholinesterase promotes neural differentiation[49]. ACSL4 has been reported to be correlated with X-linked mental retardation, which leads to a higher incidence of mental disorders[50]. Recent studies have shown that, like ACSL6, ACSL4 showed specificity for polyunsaturated fatty acids, such as AA and eicosapentaenoic acid, which may contribute to changes in function of specific tissue[4,28].

ACSL AND OTHER DISEASES

Studies have shown that ACSLs also play a role in other diseases. Methylation of ACSL3 5’-CpG island (CGI) is thought to be correlated with maternal exposure to polycyclic aromatic hydrocarbons in the air, which is also closely correlated with the increased incidence of asthma, suggesting that ACSL3 may play a role in asthma[51,52]. Studies have shown that ACSL4 stimulates the release of prostaglandin E2 by arterial smooth muscle cells, which promotes atherosclerosis formation, suggesting the involvement of ACSL4 in atherosclerosis[23,53,54]. ACSL4 has also been linked to uterus abnormalities, Alport syndrome and elliptocytosis[55,56]. ACSL5 transcription is elevated in systemic lupus erythematosus (SLE), and the treatment of SLE with corticosteroids decreased ACSL5 transcription[36]. Patients with inflammatory bowel disease showed over-expression of ACSL1 and ACSL5 mRNAs in the terminal ileum and colon[57].

CONCLUSION

Long-chain Acyl-CoA synthetase plays a crucial role in fatty acid metabolism. Fatty acids, saturated or unsaturated, are a major source of energy in humans and are essential, particularly unsaturated fatty acids. Fatty acids need to be activated to form acyl-CoA before they enter a metabolic pathway, including both anabolic and catabolic pathways. Recent research on ACSLs mainly focused on the effect on fatty acid metabolism and less on the influence on cell proliferation and apoptosis. The mechanisms involved in fatty acid metabolism and cell proliferation and apoptosis are unclear. Further studies to elucidate the function of ACSL enzymes would be highly beneficial.

Footnotes

Supported by National Natural Science Foundation of China, No. 81373465.

Conflict-of-interest: We declare that we have no financial and personal relationships with other people or organizations that can inappropriately influence our work. There is no professional or other personal interest of any nature or kind in any product, service and/or company that could be construed as influencing the position presented in, or the review of, the manuscript entitled.

Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/

Peer-review started: September 30, 2014

First decision: October 29, 2014

Article in press: January 21, 2015

P- Reviewer: Crenn PP, Matthews V S- Editor: Ma YJ L- Editor: Wang TQ E- Editor: Zhang DN

