Next Article in Journal
“Fuel for the Damage Induced”: Untargeted Metabolomics in Elite Rugby Union Match Play
Next Article in Special Issue
Identification of a Variant in APOB Gene as a Major Cause of Hypobetalipoproteinemia in Lebanese Families
Previous Article in Journal
A Mediation Approach to Discovering Causal Relationships between the Metabolome and DNA Methylation in Type 1 Diabetes
Previous Article in Special Issue
Hepatocyte Small Heterodimer Partner Mediates Sex-Specific Effects on Triglyceride Metabolism via Androgen Receptor in Male Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Acyl-Coenzyme A: Cholesterol Acyltransferase (ACAT) in Cholesterol Metabolism: From Its Discovery to Clinical Trials and the Genomics Era

Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH 44195, USA
*
Author to whom correspondence should be addressed.
Metabolites 2021, 11(8), 543; https://doi.org/10.3390/metabo11080543
Submission received: 30 July 2021 / Revised: 10 August 2021 / Accepted: 12 August 2021 / Published: 14 August 2021

Abstract

:
The purification and cloning of the acyl-coenzyme A: cholesterol acyltransferase (ACAT) enzymes and the sterol O-acyltransferase (SOAT) genes has opened new areas of interest in cholesterol metabolism given their profound effects on foam cell biology and intestinal lipid absorption. The generation of mouse models deficient in Soat1 or Soat2 confirmed the importance of their gene products on cholesterol esterification and lipoprotein physiology. Although these studies supported clinical trials which used non-selective ACAT inhibitors, these trials did not report benefits, and one showed an increased risk. Early genetic studies have implicated common variants in both genes with human traits, including lipoprotein levels, coronary artery disease, and Alzheimer’s disease; however, modern genome-wide association studies have not replicated these associations. In contrast, the common SOAT1 variants are most reproducibly associated with testosterone levels.

Graphical Abstract

1. Introduction

The acyl-coenzyme A:cholesterol acyltransferase (ACAT; EC 2.3.1.26) enzyme family consists of membrane-spanning proteins, which are primarily located in the endoplasmic reticulum [1]. These enzymes act to esterify the 3-hydroxyl position of cellular free cholesterol (FC) with a fatty acid-CoA creating cholesteryl ester (CE), thus playing an important role in cholesterol homeostasis [2]. A different enzyme, lecithin cholesterol acyltransferase (LCAT), can esterify plasma FC within HDL, which is reviewed elsewhere [3]. In mammals, the ACAT family contains two isoenzymes, ACAT1 and ACAT2 [4,5,6]. ACAT1/2 are members of the membrane-bound O-acyltransferase (MBOAT) family, which includes the closely related acyl-CoA:diacylglycerol acyltransferase-1 (DGAT1) [7]. MBOAT proteins are characterized by their multiple transmembrane domains and a catalytic histidine residue in a hydrophobic domain [8].
We first deal with the confusing nomenclature of the ACAT1/2 enzymes, which are alternatively termed sterol O-acyltransferases (SOAT) and then lead to the human gene names SOAT1 (ensemble identifier ENSG00000057252) and SOAT2 (ENSG00000167780), whereas the mouse genes are called Soat1 and Soat2 [9,10]. The human ACAT1 (ENSG00000075239) and ACAT2 (ENSG0000012043) genes encode cytosolic enzymes with acetyl-CoA acetyltransferase/thiolase activities, which are independent from cholesterol metabolism. However, older studies may refer to the SOAT genes as ACAT genes, hampering a search of the literature. For the remainder of this article, we will refer to ACAT1/2 proteins and SOAT1/2 genes. Given that ACAT1 is expressed more widely and robustly than ACAT2, here we will focus on ACAT1/SOAT1.

2. The Structure of ACAT1

The ACAT enzymes are large transmembrane proteins that are difficult to purify with retained activity. Therefore, purification of the ACAT enzymes is dependent on elegant cellular, molecular, and biochemical studies performed in the lab of T.Y. Chang starting in 1988, as described in his review paper [1]. Briefly, mutant Chinese hamster ovary (CHO) cells were selected that were deficient in ACAT activity and susceptible to free-cholesterol-mediated cell death [2]. These cells were transfected with human genomic DNA to isolate a clonal cell population with restored ACAT activity [11]. A cDNA library prepared from the ACAT-restored CHO line was used to make a new ACAT-restored CHO cell line leading to the molecular cloning of a human SOAT1 cDNA, the 4.0 kb “K1 cDNA” with a 1.7 kb open reading frame [12]. Using fluorescence in situ hybridization and Southern blot analyses of human/hamster somatic cell hybrid panels, the SOAT1 gene was mapped to the human chromosome 1q25 [13]. Then, Chang’s lab prepared an expression plasmid adding a his-tag at the N-terminus of the SOAT1 cDNA, and additionally made monoclonal antibodies to another recombinant ACAT1 fragment [14]. The his-tagged ACAT1 was stably transfected into ACAT-deficient CHO cells and an optimized cell-free assay for ACAT activity was developed, leading to the initial purification of recombinant ACAT1 whose CE product is formed at increasing FC concentration in a sigmoidal manner, indicating the potential for catalytic and allosteric interactions with FC [14]. Among several other sterols, FC was the most potent substrate and activator of ACAT1 [15]. The steroid hormone precursor pregnenolone, made in the mitochondria, is also an ACAT1 substrate, but it cannot activate cholesterol esterification, implying that pregnenolone only binds to the catalytic site and not the allosteric site, whereas FC binds to both sites and can activate pregnenolone esterification [16]. Using recombinant ACAT1, oleoyl-CoA (18:1) vs. stearoyl-CoA (18:0) is the preferred substrate for ACAT1, leading to augmented CE production and demonstrating specific and saturable binding to ACAT1 [17]. Among unsaturated fatty acids, the ACAT1 preferentially uses oleoyl-CoA (18:1) vs. linolenoyl CoA (18:3), arachidonyl CoA (20:4), and eicosapentaenoyl CoA 20:5 [18]. The cholesterol vs. sitosterol substrate specificity of ACAT1 was compared to ACAT2 and LCAT. ACAT2 was highly selective for cholesterol, whereas ACAT1 and LCAT only slightly preferred cholesterol vs. sitosterol [19].
ACAT1 consists of two homodimers (dimer of dimers). ACAT1 contains nine transmembrane domains (TMD) and various site-directed mutagenesis studies have identified the catalytic His460 residue involved in FC binding in TMD7, and a portion of the N-terminal domain and TMD8 as being important for subunit interactions [1]. The cryo-electron microscopy structure of human ACAT1 was recently described, confirming the dimer of dimers structure and revealing three intra-ER helixes and one extra-ER helix in addition to the nine TMD-helixes [20]. Oleoyl-CoA gains entry via the C-tunnel in the structure and was found in the active site of the purified enzyme; by altering two residues in the C-tunnel, the enzyme activity was lost and the oleoyl-CoA level in the enzyme was decreased [20]. The structure demonstrates why oleoyl-CoA that bends at the Δ9 double bond fits better than nonsaturated or other unsaturated fatty acyl-CoAs; amino acid substitutions in the vicinity of the oleoyl tail also deceased activity of the enzyme [20]. The T-tunnel that converges with the C-tunnel in the active site may be used for the entry of FC [20].