References

  • 1.Soupene E, Kuypers FA. Mammalian long-chain acyl-CoA synthetases. Exp Biol Med (Maywood) 2008;233:507–521. doi: 10.3181/0710-MR-287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Suzuki H, Kawarabayasi Y, Kondo J, Abe T, Nishikawa K, Kimura S, Hashimoto T, Yamamoto T. Structure and regulation of rat long-chain acyl-CoA synthetase. J Biol Chem. 1990;265:8681–8685. [PubMed] [Google Scholar]
  • 3.Oikawa E, Iijima H, Suzuki T, Sasano H, Sato H, Kamataki A, Nagura H, Kang MJ, Fujino T, Suzuki H, et al. A novel acyl-CoA synthetase, ACS5, expressed in intestinal epithelial cells and proliferating preadipocytes. J Biochem. 1998;124:679–685. doi: 10.1093/oxfordjournals.jbchem.a022165. [DOI] [PubMed] [Google Scholar]
  • 4.Kang MJ, Fujino T, Sasano H, Minekura H, Yabuki N, Nagura H, Iijima H, Yamamoto TT. A novel arachidonate-preferring acyl-CoA synthetase is present in steroidogenic cells of the rat adrenal, ovary, and testis. Proc Natl Acad Sci USA. 1997;94:2880–2884. doi: 10.1073/pnas.94.7.2880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Fujino T, Yamamoto T. Cloning and functional expression of a novel long-chain acyl-CoA synthetase expressed in brain. J Biochem. 1992;111:197–203. doi: 10.1093/oxfordjournals.jbchem.a123737. [DOI] [PubMed] [Google Scholar]
  • 6.Wu M, Liu H, Chen W, Fujimoto Y, Liu J. Hepatic expression of long-chain acyl-CoA synthetase 3 is upregulated in hyperlipidemic hamsters. Lipids. 2009;44:989–998. doi: 10.1007/s11745-009-3341-3. [DOI] [PubMed] [Google Scholar]
  • 7.Mashek DG, McKenzie MA, Van Horn CG, Coleman RA. Rat long chain acyl-CoA synthetase 5 increases fatty acid uptake and partitioning to cellular triacylglycerol in McArdle-RH7777 cells. J Biol Chem. 2006;281:945–950. doi: 10.1074/jbc.M507646200. [DOI] [PubMed] [Google Scholar]
  • 8.Li LO, Ellis JM, Paich HA, Wang S, Gong N, Altshuller G, Thresher RJ, Koves TR, Watkins SM, Muoio DM, et al. Liver-specific loss of long chain acyl-CoA synthetase-1 decreases triacylglycerol synthesis and beta-oxidation and alters phospholipid fatty acid composition. J Biol Chem. 2009;284:27816–27826. doi: 10.1074/jbc.M109.022467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Li LO, Mashek DG, An J, Doughman SD, Newgard CB, Coleman RA. Overexpression of rat long chain acyl-coa synthetase 1 alters fatty acid metabolism in rat primary hepatocytes. J Biol Chem. 2006;281:37246–37255. doi: 10.1074/jbc.M604427200. [DOI] [PubMed] [Google Scholar]
  • 10.Ellis JM, Li LO, Wu PC, Koves TR, Ilkayeva O, Stevens RD, Watkins SM, Muoio DM, Coleman RA. Adipose acyl-CoA synthetase-1 directs fatty acids toward beta-oxidation and is required for cold thermogenesis. Cell Metab. 2010;12:53–64. doi: 10.1016/j.cmet.2010.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Parkes HA, Preston E, Wilks D, Ballesteros M, Carpenter L, Wood L, Kraegen EW, Furler SM, Cooney GJ. Overexpression of acyl-CoA synthetase-1 increases lipid deposition in hepatic (HepG2) cells and rodent liver in vivo. Am J Physiol Endocrinol Metab. 2006;291:E737–E744. doi: 10.1152/ajpendo.00112.2006. [DOI] [PubMed] [Google Scholar]
  • 12.Wang Z, Yang YX, Wang GD, Bian LH, Yang JM, Men JH, Yao H, Li Y, Lu Ying. Starch digestion and glycemic indexes. Weisheng Yanjiu. 2003;32:622–624. [PubMed] [Google Scholar]
  • 13.Liu Y, Shi WR, Hong ZF, Zheng HY, Li Y. Effect of free fatty acids on long-chain acyl-CoA synthetase 1 expression level and lipid metabolism in liver cells. Yingyang Xuebao. 2013;35:232–235, 240. [Google Scholar]
  • 14.Fujino T, Kang MJ, Suzuki H, Iijima H, Yamamoto T. Molecular characterization and expression of rat acyl-CoA synthetase 3. J Biol Chem. 1996;271:16748–16752. doi: 10.1074/jbc.271.28.16748. [DOI] [PubMed] [Google Scholar]
  • 15.Murphy DJ. The biogenesis and functions of lipid bodies in animals, plants and microorganisms. Prog Lipid Res. 2001;40:325–438. doi: 10.1016/s0163-7827(01)00013-3. [DOI] [PubMed] [Google Scholar]
  • 16.Fujimoto Y, Itabe H, Kinoshita T, Homma KJ, Onoduka J, Mori M, Yamaguchi S, Makita M, Higashi Y, Yamashita A, et al. Involvement of ACSL in local synthesis of neutral lipids in cytoplasmic lipid droplets in human hepatocyte HuH7. J Lipid Res. 2007;48:1280–1292. doi: 10.1194/jlr.M700050-JLR200. [DOI] [PubMed] [Google Scholar]
  • 17.Yao H, Ye J. Long chain acyl-CoA synthetase 3-mediated phosphatidylcholine synthesis is required for assembly of very low density lipoproteins in human hepatoma Huh7 cells. J Biol Chem. 2008;283:849–854. doi: 10.1074/jbc.M706160200. [DOI] [PubMed] [Google Scholar]
  • 18.Poppelreuther M, Rudolph B, Du C, Großmann R, Becker M, Thiele C, Ehehalt R, Füllekrug J. The N-terminal region of acyl-CoA synthetase 3 is essential for both the localization on lipid droplets and the function in fatty acid uptake. J Lipid Res. 2012;53:888–900. doi: 10.1194/jlr.M024562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Schroeder F, Petrescu AD, Huang H, Atshaves BP, McIntosh AL, Martin GG, Hostetler HA, Vespa A, Landrock D, Landrock KK, et al. Role of fatty acid binding proteins and long chain fatty acids in modulating nuclear receptors and gene transcription. Lipids. 2008;43:1–17. doi: 10.1007/s11745-007-3111-z. [DOI] [PubMed] [Google Scholar]
  • 20.Mashek DG, Li LO, Coleman RA. Long-chain acyl-CoA synthetases and fatty acid channeling. Future Lipidol. 2007;2:465–476. doi: 10.2217/17460875.2.4.465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wu X, Li Y, Wang J, Wen X, Marcus MT, Daniels G, Zhang DY, Ye F, Wang LH, Du X, et al. Long chain fatty Acyl-CoA synthetase 4 is a biomarker for and mediator of hormone resistance in human breast cancer. PLoS One. 2013;8:e77060. doi: 10.1371/journal.pone.0077060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Maloberti P, Castilla R, Castillo F, Cornejo Maciel F, Mendez CF, Paz C, Podestá EJ. Silencing the expression of mitochondrial acyl-CoA thioesterase I and acyl-CoA synthetase 4 inhibits hormone-induced steroidogenesis. FEBS J. 2005;272:1804–1814. doi: 10.1111/j.1742-4658.2005.04616.x. [DOI] [PubMed] [Google Scholar]
  • 23.Golej DL, Askari B, Kramer F, Barnhart S, Vivekanandan-Giri A, Pennathur S, Bornfeldt KE. Long-chain acyl-CoA synthetase 4 modulates prostaglandin E2 release from human arterial smooth muscle cells. J Lipid Res. 2011;52:782–793. doi: 10.1194/jlr.M013292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cooke M, Orlando U, Maloberti P, Podestá EJ, Cornejo Maciel F. Tyrosine phosphatase SHP2 regulates the expression of acyl-CoA synthetase ACSL4. J Lipid Res. 2011;52:1936–1948. doi: 10.1194/jlr.M015552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bu SY, Mashek MT, Mashek DG. Suppression of long chain acyl-CoA synthetase 3 decreases hepatic de novo fatty acid synthesis through decreased transcriptional activity. J Biol Chem. 2009;284:30474–30483. doi: 10.1074/jbc.M109.036665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Meller N, Morgan ME, Wong WP, Altemus JB, Sehayek E. Targeting of Acyl-CoA synthetase 5 decreases jejunal fatty acid activation with no effect on dietary long-chain fatty acid absorption. Lipids Health Dis. 2013;12:88. doi: 10.1186/1476-511X-12-88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bu SY, Mashek DG. Hepatic long-chain acyl-CoA synthetase 5 mediates fatty acid channeling between anabolic and catabolic pathways. J Lipid Res. 2010;51:3270–3280. doi: 10.1194/jlr.M009407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Marszalek JR, Kitidis C, Dirusso CC, Lodish HF. Long-chain acyl-CoA synthetase 6 preferentially promotes DHA metabolism. J Biol Chem. 2005;280:10817–10826. doi: 10.1074/jbc.M411750200. [DOI] [PubMed] [Google Scholar]
  • 29.Klaus C, Jeon MK, Kaemmerer E, Gassler N. Intestinal acyl-CoA synthetase 5: activation of long chain fatty acids and behind. World J Gastroenterol. 2013;19:7369–7373. doi: 10.3748/wjg.v19.i42.7369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Schoonjans K, Staels B, Grimaldi P, Auwerx J. Acyl-CoA synthetase mRNA expression is controlled by fibric-acid derivatives, feeding and liver proliferation. Eur J Biochem. 1993;216:615–622. doi: 10.1111/j.1432-1033.1993.tb18181.x. [DOI] [PubMed] [Google Scholar]