3. The Role of ACAT in Lipid Metabolism and Atherosclerosis Derived from Knockout Mouse Studies

Both ACAT1/2 can convert cellular FC and free fatty acid to CE, which is an inert form that can be stored in lipid droplets. ACAT1 is expressed in many tissues, including macrophages and adrenal glands, and macrophage ACAT1 was thought to play a role in the formation of foam cells in atherosclerotic plaques [21]. Farese and colleagues disrupted the mouse Soat1 gene by homologous recombination, which resulted in decreased cholesterol esterification in the Soat1−/− embryonic fibroblasts and adrenal lysates, and markedly reduced CE levels in the adrenal glands and in peritoneal macrophages incubated with acetylated LDL. In contrast, the livers of Soat1−/− mice contained substantial amounts of CE and exhibited no reduction in cholesterol esterification activity. The tissue-specific reductions in cholesterol esterification provided strong evidence that the mammalian intracellular CE formation involves more than one ACAT enzyme, promoting the discovery of ACAT2/SOAT2. Soat1−/− mice also had higher plasma total cholesterol (TC) and high-density lipoprotein cholesterol (HDL-C) vs. controls when mice were fed either a chow or high-fat diet [21]. These global Soat1−/− mice were also characterized to have leukocytosis and an increase in bone marrow cell proliferation [22]. Later, ACAT2 was cloned and knockout of Soat2 in mice resulted in reduced CE synthesis in the small intestine and liver, which, in turn, limited intestinal cholesterol absorption, hepatic cholesterol gallstone formation, and the accumulation of CE in plasma lipoproteins [23,24].
The role of Soat1 in atherosclerosis is controversial. Farese, along with collaborators Linton and Fazio, bred Soat1−/− mice to Ldlr−/− and Apoe−/− mouse models of hyperlipidemia. On the Apoe−/− background, Soat1 deficiency led to skin xanthomas even on a chow diet; on a Western-type diet, plasma total cholesterol was markedly reduced in the Soat1−/− mice [25]. Skin xanthomas and decreased plasma cholesterol were also observed in the Ldlr−/− with Soat1-deficient mice upon feeding of a synthetic diet with 1.25% cholesterol and 0.5% cholate. These skin xanthomas were attributed to a macrophage Soat1 deficiency via bone marrow transplantation (BMT) studies [25]. To determine if the macrophage Soat1 alters atherosclerosis without an effect on plasma cholesterol, Fazio and Linton performed BMT from WT and Soat1−/− donor mice into Ldlr−/− host mice, and found larger aortic root and surface lesions in the mice receiving the Soat1−/− marrow [26]. In contrast, Ishibashi and colleagues found smaller aortic root lesions with less CE content in Soat1−/− vs. Soat1+/+ mice bred onto the Apoe- or Ldlr-deficient backgrounds, despite observing a lower plasma cholesterol only in the Western-type diet fed Apoe−/− mice, and not in the Ldlr−/− mice fed a diet with 1.25% cholesterol and 0.5% cholate [27].
Chang and colleagues bred Soat1 floxed mice and crossed them with LysM-Cre mice to generate myeloid-specific Soat1-deficient mice (SoatM/−M), which did not have leukocytosis as seen in the global KO mice [28]. Upon crossing these mice with Apoe−/− mice, they found significantly decreased aortic and aortic root lesions vs. Apoe−/− control mice accompanied by decreased lesion macrophages, macrophage recruitment, and macrophage β1-integrin; however, these mice developed larger skin xanthomas [28]. These findings were extended to advanced lesions, where decreased cholesterol clefts were seen in the Soat1−M/−MApoe−/− mice [29].
Farese and colleagues then examined the contribution of Soat2-derived CE to atherosclerosis by crossing Soat2−/− mice with Apoe-deficient mice [30]. Soat2−/− Apoe−/− mice and Soat2+/+ Apoe−/− control mice showed a similar increase in plasma apoB and total plasma lipids; however, the lipid content of the apoB-containing lipoproteins in the double-null mice contained primarily triglycerides rather than CE. At 30 weeks of age, the Soat2−/− Apoe−/− mice fed with a chow diet had smaller atherosclerosis lesions in all aortic regions (arch, thorax, and abdomen) than the Soat2+/+ Apoe−/− controls. These investigators surmised that the reason for smaller lesions in the Soat2−/− Apoe−/− mice was due to high triglycerides but low CE levels in plasma. These findings highlight the crucial role of Soat2-mediated effects on plasma CE and atherosclerosis in mice, supporting the therapeutic potential for SOAT2 inhibition.

4. ACAT Inhibitors in Pre-Clinical Studies

Due to their role in cholesterol metabolism, ACAT enzymes became a logical target for pharmacological inhibition to protect against atherosclerosis. Since ACAT1 is responsible for CE accumulation in macrophage foam cells, its inhibition was thought to reduce cholesterol accumulation in atherosclerotic lesions. Conversely, intestinal and liver ACAT2 plays an important role in cholesterol absorption; thus, its inhibition was thought to lead to reduced cholesterol absorption and serum levels. Therefore, many preclinical studies have been performed using various ACAT inhibitors, as demonstrated by the select examples below. These and additional examples are shown in Table 1.
  • Rabbit models
In 1991, Krause and colleagues used the Parke-Davis ACAT inhibitor CI-976 in New Zealand white rabbits being fed a high cholesterol diet and reported an increased regression of pre-existing lesions and a reduced formation of new lesions [49]. A different ACAT inhibitor, avasimibe (CI-1011), when given after changing from a high cholesterol to a normal chow diet, was also found to reduce pre-existing lesion cholesterol and macrophage content in rabbits, despite having no significant effects on plasma cholesterol levels [39]. Similar findings in rabbit studies have been observed with several other ACAT inhibitors, such as NTE-122 and F-1394 [58,59].
  • Mouse models
Avasimibe was found to lower plasma cholesterol levels in hypercholesterolemic apoE*3-Leiden mice fed a high-fat, 0.5% cholesterol, 0.1% cholate diet, leading to reductions in the aortic root lesion area, lesion severity, and monocyte adherence to the aortic root endothelium [41]. Fisher and colleagues demonstrated that the ACAT inhibitor F-1394 led to a dose-dependent decrease in the atherosclerotic lesion area and macrophage content, accompanied by a decrease in total plasma cholesterol in Apoe−/− mice fed a Western-type diet [34]. These results were extended to pre-existing lesions, where the mice treated with an ACAT inhibitor had slower lesion progression, with less macrophage, FC, and CE accumulation [35]. After switching Apoe−/− mice from a Western-type diet to a chow diet, the short-term addition of F-1394 decreased the lesion macrophage pro-inflammatory gene expression and increased the inflammation resolving gene expression [36]. Pactimibe sulfate (CS-505) treatment of Apoe−/− mice fed a chow diet decreased plasma cholesterol along with aortic root lesion development and progression, with the lesions being more significantly reduced with pactimibe sulfate vs. avasimibe treatment, despite similar effects on plasma cholesterol [43]. This compound was later used in human clinical trials (see below).
  • Adverse effects of ACAT inhibitors
The ACAT inhibitor PD 132301-2 was found to have adrenal toxicity in guinea pigs and dogs, leading to decreased plasma cortisol at baseline and after ACTH stimulation, and was associated with decreased adrenal cholesterol levels and increased release of liver enzymes into plasma [60,61]. Ex vivo adrenocortical cells treated with PD 132301-2 showed decreased mitochondrial function and ATP levels that preceded cell death [62].
Rothblat and colleagues found that an ACAT inhibitor treatment of cholesterol-loaded mouse macrophages led to increased cell death due to the buildup of free cholesterol [63,64]. Later, this group demonstrated that ACAT inhibition in cholesterol-loaded mouse macrophages led to intracellular FC crystal formation, and release of these crystals from the cells which continued to grow in size [65]. Tabas and colleagues published a series of elegant papers which examined the mechanism of macrophage cell death by FC loading via incubation with acetylated LDL in the presence of an ACAT inhibitor. The cell death was attributed to apoptosis, as it was blocked by caspase inhibitors, and, macrophages from the mice with a dysfunctional Fas ligand or Fas receptor were resistant to FC-loading-induced apoptosis [66]. In addition to the Fas pathway, they also showed that macrophage FC loading decreased the mitochondrial membrane potential leading to increased Bax levels and mitochondrial cytochrome C release, which resulted in caspase activation and apoptotic cell death [67]. More details were elucidated about the signaling involved in ACAT-inhibitor-mediated FC macrophage apoptosis. FC loading in macrophages activates the unfolded protein response, leading to the induction of the CHOP protein, and this induction requires the p38 MAP kinase activation cascade [68]. In addition, ligation of the scavenger receptor A (which mediates uptake of acetylated LDL) and JNK2 kinase are also necessary for FC-loading-induced macrophage apoptosis [68].

5. Clinical Trials of ACAT Inhibitors

Animal studies of ACAT inhibitors yielded sufficient evidence of anti-atherosclerosis activity that three human clinical trials were published using two different ACAT inhibitors. In a multi-center, randomized, double-blind, placebo-controlled clinical trial, placebo or three dose of avasimibe, along with lipid-lowering therapy, were administered to 639 coronary artery disease patients to examine changes in coronary atheroma volumes measured by intravascular ultrasound (IVUS) [69]. Compared to baseline atheroma volumes, there was no significant benefit of avasimibe after 24 months of treatment [69]. Additionally, the treated patients had increased LDL-C vs. baseline values.
Similarly, in the ACTIVATE trial, Nissen and colleagues enrolled 408 patients with confirmed coronary artery disease, in a multi-center, randomized, double-blind, placebo-controlled clinical trial of the ACAT inhibitor pactimibe [70]. Eighteen months of treatment vs. baseline failed to show a significant improvement in atheroma volume assessed by IVUS. Additionally, the treated vs. placebo patients had significantly less atheroma regression in the most diseased artery segment [70].
The CAPTIVATE trial was another multi-center, randomized, double-blind, placebo-controlled clinical trial which used the ACAT inhibitor pactimibe in 892 patients with heterozygous familial hypercholesterolemia, to assess carotid artery intimal-media thickness at baseline and at 12, 18 and 24 months later [71]. This trial was terminated prematurely after an average follow-up of 15 months when the results of the ACTIVATE trial became available. LDL-C increased by 7.3% in the pactimibe group, which was significantly more than the 1.4% increase in the placebo group. More importantly, major cardiovascular events were significantly increased in the treated vs. placebo patients [71].
There has been speculation about why ACAT inhibitors were successful in some animal models but not in human clinical trials. Global ACAT inhibition may lead to excess free cholesterol that can lead to cytotoxicity and proinflammatory effects that may not be beneficial for atherosclerosis, leading to the suggestion that ACAT2 selective inhibition might overcome this [72]. Avasimibe may have off-target effects that are also contraindicated, such as the activation of the human pregnane X receptor and the induction of CYP3A4, CYP2C9, and CYP2B6 among other genes [73,74], which may increase the catabolism of protective drugs, such as statins.