  • 31.Monaco ME, Creighton CJ, Lee P, Zou X, Topham MK, Stafforini DM. Expression of Long-chain Fatty Acyl-CoA Synthetase 4 in Breast and Prostate Cancers Is Associated with Sex Steroid Hormone Receptor Negativity. Transl Oncol. 2010;3:91–98. doi: 10.1593/tlo.09202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Liang YC, Wu CH, Chu JS, Wang CK, Hung LF, Wang YJ, Ho YS, Chang JG, Lin SY. Involvement of fatty acid-CoA ligase 4 in hepatocellular carcinoma growth: roles of cyclic AMP and p38 mitogen-activated protein kinase. World J Gastroenterol. 2005;11:2557–2563. doi: 10.3748/wjg.v11.i17.2557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Cao Y, Dave KB, Doan TP, Prescott SM. Fatty acid CoA ligase 4 is up-regulated in colon adenocarcinoma. Cancer Res. 2001;61:8429–8434. [PubMed] [Google Scholar]
  • 34.Xu C, Wang G, Hao Y, Zhi J, Zhang L, Chang C. Correlation analysis between gene expression profile of rat liver tissues and high-fat emulsion-induced nonalcoholic fatty liver. Dig Dis Sci. 2011;56:2299–2308. doi: 10.1007/s10620-011-1599-9. [DOI] [PubMed] [Google Scholar]
  • 35.Gassler N, Roth W, Funke B, Schneider A, Herzog F, Tischendorf JJ, Grund K, Penzel R, Bravo IG, Mariadason J, et al. Regulation of enterocyte apoptosis by acyl-CoA synthetase 5 splicing. Gastroenterology. 2007;133:587–598. doi: 10.1053/j.gastro.2007.06.005. [DOI] [PubMed] [Google Scholar]
  • 36.Catalá-Rabasa A, Ndagire D, Sabio JM, Fedetz M, Matesanz F, Alcina A. High ACSL5 transcript levels associate with systemic lupus erythematosus and apoptosis in Jurkat T lymphocytes and peripheral blood cells. PLoS One. 2011;6:e28591. doi: 10.1371/journal.pone.0028591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Li X, Gonzalez O, Shen X, Barnhart S, Kramer F, Kanter JE, Vivekanandan-Giri A, Tsuchiya K, Handa P, Pennathur S, et al. Endothelial acyl-CoA synthetase 1 is not required for inflammatory and apoptotic effects of a saturated fatty acid-rich environment. Arterioscler Thromb Vasc Biol. 2013;33:232–240. doi: 10.1161/ATVBAHA.112.252239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Filip-Ciubotaru F, Foia L, Manciuc C, Grigore C. [PPARs: structure, mechanisms of action and control. Note I] Rev Med Chir Soc Med Nat Iasi. 2011;115:477–484. [PubMed] [Google Scholar]
  • 39.Xin X, Yan HZ, Li WQ, Yu HY. Studies on PPAR α and γ participating in progression of liver fibrosis by regulating ACSL1. Linchuang Gandanbing Zazhi. 2014;30:700–702. [Google Scholar]
  • 40.Reinartz A, Ehling J, Leue A, Liedtke C, Schneider U, Kopitz J, Weiss T, Hellerbrand C, Weiskirchen R, Knüchel R, et al. Lipid-induced up-regulation of human acyl-CoA synthetase 5 promotes hepatocellular apoptosis. Biochim Biophys Acta. 2010;1801:1025–1035. doi: 10.1016/j.bbalip.2010.04.010. [DOI] [PubMed] [Google Scholar]
  • 41.Stepanova M, Hossain N, Afendy A, Perry K, Goodman ZD, Baranova A, Younossi Z. Hepatic gene expression of Caucasian and African-American patients with obesity-related non-alcoholic fatty liver disease. Obes Surg. 2010;20:640–650. doi: 10.1007/s11695-010-0078-2. [DOI] [PubMed] [Google Scholar]
  • 42.Pyper SR, Viswakarma N, Yu S, Reddy JK. PPARalpha: energy combustion, hypolipidemia, inflammation and cancer. Nucl Recept Signal. 2010;8:e002. doi: 10.1621/nrs.08002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Uto H, Nakanishi C, Ido A, Hasuike S, Kusumoto K, Abe H, Numata M, Nagata K, Hayashi K, Tsubouchi H. The peroxisome proliferator-activated receptor-gamma agonist, pioglitazone, inhibits fat accumulation and fibrosis in the livers of rats fed a choline-deficient, l-amino acid-defined diet. Hepatol Res. 2005;32:235–242. doi: 10.1016/j.hepres.2005.05.008. [DOI] [PubMed] [Google Scholar]