6. ACAT Genetics and Genomics

6.1. Mouse Soat1 Genetics

Adult male AKR mice have adrenocortical lipid depletion [75]. Farese and colleagues showed that this was due to a deletion of the first coding exon of the Soat1 gene, leading to an N-terminally truncated ACAT1 protein; however, ACAT enzyme activity was still present in the adrenal membranes and transfected CHO cells [76]. Using classical mouse genetic methods combined with CRISPR-Cas9 gene editing, our lab independently identified that the mouse Soat1 gene was responsible for macrophage CE levels after cholesterol loading, with the AKR allele having an exon 2 deletion leading to a lower CE accumulation [77]. Furthermore, the AKR decrease in macrophage ACAT activity can lead to more FC accumulation that can induce CHOP protein expression by acetylated LDL loading in the absence of an ACAT inhibitor [78].

6.2. Human SOAT1/2 Genes and Expression

Searching for the human SOAT1 gene using the UCSC genome browser (http://genome.ucsc.edu, accessed on 30 July 2021) shows that its 16 exons reside on chromosome 1 from 179,293,718–179,358,679 bp (GRCh38). Chang’s lab previously reported four human SOAT1 mRNAs on Northern blots [79]. Additionally, one of the mRNAs contained two additional exons, a 1277 bp exon derived from chromosome 7, called exon Xa that is not conserved in mice, and a second 10 bp exon that was from an unknown location, called exon Xb [79]. Our analysis of the exon Xa sequence places it at 120,669,474–120,670,751 (GRCh38—strand) within an intron of the KCND2 gene. Thus, this mRNA was thought to be due to trans-splicing that was confirmed by using human RNA with RNAse protection and cDNA PCR assays [79]. In transfection studies, another SOAT1 isoform was characterized that contains not only sequences from chromosomes 1 and 7, but also a sequence derived from the antisense strand of a plasmid-derived ampicillin resistance gene, claiming an exogenous–endogenous trans-splicing system [80]. The existence of trans-splicing in human mRNAs is controversial, as generation of cDNA libraries can lead to many artifactual chimeric mRNAs [81,82]. To provide a fresh look at this, we searched the GTEx project (gtexportal.org) and identified the adrenal gland, cultured fibroblasts, lung, EBV transformed lymphocytes, prostate, cervix, and spleen as the human tissues with the highest expression of SOAT1. Within the GTEx portal, we searched RNAseq reads in these tissues through the IGV Browser and found abundant reads in the SOAT1 exons on chromosome 1, but no reads through the Exon Xa on chromosome 7. Thus, we found no convincing RNAseq evidence for this chimeric transcript in human tissue.
The human SOAT2 gene contains 15 exons and maps to chromosome 12q13 from 53,103,485–53,124,534 bp (GRCh38). The GTEx portal shows the highest expression in the human spleen, small intestine, tibial nerve, aorta, and cervix; and surprisingly, the liver is listed as the 10th highest expressing tissue.

6.3. Human SOAT1/2 Common and Rare Genetic Variants and Disease Association

Before the era of genome-wide association studies (GWAS), researchers studied one or several common variants for association with a trait in relatively small studies involving humans, using p < 0.05 as the threshold for significance, which has yielded many false positive results [83]. There have been multiple studies that include subjects of Asian, European, and African descent reporting a single nucleotide polymorphism (SNP) in or near the SOAT1 or SOAT2 gene to be associated with lipoprotein levels, incidence of atherosclerotic disease, or Chagas disease, some of which were sex-specific or population specific [84,85,86,87,88,89,90,91,92,93]. In a well-powered meta-analysis, the SOAT1 SNP rs4421551 was associated with HDL-C (p = 4.81 × 10−3, n = 7840 subjects) [94]. There have also been SOAT1 variants that are associated with Alzheimer’s disease [95,96]. Knockout of mouse Soat1 or ACAT1 inhibitors has been found to decrease β-amyloid levels in mouse models of Alzheimer’s disease [97,98,99,100].
In the modern GWAS era, the threshold for genetic association significance is much more stringent at p < 5 × 10−8. Searching the GWAS catalog in July 2021 found only seven associations for SNPs nearest to the SOAT1 gene, four of which are for testosterone levels, and one each for morningness, type 2 diabetes, and white matter microstructure (Table 2). There are abundant SOAT1 expression quantitative trait locus (eQTL) SNPs associated with SOAT1 expression in many tissues in the GTEx portal. The finding of no SOAT1 GWAS SNPs associated with lipoprotein levels, atherosclerotic disease, or Alzheimer’s disease, which have been the objectives of large, well-powered GWAS studies, indicates that the SOAT1 expression levels do not play a significant role in these phenotypes. Of the three SNPs associated with testosterone levels, two (rs2152318 and rs2248979) were strong eQTL SNPs associated with SOAT1 expression in the adrenal gland. This may be biologically plausible, as adrenal steroids can be converted to androgens [101]. Likewise, searching the GWAS catalog for SOAT2 identified only one association for age-related macular degeneration, although this failed to meet the stringent GWAS p-value threshold (Table 2). Again, numerous eQTL SNPs associated with SOAT2 expression are found in the GTEx portal; however, not in the small intestine or liver, the two tissues that might affect lipid metabolism the most.
One limitation of GWAS is that they are underpowered for rare variants, whose discovery is mainly through exome and whole genome sequencing. We searched the ClinVar database (ncbi.nlm.nih.gov/clinvar/, accessed on 30 July 2021) for rare SOAT1/2 variants in July 2021. SOAT1 rare variants in the coding region include four synonymous (all classified as benign) and one non-synonymous variant (rs73048613, Gln204GLu), the latter of which is enriched in African populations (~4%) and is very rarely found in European populations, but is classified as benign by computational analysis and by one submitter. SOAT2 rare variants in the coding region include two synonymous (both classified as benign) and one non-synonymous variant (rs149485045, Arg417Gln), the later which has a <1% allele frequency in all populations, but also being classified as benign. In summary, neither common nor rare variants in SOAT1/2 are associated with lipoprotein levels, atherosclerosis, or Alzheimer’s disease, despite abundant biochemical and mouse studies showing effects on related phenotypes. Thus, a role for selective ACAT2 inhibition as a human therapeutic is not supported by human genetic studies.

7. Conclusions

ACAT enzymes play essential roles in the storage of cellular cholesterol as cholesteryl esters, which help to protect cells from the adverse effects of excess free cholesterol. Long-standing research projects, including mouse gene knockout studies, have led to understanding the tissue expression differences and the roles of ACAT1 and ACAT2 in lipoprotein metabolism, adrenal function, and cholesterol absorption, which supported preclinical and clinical trials of ACAT inhibitors. Clinical trials using non-selective ACAT inhibitors have not proven to be effective in reducing atheroma, and can lead to a small increase in LDL-C in familial hypercholesterolemia heterozygous patients. In addition, there is little support for ACAT enzymes as a drug target for cardiovascular disease based on the lack of findings from SOAT1 and SOAT2 genome-wide association studies, and the absence of rare variants associated with relevant phenotypes.

Author Contributions

Conceptualization, Q.H. and J.D.S.; writing—original draft preparation, Q.H. and J.D.S.; writing—review and editing, Q.H. and J.D.S.; funding acquisition, J.D.S. Both authors have read and agreed to the published version of the manuscript.

Funding

These investigators were funded by National Institutes of Health grant numbers R01HL128268 and R01HL156499.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chang, T.Y.; Li, B.L.; Chang, C.C.; Urano, Y. Acyl-coenzyme A:cholesterol acyltransferases. Am. J. Physiol. Endocrinol. Metab. 2009, 297, E1–E9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cadigan, K.M.; Heider, J.G.; Chang, T.Y. Isolation and characterization of Chinese hamster ovary cell mutants deficient in acyl-coenzyme A:cholesterol acyltransferase activity. J. Biol. Chem. 1988, 263, 274–282. [Google Scholar] [CrossRef]
  3. Pavanello, C.; Calabresi, L. Genetic, biochemical, and clinical features of LCAT deficiency: Update for 2020. Curr. Opin. Lipidol. 2020, 31, 232–237. [Google Scholar] [CrossRef]
  4. Anderson, R.A.; Joyce, C.; Davis, M.; Reagan, J.W.; Clark, M.; Shelness, G.S.; Rudel, L.L. Identification of a form of acyl-CoA:cholesterol acyltransferase specific to liver and intestine in nonhuman primates. J. Biol. Chem. 1998, 273, 26747–26754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Chang, T.Y.; Chang, C.C.; Lin, S.; Yu, C.; Li, B.L.; Miyazaki, A. Roles of acyl-coenzyme A:cholesterol acyltransferase-1 and -2. Curr. Opin. Lipidol. 2001, 12, 289–296. [Google Scholar] [CrossRef] [Green Version]
  6. Cases, S.; Novak, S.; Zheng, Y.W.; Myers, H.M.; Lear, S.R.; Sande, E.; Welch, C.B.; Lusis, A.J.; Spencer, T.A.; Krause, B.R.; et al. ACAT-2, a second mammalian acyl-CoA:cholesterol acyltransferase. Its cloning, expression, and characterization. J. Biol. Chem. 1998, 273, 26755–26764. [Google Scholar] [CrossRef] [Green Version]
  7. Cases, S.; Smith, S.J.; Zheng, Y.W.; Myers, H.M.; Lear, S.R.; Sande, E.; Novak, S.; Collins, C.; Welch, C.B.; Lusis, A.J.; et al. Identification of a gene encoding an acyl CoA:diacylglycerol acyltransferase, a key enzyme in triacylglycerol synthesis. Proc. Natl. Acad. Sci. USA 1998, 95, 13018–13023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Hofmann, K. A superfamily of membrane-bound O-acyltransferases with implications for wnt signaling. Trends Biochem. Sci. 2000, 25, 111–112. [Google Scholar] [CrossRef]
  9. Rogers, M.A.; Liu, J.; Song, B.L.; Li, B.L.; Chang, C.C.; Chang, T.Y. Acyl-CoA:cholesterol acyltransferases (ACATs/SOATs): Enzymes with multiple sterols as substrates and as activators. J. Steroid Biochem. Mol. Biol. 2015, 151, 102–107. [Google Scholar] [CrossRef] [Green Version]
  10. Chang, C.; Dong, R.; Miyazaki, A.; Sakashita, N.; Zhang, Y.; Liu, J.; Guo, M.; Li, B.L.; Chang, T.Y. Human acyl-CoA:cholesterol acyltransferase (ACAT) and its potential as a target for pharmaceutical intervention against atherosclerosis. Acta Biochim. Biophys. Sin. 2006, 38, 151–156. [Google Scholar] [CrossRef] [Green Version]
  11. Cadigan, K.M.; Chang, C.C.; Chang, T.Y. Isolation of Chinese hamster ovary cell lines expressing human acyl-coenzyme A/cholesterol acyltransferase activity. J. Cell Biol. 1989, 108, 2201–2210. [Google Scholar] [CrossRef] [Green Version]
  12. Chang, C.C.; Huh, H.Y.; Cadigan, K.M.; Chang, T.Y. Molecular cloning and functional expression of human acyl-coenzyme A:cholesterol acyltransferase cDNA in mutant Chinese hamster ovary cells. J. Biol. Chem. 1993, 268, 20747–20755. [Google Scholar] [CrossRef]
  13. Chang, C.C.; Noll, W.W.; Nutile-McMenemy, N.; Lindsay, E.A.; Baldini, A.; Chang, W.; Chang, T.Y. Localization of acyl coenzyme A:cholesterol acyltransferase gene to human chromosome 1q25. Somat. Cell Mol. Genet. 1994, 20, 71–74. [Google Scholar] [CrossRef]
  14. Chang, C.C.; Lee, C.Y.; Chang, E.T.; Cruz, J.C.; Levesque, M.C.; Chang, T.Y. Recombinant acyl-CoA:cholesterol acyltransferase-1 (ACAT-1) purified to essential homogeneity utilizes cholesterol in mixed micelles or in vesicles in a highly cooperative manner. J. Biol. Chem. 1998, 273, 35132–35141. [Google Scholar] [CrossRef] [Green Version]
  15. Zhang, Y.; Yu, C.; Liu, J.; Spencer, T.A.; Chang, C.C.Y.; Chang, T.-Y. Cholesterol is superior to 7-ketocholesterol or 7 alpha-hydroxycholesterol as an allosteric activator for acyl-coenzyme A:cholesterol acyltransferase 1. J. Biol. Chem. 2003, 278, 11642–11647. [Google Scholar] [CrossRef] [Green Version]
  16. Rogers, M.A.; Liu, J.; Kushnir, M.M.; Bryleva, E.; Rockwood, A.L.; Meikle, A.W.; Shapiro, D.; Vaisman, B.L.; Remaley, A.T.; Chang, C.C.Y.; et al. Cellular pregnenolone esterification by acyl-CoA:cholesterol acyltransferase. J. Biol. Chem. 2012, 287, 17483–17492. [Google Scholar] [CrossRef] [Green Version]
  17. Chang, C.C.; Miyazaki, A.; Dong, R.; Kheirollah, A.; Yu, C.; Geng, Y.; Higgs, H.N.; Chang, T.Y. Purification of recombinant acyl-coenzyme A:cholesterol acyltransferase 1 (ACAT1) from H293 cells and binding studies between the enzyme and substrates using difference intrinsic fluorescence spectroscopy. Biochemistry 2010, 49, 9957–9963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Seo, T.; Oelkers, P.M.; Giattina, M.R.; Worgall, T.S.; Sturley, S.L.; Deckelbaum, R.J. Differential modulation of ACAT1 and ACAT2 transcription and activity by long chain free fatty acids in cultured cells. Biochemistry 2001, 40, 4756–4762. [Google Scholar] [CrossRef] [PubMed]
  19. Temel, R.E.; Gebre, A.K.; Parks, J.S.; Rudel, L.L. Compared with Acyl-CoA:cholesterol O-acyltransferase (ACAT) 1 and lecithin:cholesterol acyltransferase, ACAT2 displays the greatest capacity to differentiate cholesterol from sitosterol. J. Biol. Chem. 2003, 278, 47594–47601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Qian, H.; Zhao, X.; Yan, R.; Yao, X.; Gao, S.; Sun, X.; Du, X.; Yang, H.; Wong, C.C.L.; Yan, N. Structural basis for catalysis and substrate specificity of human ACAT1. Nature 2020, 581, 333–338. [Google Scholar] [CrossRef] [PubMed]
  21. Meiner, V.L.; Cases, S.; Myers, H.M.; Sande, E.R.; Bellosta, S.; Schambelan, M.; Pitas, R.E.; McGuire, J.; Herz, J.; Farese, R.V.J. Disruption of the acyl-CoA:cholesterol acyltransferase gene in mice: Evidence suggesting multiple cholesterol esterification enzymes in mammals. Proc. Natl. Acad. Sci. USA 1996, 93, 14041–14046. [Google Scholar] [CrossRef] [Green Version]
  22. Huang, L.H.; Gui, J.; Artinger, E.; Craig, R.; Berwin, B.L.; Ernst, P.A.; Chang, C.C.; Chang, T.Y. Acat1 gene ablation in mice increases hematopoietic progenitor cell proliferation in bone marrow and causes leukocytosis. Arter. Thromb. Vasc. Biol. 2013, 33, 2081–2087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Buhman, K.K.; Accad, M.; Novak, S.; Choi, R.S.; Wong, J.S.; Hamilton, R.L.; Turley, S.; Farese, R.V.J. Resistance to diet-induced hypercholesterolemia and gallstone formation in ACAT2-deficient mice. Nat. Med. 2000, 6, 1341–1347. [Google Scholar] [CrossRef]
  24. Repa, J.J.; Buhman, K.K.; Farese, R.V.J.; Dietschy, J.M.; Turley, S.D. ACAT2 deficiency limits cholesterol absorption in the cholesterol-fed mouse: Impact on hepatic cholesterol homeostasis. Hepatology 2004, 40, 1088–1097. [Google Scholar] [CrossRef] [PubMed]