  • 44.Dong B, Kan CF, Singh AB, Liu J. High-fructose diet downregulates long-chain acyl-CoA synthetase 3 expression in liver of hamsters via impairing LXR/RXR signaling pathway. J Lipid Res. 2013;54:1241–1254. doi: 10.1194/jlr.M032599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Phillips CM, Goumidi L, Bertrais S, Field MR, Cupples LA, Ordovas JM, Defoort C, Lovegrove JA, Drevon CA, Gibney MJ, et al. Gene-nutrient interactions with dietary fat modulate the association between genetic variation of the ACSL1 gene and metabolic syndrome. J Lipid Res. 2010;51:1793–1800. doi: 10.1194/jlr.M003046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Moller DE, Kaufman KD. Metabolic syndrome: a clinical and molecular perspective. Annu Rev Med. 2005;56:45–62. doi: 10.1146/annurev.med.56.082103.104751. [DOI] [PubMed] [Google Scholar]
  • 47.Kanter JE, Kramer F, Barnhart S, Averill MM, Vivekanandan-Giri A, Vickery T, Li LO, Becker L, Yuan W, Chait A, et al. Diabetes promotes an inflammatory macrophage phenotype and atherosclerosis through acyl-CoA synthetase 1. Proc Natl Acad Sci USA. 2012;109:E715–E724. doi: 10.1073/pnas.1111600109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kim HC, Lee SW, Cho YY, Lim JM, Ryoo ZY, Lee EJ. RNA interference of long-chain acyl-CoA synthetase 6 suppresses the neurite outgrowth of mouse neuroblastoma NB41A3 cells. Mol Med Rep. 2009;2:669–674. doi: 10.3892/mmr_00000155. [DOI] [PubMed] [Google Scholar]
  • 49.Soreq H, Seidman S. Acetylcholinesterase--new roles for an old actor. Nat Rev Neurosci. 2001;2:294–302. doi: 10.1038/35067589. [DOI] [PubMed] [Google Scholar]
  • 50.Meloni I, Parri V, De Filippis R, Ariani F, Artuso R, Bruttini M, Katzaki E, Longo I, Mari F, Bellan C, et al. The XLMR gene ACSL4 plays a role in dendritic spine architecture. Neuroscience. 2009;159:657–669. doi: 10.1016/j.neuroscience.2008.11.056. [DOI] [PubMed] [Google Scholar]
  • 51.Perera F, Tang WY, Herbstman J, Tang D, Levin L, Miller R, Ho SM. Relation of DNA methylation of 5’-CpG island of ACSL3 to transplacental exposure to airborne polycyclic aromatic hydrocarbons and childhood asthma. PLoS One. 2009;4:e4488. doi: 10.1371/journal.pone.0004488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yuan XZ, Jin H, Bao YB, Piao MY. Advance of research on polycyclic aromatic hydrocarbons associated with asthma. Yanbing Daxue Yixue Zazhi. 2012;35:70–74. [Google Scholar]
  • 53.Wang M, Song WL, Cheng Y, Fitzgerald GA. Microsomal prostaglandin E synthase-1 inhibition in cardiovascular inflammatory disease. J Intern Med. 2008;263:500–505. doi: 10.1111/j.1365-2796.2008.01938.x. [DOI] [PubMed] [Google Scholar]
  • 54.Mandelbaum M, Kolega J, Dolan JM, Siddiqui AH, Meng H. A critical role for proinflammatory behavior of smooth muscle cells in hemodynamic initiation of intracranial aneurysm. PLoS One. 2013;8:e74357. doi: 10.1371/journal.pone.0074357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cho YY, Kang MJ, Sone H, Suzuki T, Abe M, Igarashi M, Tokunaga T, Ogawa S, Takei YA, Miyazawa T, et al. Abnormal uterus with polycysts, accumulation of uterine prostaglandins, and reduced fertility in mice heterozygous for acyl-CoA synthetase 4 deficiency. Biochem Biophys Res Commun. 2001;284:993–997. doi: 10.1006/bbrc.2001.5065. [DOI] [PubMed] [Google Scholar]
  • 56.Piccini M, Vitelli F, Bruttini M, Pober BR, Jonsson JJ, Villanova M, Zollo M, Borsani G, Ballabio A, Renieri A. FACL4, a new gene encoding long-chain acyl-CoA synthetase 4, is deleted in a family with Alport syndrome, elliptocytosis, and mental retardation. Genomics. 1998;47:350–358. doi: 10.1006/geno.1997.5104. [DOI] [PubMed] [Google Scholar]
  • 57.Heimerl S, Moehle C, Zahn A, Boettcher A, Stremmel W, Langmann T, Schmitz G. Alterations in intestinal fatty acid metabolism in inflammatory bowel disease. Biochim Biophys Acta. 2006;1762:341–350. doi: 10.1016/j.bbadis.2005.12.006. [DOI] [PubMed] [Google Scholar]

Articles from World Journal of Gastroenterology : WJG are provided here courtesy of Baishideng Publishing Group Inc

RESOURCES