  25. Accad, M.; Smith, S.J.; Newland, D.L.; Sanan, D.A.; King, L.E.J.; Linton, M.F.; Fazio, S.; Farese, R.V.J. Massive xanthomatosis and altered composition of atherosclerotic lesions in hyperlipidemic mice lacking acyl CoA:cholesterol acyltransferase 1. J. Clin. Investig. 2000, 105, 711–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Fazio, S.; Major, A.S.; Swift, L.L.; Gleaves, L.A.; Accad, M.; Linton, M.F.; Farese, R.V.J. Increased atherosclerosis in LDL receptor-null mice lacking ACAT1 in macrophages. J. Clin. Investig. 2001, 107, 163–171. [Google Scholar] [CrossRef] [Green Version]
  27. Yagyu, H.; Kitamine, T.; Osuga, J.I.; Tozawa, R.I.; Chen, Z.; Kaji, Y.; Oka, T.; Perrey, S.; Tamura, Y.; Ohashi, K.; et al. Absence of ACAT-1 attenuates atherosclerosis but ’causes dry eye and cutaneous xanthomatosis mice with congenital hyperlipidemia. J. Biol. Chem. 2000, 275, 21324–21330. [Google Scholar] [CrossRef] [Green Version]
  28. Huang, L.H.; Melton, E.M.; Li, H.; Sohn, P.; Rogers, M.A.; Mulligan-Kehoe, M.J.; Fiering, S.N.; Hickey, W.F.; Chang, C.C.; Chang, T.Y. Myeloid Acyl-CoA:Cholesterol Acyltransferase 1 Deficiency Reduces Lesion Macrophage Content and Suppresses Atherosclerosis Progression. J. Biol. Chem. 2016, 291, 6232–6244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Melton, E.M.; Li, H.; Benson, J.; Sohn, P.; Huang, L.-H.; Song, B.-L.; Li, B.-L.; Chang, C.C.Y.; Chang, T.-Y. Myeloid Acat1/Soat1 KO attenuates pro-inflammatory responses in macrophages and protects against atherosclerosis in a model of advanced lesions. J. Biol. Chem. 2019, 294, 15836–15849. [Google Scholar] [CrossRef]
  30. Willner, E.L.; Tow, B.; Buhman, K.K.; Wilson, M.; Sanan, D.A.; Rudel, L.L.; Farese, R.V.J. Deficiency of acyl CoA:cholesterol acyltransferase 2 prevents atherosclerosis in apolipoprotein E-deficient mice. Proc. Natl. Acad. Sci. USA 2003, 100, 1262–1267. [Google Scholar] [CrossRef] [Green Version]
  31. Kusunoki, J.; Aragane, K.; Kitamine, T.; Higashinakagawa, S.; Kase, N.; Yamaura, T.; Ohnishi, H. Hypocholesterolemic action and prevention of cholesterol absorption via the gut by F-1394, a potent acyl-CoA:cholesterol acyltransferase (ACAT) inhibitor, in cholesterol diet-fed rats. Jpn. J. Pharmacol. 1995, 69, 53–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Chiwata, T.; Aragane, K.; Fujinami, K.; Kojima, K.; Ishibashi, S.; Yamada, N.; Kusunoki, J. Direct effect of an acyl-CoA:cholesterol acyltransferase inhibitor, F-1394, on atherosclerosis in apolipoprotein E and low density lipoprotein receptor double knockout mice. Br. J. Pharmacol. 2001, 133, 1005–1012. [Google Scholar] [CrossRef] [Green Version]
  33. Aragane, K.; Fujinami, K.; Kojima, K.; Kusunoki, J. ACAT inhibitor F-1394 prevents intimal hyperplasia induced by balloon injury in rabbits. J. Lipid Res. 2001, 42, 480–488. [Google Scholar] [CrossRef]
  34. Kusunoki, J.; Hansoty, D.K.; Aragane, K.; Fallon, J.T.; Badimon, J.J.; Fisher, E.A. Acyl-CoA:cholesterol acyltransferase inhibition reduces atherosclerosis in apolipoprotein E-deficient mice. Circulation 2001, 103, 2604–2609. [Google Scholar] [CrossRef] [Green Version]
  35. Rong, J.X.; Blachford, C.; Feig, J.E.; Bander, I.; Mayne, J.; Kusunoki, J.; Miller, C.; Davis, M.; Wilson, M.; Dehn, S.; et al. ACAT inhibition reduces the progression of preexisting, advanced atherosclerotic mouse lesions without plaque or systemic toxicity. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 4–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Amengual, J.; Ogando, Y.; Nikain, C.; Quezada, A.; Qian, K.; Vaisar, T.; Fisher, E.A. Short-Term Acyl-CoA:Cholesterol Acyltransferase Inhibition, Combined with Apoprotein A1 Overexpression, Promotes Atherosclerosis Inflammation Resolution in Mice. Mol. Pharmacol. 2021, 99, 175–183. [Google Scholar] [CrossRef]
  37. Ramharack, R.; Spahr, M.A.; Sekerke, C.S.; Stanfield, R.L.; Bousley, R.F.; Lee, H.T.; Krause, B.K. CI-1011 lowers lipoprotein(a) and plasma cholesterol concentrations in chow-fed cynomolgus monkeys. Atherosclerosis 1998, 136, 79–87. [Google Scholar] [CrossRef]
  38. Burnett, J.R.; Wilcox, L.J.; Telford, D.E.; Kleinstiver, S.J.; Barrett, P.H.; Newton, R.S.; Huff, M.W. Inhibition of ACAT by avasimibe decreases both VLDL and LDL apolipoprotein B production in miniature pigs. J. Lipid Res. 1999, 40, 1317–1327. [Google Scholar] [CrossRef]
  39. Bocan, T.M.; Krause, B.R.; Rosebury, W.S.; Mueller, S.B.; Lu, X.; Dagle, C.; Major, T.; Lathia, C.; Lee, H. The ACAT inhibitor avasimibe reduces macrophages and matrix metalloproteinase expression in atherosclerotic lesions of hypercholesterolemic rabbits. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 70–79. [Google Scholar] [CrossRef] [Green Version]
  40. Robertson, D.G.; Breider, M.A.; Milad, M.A. Preclinical safety evaluation of avasimibe in beagle dogs: An ACAT inhibitor with minimal adrenal effects. Toxicol. Sci. 2001, 59, 324–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Delsing, D.J.; Offerman, E.H.; van Duyvenvoorde, W.; van Der Boom, H.; de Wit, E.C.; Gijbels, M.J.; van Der Laarse, A.; Jukema, J.W.; Havekes, L.M.; Princen, H.M. Acyl-CoA:cholesterol acyltransferase inhibitor avasimibe reduces atherosclerosis in addition to its cholesterol-lowering effect in ApoE*3-Leiden mice. Circulation 2001, 103, 1778–1786. [Google Scholar] [CrossRef]
  42. Burnett, J.R.; Telford, D.E.; Barrett, P.H.R.; Huff, M.W. The ACAT inhibitor avasimibe increases the fractional clearance rate of postprandial triglyceride-rich lipoproteins in miniature pigs. Biochim. Biophys. Acta 2005, 1738, 10–18. [Google Scholar] [CrossRef] [PubMed]
  43. Terasaka, N.; Miyazaki, A.; Kasanuki, N.; Ito, K.; Ubukata, N.; Koieyama, T.; Kitayama, K.; Tanimoto, T.; Maeda, N.; Inaba, T. ACAT inhibitor pactimibe sulfate (CS-505) reduces and stabilizes atherosclerotic lesions by cholesterol-lowering and direct effects in apolipoprotein E-deficient mice. Atherosclerosis 2007, 190, 239–247. [Google Scholar] [CrossRef]
  44. Worthley, S.G.; Helft, G.; Corti, R.; Worthley, M.I.; Chew, D.P.; Fayad, Z.A.; Zaman, A.G.; Fallon, J.T.; Fuster, V.; Badimon, J.J. Statin therapy alone and in combination with an acyl-CoA:cholesterol O-acyltransferase inhibitor on experimental atherosclerosis. Pathophysiol. Haemost. Thromb. 2007, 36, 9–17. [Google Scholar] [CrossRef]
  45. Kitayama, K.; Koga, T.; Maeda, N.; Inaba, T.; Fujioka, T. Pactimibe stabilizes atherosclerotic plaque through macrophage acyl-CoA:cholesterol acyltransferase inhibition in WHHL rabbits. Eur. J. Pharmacol. 2006, 539, 81–88. [Google Scholar] [CrossRef] [PubMed]
  46. Kitayama, K.; Tanimoto, T.; Koga, T.; Terasaka, N.; Fujioka, T.; Inaba, T. Importance of acyl-coenzyme A:cholesterol acyltransferase 1/2 dual inhibition for anti-atherosclerotic potency of pactimibe. Eur. J. Pharmacol. 2006, 540, 121–130. [Google Scholar] [CrossRef] [PubMed]
  47. Kitayama, K.; Koga, T.; Inaba, T.; Fujioka, T. Multiple mechanisms of hypocholesterolemic action of pactimibe, a novel acyl-coenzyme A:cholesterol acyltransferase inhibitor. Eur. J. Pharmacol. 2006, 543, 123–132. [Google Scholar] [CrossRef]
  48. Yoshinaka, Y.; Shibata, H.; Kobayashi, H.; Kuriyama, H.; Shibuya, K.; Tanabe, S.; Watanabe, T.; Miyazaki, A. A selective ACAT-1 inhibitor, K-604, stimulates collagen production in cultured smooth muscle cells and alters plaque phenotype in apolipoprotein E-knockout mice. Atherosclerosis 2010, 213, 85–91. [Google Scholar] [CrossRef]
  49. Bocan, T.M.; Mueller, S.B.; Uhlendorf, P.D.; Newton, R.S.; Krause, B.R. Comparison of CI-976, an ACAT inhibitor, and selected lipid-lowering agents for antiatherosclerotic activity in iliac-femoral and thoracic aortic lesions. A biochemical, morphological, and morphometric evaluation. Arterioscler. Thromb. J. Vasc. Biol. 1991, 11, 1830–1843. [Google Scholar] [CrossRef] [Green Version]
  50. Krause, B.R.; Bousley, R.F.; Kieft, K.A.; Stanfield, R.L. Effect of the ACAT inhibitor CI-976 on plasma cholesterol concentrations and distribution in hamsters fed zero- and low-cholesterol diets. Clin. Biochem. 1992, 25, 371–377. [Google Scholar] [CrossRef]
  51. Krause, B.R.; Anderson, M.; Bisgaier, C.L.; Bocan, T.; Bousley, R.; DeHart, P.; Essenburg, A.; Hamelehle, K.; Homan, R.; Kieft, K. In vivo evidence that the lipid-regulating activity of the ACAT inhibitor CI-976 in rats is due to inhibition of both intestinal and liver ACAT. J. Lipid Res. 1993, 34, 279–294. [Google Scholar] [CrossRef]
  52. Bocan, T.M.; Mueller, S.B.; Uhlendorf, P.D.; Brown, E.Q.; Mazur, M.J.; Black, A.E. Inhibition of acyl-CoA cholesterol O-acyltransferase reduces the cholesteryl ester enrichment of atherosclerotic lesions in the Yucatan micropig. Atherosclerosis 1993, 99, 175–186. [Google Scholar] [CrossRef]
  53. Krause, B.R.; Pape, M.E.; Kieft, K.; Auerbach, B.; Bisgaier, C.L.; Homan, R.; Newton, R.S. ACAT inhibition decreases LDL cholesterol in rabbits fed a cholesterol-free diet. Marked changes in LDL cholesterol without changes in LDL receptor mRNA abundance. Arterioscler. Thromb. J. Vasc. Biol. 1994, 14, 598–604. [Google Scholar] [CrossRef] [Green Version]
  54. Bocan, T.M.; Mueller, S.B.; Brown, E.Q.; Lee, P.; Bocan, M.J.; Rea, T.; Pape, M.E. HMG-CoA reductase and ACAT inhibitors act synergistically to lower plasma cholesterol and limit atherosclerotic lesion development in the cholesterol-fed rabbit. Atherosclerosis 1998, 139, 21–30. [Google Scholar] [CrossRef]
  55. Vaziri, N.D.; Liang, K.H. Acyl-coenzyme A:cholesterol acyltransferase inhibition ameliorates proteinuria, hyperlipidemia, lecithin-cholesterol acyltransferase, SRB-1, and low-denisty lipoprotein receptor deficiencies in nephrotic syndrome. Circulation 2004, 110, 419–425. [Google Scholar] [CrossRef] [Green Version]
  56. Ikenoya, M.; Yoshinaka, Y.; Kobayashi, H.; Kawamine, K.; Shibuya, K.; Sato, F.; Sawanobori, K.; Watanabe, T.; Miyazaki, A. A selective ACAT-1 inhibitor, K-604, suppresses fatty streak lesions in fat-fed hamsters without affecting plasma cholesterol levels. Atherosclerosis 2007, 191, 290–297. [Google Scholar] [CrossRef]
  57. Krause, B.R.; Black, A.; Bousley, R.; Essenburg, A.; Cornicelli, J.; Holmes, A.; Homan, R.; Kieft, K.; Sekerke, C.; Shaw-Hes, M.K. Divergent pharmacologic activities of PD 132301-2 and CL 277,082, urea inhibitors of acyl-CoA:cholesterol acyltransferase. J. Pharmacol. Exp. Ther. 1993, 267, 734–743. [Google Scholar] [PubMed]
  58. Azuma, Y.; Date, K.; Ohno, K.; Matsushiro, S.; Nobuhara, Y.; Yamada, T. NTE-122, an acyl-coa:cholesterol acyltransferase inhibitor, prevents the progression of atherogenesis in cholesterol-fed rabbits. Jpn. J. Pharmacol. 2001, 86, 120–123. [Google Scholar] [CrossRef] [Green Version]
  59. Aragane, K.; Kojima, K.; Fujinami, K.; Kamei, J.; Kusunoki, J. Effect of F-1394, an acyl-CoA:cholesterol acyltransferase inhibitor, on atherosclerosis induced by high cholesterol diet in rabbits. Atherosclerosis 2001, 158, 139–145. [Google Scholar] [CrossRef]
  60. Dominick, M.A.; McGuire, E.J.; Reindel, J.F.; Bobrowski, W.F.; Bocan, T.M.; Gough, A.W. Subacute toxicity of a novel inhibitor of acyl-CoA: Cholesterol acyltransferase in beagle dogs. Fundam. Appl. Toxicol. 1993, 20, 217–224. [Google Scholar] [CrossRef] [PubMed]
  61. Dominick, M.A.; Bobrowski, W.A.; MacDonald, J.R.; Gough, A.W. Morphogenesis of a zone-specific adrenocortical cytotoxicity in guinea pigs administered PD 132301-2, an inhibitor of acyl-CoA:cholesterol acyltransferase. Toxicol. Pathol. 1993, 21, 54–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Vernetti, L.A.; MacDonald, J.R.; Wolfgang, G.H.; Dominick, M.A.; Pegg, D.G. ATP depletion is associated with cytotoxicity of a novel lipid regulator in guinea pig adrenocortical cells. Toxicol. Appl. Pharmacol. 1993, 118, 30–38. [Google Scholar] [CrossRef]
  63. Warner, G.J.; Stoudt, G.; Bamberger, M.; Johnson, W.J.; Rothblat, G.H. Cell toxicity induced by inhibition of acyl coenzyme A:cholesterol acyltransferase and accumulation of unesterified cholesterol. J. Biol. Chem. 1995, 270, 5772–5778. [Google Scholar] [CrossRef] [Green Version]
  64. Kellner-Weibel, G.; Jerome, W.G.; Small, D.M.; Warner, G.J.; Stoltenborg, J.K.; Kearney, M.A.; Corjay, M.H.; Phillips, M.C.; Rothblat, G.H. Effects of intracellular free cholesterol accumulation on macrophage viability: A model for foam cell death. Arterioscler. Thromb. Vasc. Biol. 1998, 18, 423–431. [Google Scholar] [CrossRef] [Green Version]
  65. Kellner-Weibel, G.; Yancey, P.G.; Jerome, W.G.; Walser, T.; Mason, R.P.; Phillips, M.C.; Rothblat, G.H. Crystallization of free cholesterol in model macrophage foam cells. Arterioscler. Thromb. Vasc. Biol. 1999, 19, 1891–1898. [Google Scholar] [CrossRef]
  66. Yao, P.M.; Tabas, I. Free cholesterol loading of macrophages induces apoptosis involving the fas pathway. J. Biol. Chem. 2000, 275, 23807–23813. [Google Scholar] [CrossRef] [Green Version]
  67. Yao, P.M.; Tabas, I. Free cholesterol loading of macrophages is associated with widespread mitochondrial dysfunction and activation of the mitochondrial apoptosis pathway. J. Biol. Chem. 2001, 276, 42468–42476. [Google Scholar] [CrossRef] [Green Version]
  68. Devries-Seimon, T.; Li, Y.; Yao, P.M.; Stone, E.; Wang, Y.; Davis, R.J.; Flavell, R.; Tabas, I. Cholesterol-induced macrophage apoptosis requires ER stress pathways and engagement of the type A scavenger receptor. J. Cell Biol. 2005, 171, 61–73. [Google Scholar] [CrossRef] [PubMed]
  69. Tardif, J.-C.; Grégoire, J.; L’Allier, P.L.; Anderson, T.J.; Bertrand, O.; Reeves, F.; Title, L.M.; Alfonso, F.; Schampaert, E.; Hassan, A.; et al. Effects of the acyl coenzyme A:cholesterol acyltransferase inhibitor avasimibe on human atherosclerotic lesions. Circulation 2004, 110, 3372–3377. [Google Scholar] [CrossRef] [Green Version]
  70. Nissen, S.E.; Tuzcu, E.M.; Brewer, H.B.; Sipahi, I.; Nicholls, S.J.; Ganz, P.; Schoenhagen, P.; Waters, D.D.; Pepine, C.J.; Crowe, T.D.; et al. Effect of ACAT inhibition on the progression of coronary atherosclerosis. N. Engl. J. Med. 2006, 354, 1253–1263. [Google Scholar] [CrossRef] [PubMed]
  71. Meuwese, M.C.; de Groot, E.; Duivenvoorden, R.; Trip, M.D.; Ose, L.; Maritz, F.J.; Basart, D.C.G.; Kastelein, J.J.P.; Habib, R.; Davidson, M.H.; et al. ACAT inhibition and progression of carotid atherosclerosis in patients with familial hypercholesterolemia: The CAPTIVATE randomized trial. JAMA 2009, 301, 1131–1139. [Google Scholar] [CrossRef]
  72. Farese, R.V.J. The nine lives of ACAT inhibitors. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 1684–1686. [Google Scholar] [CrossRef] [Green Version]
  73. Sahi, J.; Milad, M.A.; Zheng, X.; Rose, K.A.; Wang, H.; Stilgenbauer, L.; Gilbert, D.; Jolley, S.; Stern, R.H.; LeCluyse, E.L. Avasimibe induces CYP3A4 and multiple drug resistance protein 1 gene expression through activation of the pregnane X receptor. J. Pharmacol. Exp. Ther. 2003, 306, 1027–1034. [Google Scholar] [CrossRef]
  74. Sahi, J.; Stern, R.H.; Milad, M.A.; Rose, K.A.; Gibson, G.; Zheng, X.; Stilgenbauer, L.; Sadagopan, N.; Jolley, S.; Gilbert, D.; et al. Effects of avasimibe on cytochrome P450 2C9 expression in vitro and in vivo. Drug Metab. Dispos. 2004, 32, 1370–1376. [Google Scholar] [CrossRef] [Green Version]
  75. ARNESEN, K. Constitutional difference in lipid content of adrenals in two strains of mice and their hybrids. Acta Endocrinol. 1955, 18, 396–401. [Google Scholar] [CrossRef] [PubMed]
  76. Meiner, V.L.; Welch, C.L.; Cases, S.; Myers, H.M.; Sande, E.; Lusis, A.J.; Farese, R.V.J. Adrenocortical lipid depletion gene (ald) in AKR mice is associated with an acyl-CoA:cholesterol acyltransferase (ACAT) mutation. J. Biol. Chem. 1998, 273, 1064–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Hai, Q.; Ritchey, B.; Robinet, P.; Alzayed, A.M.; Brubaker, G.; Zhang, J.; Smith, J.D. Quantitative Trait Locus Mapping of Macrophage Cholesterol Metabolism and CRISPR/Cas9 Editing Implicate an ACAT1 Truncation as a Causal Modifier Variant. Arterioscler. Thromb. Vasc. Biol. 2018, 38, 83–91. [Google Scholar] [CrossRef] [PubMed]
  78. Luo, M.; Opoku, E.; Traughber, C.A.; Hai, Q.; Robinet, P.; Berisha, S.; Smith, J.D. Soat1 mediates the mouse strain effects on cholesterol loading-induced endoplasmic reticulum stress and CHOP expression in macrophages. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2021, 1866, 158825. [Google Scholar] [CrossRef] [PubMed]
  79. Li, B.L.; Li, X.L.; Duan, Z.J.; Lee, O.; Lin, S.; Ma, Z.M.; Chang, C.C.; Yang, X.Y.; Park, J.P.; Mohandas, T.K.; et al. Human acyl-CoA:cholesterol acyltransferase-1 (ACAT-1) gene organization and evidence that the 4.3-kilobase ACAT-1 mRNA is produced from two different chromosomes. J. Biol. Chem. 1999, 274, 11060–11071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Hu, G.-J.; Chen, J.; Zhao, X.-N.; Xu, J.-J.; Guo, D.-Q.; Lu, M.; Zhu, M.; Xiong, Y.; Li, Q.; Chang, C.C.; et al. Production of ACAT1 56-kDa isoform in human cells via trans-splicing involving the ampicillin resistance gene. Cell Res. 2013, 23, 1007–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Lei, Q.; Li, C.; Zuo, Z.; Huang, C.; Cheng, H.; Zhou, R. Evolutionary Insights into RNA trans-Splicing in Vertebrates. Genome Biol. Evol. 2016, 8, 562–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. He, Y.; Yuan, C.; Chen, L.; Lei, M.; Zellmer, L.; Huang, H.; Liao, D.J. Transcriptional-Readthrough RNAs Reflect the Phenomenon of “A Gene Contains Gene(s)” or “Gene(s) within a Gene” in the Human Genome, and Thus Are Not Chimeric RNAs. Genes 2018, 9, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Ioannidis, J.P.; Ntzani, E.E.; Trikalinos, T.A.; Contopoulos-Ioannidis, D.G. Replication validity of genetic association studies. Nat. Genet. 2001, 29, 306–309. [Google Scholar] [CrossRef] [PubMed]
  84. Ohta, T.; Takata, K.; Katsuren, K.; Fukuyama, S. The influence of the acyl-CoA:cholesterol acyltransferase-1 gene (-77G-->A) polymorphisms on plasma lipid and apolipoprotein levels in normolipidemic and hyperlipidemic subjects. Biochim. Biophys. Acta 2004, 1682, 56–62. [Google Scholar] [CrossRef]
  85. Chen, S.N.; Cilingiroglu, M.; Todd, J.; Lombardi, R.; Willerson, J.T.; Gotto, A.M.J.; Ballantyne, C.M.; Marian, A.J. Candidate genetic analysis of plasma high-density lipoprotein-cholesterol and severity of coronary atherosclerosis. BMC Med. Genet. 2009, 10, 111. [Google Scholar] [CrossRef] [Green Version]
  86. Wu, D.-F.; Yin, R.-X.; Aung, L.H.H.; Hu, X.-J.; Cao, X.-L.; Miao, L.; Li, Q.; Yan, T.-T.; Wu, J.-Z.; Pan, S.-L. Polymorphism of rs1044925 in the acyl-CoA:cholesterol acyltransferase-1 gene and serum lipid levels in the Guangxi Bai Ku Yao and Han populations. Lipids Health Dis. 2010, 9, 139. [Google Scholar] [CrossRef] [Green Version]
  87. Wu, D.-F.; Yin, R.-X.; Aung, L.H.H.; Li, Q.; Yan, T.-T.; Zeng, X.-N.; Huang, K.-K.; Huang, P.; Wu, J.-Z.; Pan, S.-L. Sex-specific association of ACAT-1 rs1044925 SNP and serum lipid levels in the hypercholesterolemic subjects. Lipids Health Dis. 2012, 11, 9. [Google Scholar] [CrossRef] [Green Version]
  88. Wu, D.-F.; Yin, R.-X.; Cao, X.-L.; Chen, W.-X. Association between single nucleotide polymorphism rs1044925 and the risk of coronary artery disease and ischemic stroke. Int. J. Mol. Sci. 2014, 15, 3546–3559. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Wang, Y.-T.; Wang, Y.-H.; Ma, Y.-T.; Fu, Z.-Y.; Yang, Y.-N.; Ma, X.; Li, X.-M.; Adi, D.; Liu, F.; Chen, B.-D. ACAT-1 gene polymorphism is associated with increased susceptibility to coronary artery disease in Chinese Han population: A case-control study. Oncotarget 2017, 8, 89055–89063. [Google Scholar] [CrossRef] [Green Version]
  90. Wang, Y.-T.; Maitusong, B.; Ma, Y.-T.; Fu, Z.-Y.; Yang, Y.-N.; Ma, X.; Li, X.-M.; Liu, F.; Chen, B.-D. Acyl-CoA: Cholesterol acyltransferases-2 gene polymorphism is associated with increased susceptibility to coronary artery disease in Uygur population in Xinjiang, China. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Carvalho, T.B.; Padovani, C.R.; de Oliveira Junior, L.R.; Latini, A.C.P.; Kurokawa, C.S.; Pereira, P.C.M.; Dos Santos, R.M. ACAT-1 gene rs1044925 SNP and its relation with different clinical forms of chronic Chagas disease. Parasitol. Res. 2019, 118, 2343–2351. [Google Scholar] [CrossRef]
  92. Huang, L.-H.; Nishi, K.; Li, S.; Ho, T.; Dong, R.; Chang, C.C.Y.; Chang, T.-Y. Acyl-coenzyme A:cholesterol acyltransferase 1—significance of single-nucleotide polymorphism at residue 526 and the role of Pro347 near the fifth transmembrane domain. FEBS J. 2014, 281, 1773–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Klos, K.L.E.; Sing, C.F.; Boerwinkle, E.; Hamon, S.C.; Rea, T.J.; Clark, A.; Fornage, M.; Hixson, J.E. Consistent effects of genes involved in reverse cholesterol transport on plasma lipid and apolipoprotein levels in CARDIA participants. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 1828–1836. [Google Scholar] [CrossRef] [PubMed]
  94. Edmondson, A.C.; Braund, P.S.; Stylianou, I.M.; Khera, A.V.; Nelson, C.P.; Wolfe, M.L.; Derohannessian, S.L.; Keating, B.J.; Qu, L.; He, J.; et al. Dense genotyping of candidate gene loci identifies variants associated with high-density lipoprotein cholesterol. Circ. Cardiovasc. Genet. 2011, 4, 145–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Wollmer, M.A.; Streffer, J.R.; Tsolaki, M.; Grimaldi, L.M.E.; Lütjohann, D.; Thal, D.; von Bergmann, K.; Nitsch, R.M.; Hock, C.; Papassotiropoulos, A. Genetic association of acyl-coenzyme A: Cholesterol acyltransferase with cerebrospinal fluid cholesterol levels, brain amyloid load, and risk for Alzheimer’s disease. Mol. Psychiatry 2003, 8, 635–638. [Google Scholar] [CrossRef] [Green Version]
  96. Papassotiropoulos, A.; Wollmer, M.A.; Tsolaki, M.; Brunner, F.; Molyva, D.; Lütjohann, D.; Nitsch, R.M.; Hock, C. A cluster of cholesterol-related genes confers susceptibility for Alzheimer’s disease. J. Clin. Psychiatry 2005, 66, 940–947. [Google Scholar] [CrossRef]
  97. Huttunen, H.J.; Greco, C.; Kovacs, D.M. Knockdown of ACAT-1 reduces amyloidogenic processing of APP. FEBS Lett. 2007, 581, 1688–1692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Bhattacharyya, R.; Kovacs, D.M. ACAT inhibition and amyloid beta reduction. Biochim. Biophys. Acta 2010, 1801, 960–965. [Google Scholar] [CrossRef] [Green Version]
  99. Murphy, S.R.; Chang, C.C.; Dogbevia, G.; Bryleva, E.Y.; Bowen, Z.; Hasan, M.T.; Chang, T.-Y. Acat1 knockdown gene therapy decreases amyloid-β in a mouse model of Alzheimer’s disease. Mol. Ther. 2013, 21, 1497–1506. [Google Scholar] [CrossRef]
  100. Bryleva, E.Y.; Rogers, M.A.; Chang, C.C.Y.; Buen, F.; Harris, B.T.; Rousselet, E.; Seidah, N.G.; Oddo, S.; LaFerla, F.M.; Spencer, T.A.; et al. ACAT1 gene ablation increases 24(S)-hydroxycholesterol content in the brain and ameliorates amyloid pathology in mice with AD. Proc. Natl. Acad. Sci. USA 2010, 107, 3081–3086. [Google Scholar] [CrossRef] [Green Version]
  101. Naelitz, B.D.; Sharifi, N. Through the Looking-Glass: Reevaluating DHEA Metabolism Through HSD3B1 Genetics. Trends Endocrinol. Metab. 2020, 31, 680–690. [Google Scholar] [CrossRef]
  102. Jansen, P.R.; Watanabe, K.; Stringer, S.; Skene, N.; Bryois, J.; Hammerschlag, A.R.; de Leeuw, C.A.; Benjamins, J.S.; Muñoz-Manchado, A.B.; Nagel, M.; et al. Genome-wide analysis of insomnia in 1,331,010 individuals identifies new risk loci and functional pathways. Nat. Genet. 2019, 51, 394–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Ruth, K.S.; Day, F.R.; Tyrrell, J.; Thompson, D.J.; Wood, A.R.; Mahajan, A.; Beaumont, R.N.; Wittemans, L.; Martin, S.; Busch, A.S.; et al. Using human genetics to understand the disease impacts of testosterone in men and women. Nat. Med. 2020, 26, 252–258. [Google Scholar] [CrossRef] [PubMed]
  104. Vujkovic, M.; Keaton, J.M.; Lynch, J.A.; Miller, D.R.; Zhou, J.; Tcheandjieu, C.; Huffman, J.E.; Assimes, T.L.; Lorenz, K.; Zhu, X.; et al. Discovery of 318 new risk loci for type 2 diabetes and related vascular outcomes among 1.4 million participants in a multi-ancestry meta-analysis. Nat. Genet. 2020, 52, 680–691. [Google Scholar] [CrossRef]
  105. Zhao, B.; Zhang, J.; Ibrahim, J.G.; Luo, T.; Santelli, R.C.; Li, Y.; Li, T.; Shan, Y.; Zhu, Z.; Zhou, F.; et al. Large-scale GWAS reveals genetic architecture of brain white matter microstructure and genetic overlap with cognitive and mental health traits (n = 17,706). Mol. Psychiatry 2019, 1–13. [Google Scholar] [CrossRef]
  106. Yan, Q.; Ding, Y.; Liu, Y.; Sun, T.; Fritsche, L.G.; Clemons, T.; Ratnapriya, R.; Klein, M.L.; Cook, R.J.; Liu, Y.; et al. Genome-wide analysis of disease progression in age-related macular degeneration. Hum. Mol. Genet. 2018, 27, 929–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Table 1. Pre-clinical studies of the main ACAT inhibitors for atherosclerosis.
Table 1. Pre-clinical studies of the main ACAT inhibitors for atherosclerosis.
NameAnimalPlasma CholesterolAtherosclerotic Lesion EffectOther EffectsReference
F-1394RatsN.D.↓ cholesterol absorption[31]
Apoe−/− ldlr−/− miceN.S. [32]
Rabbits (balloon injury)N.S.N.D.↓ neointimal thickening[33]
Apoe−/− mice [34]
Apoe−/− miceN.S.↓ progression ↓ lesion tissue factor[35]
Apoe−/− miceN.S.N.S. [36]
Apoe−/− and human APO A1-transgenic miceN.S.N.S. [36]
CI-1011 (avasimibe)Male monkeysN.D.↓ Lp(a)[37]
Miniature pigsN.D. [38]
Male rabbitsN.S.↓ progression↓ lesion MMP expression[39]
Beagle dogsN.D.N.D.↑ Emesis, saliva, hepatic toxicity,[40]
ApoE *3-Leiden mice [41]
Miniature pigsN.D. [42]
Apoe−/− mice↓ progression [43]
RabbitsN.D.↓ progression [44]
CS-505 (pactimibe sulfate)Apoe−/− mice↓ progression↓ MMP expression[43]
RabbitsN.S.N.S.↑ lesion fibers and smooth muscle cells[45]
Hamsters [46]
HamstersN.D.↓ hepatic lipids; ↓ lipid absorption; ↑ fecal lipid excretion[47]
MonkeysN.D. [47]
Diabetic ratsN.D.N.D.↓ postprandial fat loading[47]
Apoe−/− mice [48]
CI-976RabbitsN.S.↓ progression [49]
HamstersN.D. [50]
RatsN.D.↓ liver CE[51]
MicropigsN.S.N.D.↓ liver CE[52]
Endogenous hypercholesterolemia rabbitsN.D. [53]
Rabbits [54]
Nephrotic syndrome (NS) ratsN.D.restore LDL and HDL receptors[55]
K-604 (ACAT-1 selective)Hamsters↓ (at high dose) [56]
Apoe−/− mice↓ (at high dose)N.S.↑ lesion collagen[48]
PD 132301-2RatsN.D. [57]
Guinea pigsN.D. [57]
RabbitsN.D. [57]
CL 277,082RatsN.D. [57]
Guinea pigs↓ (at high dose)N.D. [57]
RabbitsN.D. [57]
RatsN.D. [51]
N.D., not determined; N.S., not significantly different; MMP, matrix metalloproteinases.
Table 2. GWAS findings for human SOAT1/2 genes.
Table 2. GWAS findings for human SOAT1/2 genes.
GeneVariantp-ValueReported TraitReference
SOAT1rs133067286 × 10−14Morningness[102]
rs21523182 × 10−27Bioavailable testosterone levels[103]
rs22489792 × 10−12Bioavailable testosterone levels[103]
rs5694218851 × 10−62Total testosterone levels[103]
rs5694218853 × 10−37Total testosterone levels[103]
rs28161777 × 10−9Type 2 diabetes[104]
rs675632842 × 10−8White matter microstructure[105]
SOAT2rs111704177 × 10−6Age-related macular degeneration progression[106]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hai, Q.; Smith, J.D. Acyl-Coenzyme A: Cholesterol Acyltransferase (ACAT) in Cholesterol Metabolism: From Its Discovery to Clinical Trials and the Genomics Era. Metabolites 2021, 11, 543. https://doi.org/10.3390/metabo11080543

AMA Style

Hai Q, Smith JD. Acyl-Coenzyme A: Cholesterol Acyltransferase (ACAT) in Cholesterol Metabolism: From Its Discovery to Clinical Trials and the Genomics Era. Metabolites. 2021; 11(8):543. https://doi.org/10.3390/metabo11080543

Chicago/Turabian Style

Hai, Qimin, and Jonathan D. Smith. 2021. "Acyl-Coenzyme A: Cholesterol Acyltransferase (ACAT) in Cholesterol Metabolism: From Its Discovery to Clinical Trials and the Genomics Era" Metabolites 11, no. 8: 543. https://doi.org/10.3390/metabo11080543

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop