Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Anticancer Agents Med Chem ; 22(5): 897-904, 2022.
Article in English | MEDLINE | ID: mdl-34353273

ABSTRACT

BACKGROUND AND OBJECTIVE: Beauvericin (BEA), a cyclic hexadepsipeptide mycotoxin, is a potent inhibitor of the acyl-CoA: cholesterol acyltransferase enzyme 1 (ACAT1), involved in multiple tumor-correlated pathways. However, the binding mechanisms between BEA and ACAT1 were not elucidated. METHODS: BEA was purified from a mangrove entophytic Fusarium sp. KL11. Single-crystal X-ray diffraction was used to determine the structure of BEA. Wound healing assays of BEA against KB cell line and MDA-MB-231 cell line were evaluated. Inhibitory potency of BEA against ACAT1 was determined by ELISA assays. Molecular docking was carried out to illuminate the bonding mechanism between BEA and ACAT1. RESULTS: The structure of BEA was confirmed by X-ray diffraction, indicating a monoclinic crystal system with P21 space group (α = 90°, ß = 92.2216(9)°, γ= 90°). BEA displayed migration-inhibitory activities against KB cells and MDA-MB-231 cells In Vitro. ELISA assays revealed that the protein expression level of ACAT1 in KB cells was significantly decreased after BEA treatment (P <0.05). Molecular docking demonstrated that BEA formed hydrogen bond with His425 and pi-pi staking with Tyr429 in ACAT1. CONCLUSION: BEA sufficiently inhibited the proliferation and migration of KB cells and MDA-MB-231 cells by downregulating ACAT1 expression. In addition, BEA potentially possessed a strong binding affinity with ACAT1. BEA may serve as a potential lead compound for the development of a new ACAT1-targeted anticancer drug.


Subject(s)
Acetyl-CoA C-Acetyltransferase , Depsipeptides , Mycotoxins , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/biosynthesis , Depsipeptides/chemistry , Depsipeptides/pharmacology , Humans , KB Cells , Molecular Docking Simulation , Mycotoxins/pharmacology
2.
Mol Pharmacol ; 99(3): 175-183, 2021 03.
Article in English | MEDLINE | ID: mdl-33384285

ABSTRACT

Acyl-CoA:cholesterol acyltransferase (ACAT) mediates cellular cholesterol esterification. In atherosclerotic plaque macrophages, ACAT promotes cholesteryl ester accumulation, resulting in foam cell formation and atherosclerosis progression. Its complete inactivation in mice, however, showed toxic effects because of an excess of free cholesterol (FC) in macrophages, which can cause endoplasmic reticulum stress, cholesterol crystal formation, and inflammasome activation. Our previous studies showed that long-term partial ACAT inhibition, achieved by dietary supplementation with Fujirebio F1394, delays atherosclerosis progression in apoprotein E-deficient (Apoe -/-) mice by reducing plaque foam cell formation without inflammatory or toxic effects. Here, we determined whether short-term partial inhibition of ACAT, in combination with an enhanced systemic FC acceptor capacity, has synergistic benefits. Thus, we crossbred Apoe -/- with human apoprotein A1-transgenic (APOA1 tg/tg) mice, which have elevated cholesterol-effluxing high-density lipoprotein particles, and subjected Apoe -/- and APOA1 tg/tg/Apoe -/- mice to an atherogenic diet to develop advanced plaques. Then mice were either euthanized (baseline) or fed purified standard diet with or without F1394 for 4 more weeks. Plaques of APOA1 tg/tg/Apoe -/- mice fed F1394 showed a 60% reduction of macrophages accompanied by multiple other benefits, such as reduced inflammation and favorable changes in extracellular composition, in comparison with Apoe -/- baseline mice. In addition, there was no accumulation of cholesterol crystals or signs of toxicity. Overall, these results show that short-term partial ACAT inhibition, coupled to increased cholesterol efflux capacity, favorably remodels atherosclerosis lesions, supporting the potential of these combined therapies in the treatment of advanced atherosclerosis. SIGNIFICANCE STATEMENT: Short-term pharmacological inhibition of acyl-CoA:cholesterol acyltransferase-mediated cholesterol esterification, in combination with increased free cholesterol efflux acceptors, has positive effects in mice by 1) reducing the inflammatory state of the plaque macrophages and 2) favoring compositional changes associated with plaque stabilization. These effects occur without toxicity, showing the potential of these combined therapies in the treatment of advanced atherosclerosis.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Apolipoprotein A-I/genetics , Apolipoproteins E/genetics , Atherosclerosis/therapy , Cyclohexanes/administration & dosage , Dioxanes/administration & dosage , Animals , Atherosclerosis/genetics , Breeding , Cyclohexanes/pharmacology , Dietary Supplements , Dioxanes/pharmacology , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Genetic Markers/drug effects , Humans , Lipoproteins, HDL/blood , Male , Mice , Mice, Knockout , Mice, Transgenic , Treatment Outcome
3.
Nutrients ; 12(10)2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33050310

ABSTRACT

The present study aimed to determine the effect of an ethyl acetate extract of Mikania micrantha stems (EAMMS) in hypercholesterolemia-induced rats. Rats were divided into a normal group (NC) and hypercholesterolemia induced groups: hypercholesterolemia control group (PC), simvastatin group (SV) (10 mg/kg) and EAMMS extract groups at different dosages of 50, 100 and 200 mg/kg, respectively. Blood serum and tissues were collected for haematological, biochemical, histopathological, and enzyme analysis. Total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), aspartate aminotransferase (AST), alanine aminotransferase (ALT), urea, creatinine, malondialdehyde (MDA) level, as well as enzymes of HMG-CoA reductase (HMGCR) and acetyl-CoA acetyltransferase 2 (ACAT2), were measured. Feeding rats with high cholesterol diet for eight weeks resulted in a significantly (p < 0.05) increased of TC, TG, LDL-C, AST, ALT and MDA levels. Meanwhile, the administration of EAMMS extract (50, 100 and 200 mg/kg) and simvastatin (10 mg/kg) significantly reduced (p < 0.05) the levels of TC, TG, LDL-C and MDA compared to rats in the PC group. Furthermore, all EAMMS and SV-treated groups showed a higher HDL-C level compared to both NC and PC groups. No significant difference was found in the level of ALT, AST, urea and creatinine between the different dosages in EAMMS extracts. Treatment with EAMMS also exhibited the highest inhibition activity of enzyme HMGCR and ACAT2 as compared to the control group. From the histopathological examination, liver tissues in the PC group showed severe steatosis than those fed with EAMMS and normal diet. Treatment with EAMMS extract ameliorated and reduced the pathological changes in the liver. No morphological changes showed in the kidney structure of both control and treated groups. In conclusion, these findings demonstrated that EAMMS extract has anti-hypercholesterolemia properties and could be used as an alternative treatment for this disorder.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/metabolism , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/adverse effects , Diet, High-Fat/adverse effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Hypercholesterolemia/drug therapy , Lipid Peroxidation/drug effects , Mikania/chemistry , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Animals , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hypercholesterolemia/etiology , Male , Plant Extracts/isolation & purification , Rats, Sprague-Dawley
4.
Appl Environ Microbiol ; 86(7)2020 03 18.
Article in English | MEDLINE | ID: mdl-32005728

ABSTRACT

Ergosterol plays an important role in maintaining cell membrane sterol homeostasis in fungi, and as such, it is considered an effective target in antifungal chemotherapy. In yeast, the enzyme acetyl-coenzyme A (CoA) acetyltransferase (ERG10) catalyzes the Claisen condensation of two acetyl-CoA molecules to acetoacetyl-CoA in the ergosterol biosynthesis pathway and is reported as being critical for cell viability. Using yeast ERG10 for alignment, two orthologues, AfERG10A (AFUB_000550) and AfERG10B (AFUB_083570), were discovered in the opportunistic fungal pathogen Aspergillus fumigatus Despite the essentiality of AfERG10B having been previously validated, the biological function of AfERG10A remains unclear. In this study, we have characterized recombinant AfERG10A as a functional acetyl-CoA acetyltransferase catalyzing both synthetic and degradative reactions. Unexpectedly, AfERG10A localizes to the mitochondria in A. fumigatus, as shown by C-terminal green fluorescent protein (GFP) tag fusion. Both knockout and inducible promoter strategies demonstrate that Aferg10A is essential for the survival of A. fumigatus The reduced expression of Aferg10A leads to severe morphological defects and increased susceptibility to oxidative and cell wall stresses. Although the catalytic mechanism of acetyl-CoA acetyltransferase family is highly conserved, the crystal structure of AfERG10A and its complex with CoA are solved, revealing four substitutions within the CoA binding site that are different from human orthologues. Taken together, our combination of genetic and structural studies demonstrates that mitochondrial AfERG10A is essential for A. fumigatus cell viability and could be a potential drug target to feed the antifungal drug development pipeline.IMPORTANCE A growing number of people worldwide are suffering from invasive aspergillosis caused by the human opportunistic fungal pathogen A. fumigatus Current therapeutic options rely on a limited repertoire of antifungals. Ergosterol is an essential component of the fungal cell membrane as well as a target of current antifungals. Approximately 20 enzymes are involved in ergosterol biosynthesis, of which acetyl-CoA acetyltransferase (ACAT) is the first enzyme. Two ACATs in A. fumigatus are AfErg10A and AfErg10B. However, the biological function of AfErg10A is yet to be investigated. In this study, we showed that AfErg10A is localized in the mitochondria and is essential for A. fumigatus survival and morphological development. In combination with structural studies, we validated AfErg10A as a potential drug target that will facilitate the development of novel antifungals and improve the efficiency of existing drugs.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Antifungal Agents/pharmacology , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/genetics , Gene Expression Regulation, Fungal/drug effects , Mitochondrial Proteins/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Aspergillus fumigatus/enzymology , Ergosterol/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
J Biol Chem ; 295(9): 2839-2849, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31980458

ABSTRACT

Understanding the mechanisms by which viruses evade host cell immune defenses is important for developing improved antiviral therapies. In an unusual twist, human cytomegalovirus co-opts the antiviral radical SAM enzyme viperin (virus-inhibitory protein, endoplasmic reticulum-associated, interferon-inducible) to enhance viral infectivity. This process involves translocation of viperin to the mitochondrion, where it binds the ß-subunit (HADHB) of the mitochondrial trifunctional enzyme complex that catalyzes thiolysis of ß-ketoacyl-CoA esters as part of fatty acid ß-oxidation. Here we investigated how the interaction between these two enzymes alters their activities and affects cellular ATP levels. Experiments with purified enzymes indicated that viperin inhibits the thiolase activity of HADHB, but, unexpectedly, HADHB activates viperin, leading to synthesis of the antiviral nucleotide 3'-deoxy-3',4'-didehydro-CTP. Measurements of enzyme activities in lysates prepared from transfected HEK293T cells expressing these enzymes mirrored the findings obtained with purified enzymes. Thus, localizing viperin to mitochondria decreased thiolase activity, and coexpression of HADHB significantly increased viperin activity. Furthermore, targeting viperin to mitochondria also increased the rate at which HADHB is retrotranslocated out of mitochondria and degraded, providing an additional mechanism by which viperin reduces HADHB activity. Targeting viperin to mitochondria decreased cellular ATP levels by more than 50%, consistent with the enzyme disrupting fatty acid catabolism. These results provide biochemical insight into the mechanism by which human cytomegalovirus subverts viperin; they also provide a biochemical rationale for viperin's recently discovered role in regulating thermogenesis in adipose tissues.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Mitochondria/metabolism , Proteins/metabolism , Adenosine Triphosphate/metabolism , Cytomegalovirus/physiology , HEK293 Cells , Humans , Immune Evasion , Mitochondrial Trifunctional Protein, beta Subunit/antagonists & inhibitors , Mitochondrial Trifunctional Protein, beta Subunit/metabolism , Mitochondrial Trifunctional Protein, beta Subunit/physiology , Oxidoreductases Acting on CH-CH Group Donors
6.
Neuron ; 105(5): 837-854.e9, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31902528

ABSTRACT

Loss-of-function (LOF) variants of TREM2, an immune receptor expressed in microglia, increase Alzheimer's disease risk. TREM2 senses lipids and mediates myelin phagocytosis, but its role in microglial lipid metabolism is unknown. Combining chronic demyelination paradigms and cell sorting with RNA sequencing and lipidomics, we find that wild-type microglia acquire a disease-associated transcriptional state, while TREM2-deficient microglia remain largely homeostatic, leading to neuronal damage. TREM2-deficient microglia phagocytose myelin debris but fail to clear myelin cholesterol, resulting in cholesteryl ester (CE) accumulation. CE increase is also observed in APOE-deficient glial cells, reflecting impaired brain cholesterol transport. This finding replicates in myelin-treated TREM2-deficient murine macrophages and human iPSC-derived microglia, where it is rescued by an ACAT1 inhibitor and LXR agonist. Our studies identify TREM2 as a key transcriptional regulator of cholesterol transport and metabolism under conditions of chronic myelin phagocytic activity, as TREM2 LOF causes pathogenic lipid accumulation in microglia.


Subject(s)
Brain/metabolism , Cholesterol/metabolism , Macrophages/metabolism , Membrane Glycoproteins/genetics , Microglia/metabolism , Myelin Sheath/metabolism , Phagocytosis/genetics , Receptors, Immunologic/genetics , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cholesterol Esters/metabolism , Disease Models, Animal , Flow Cytometry , Humans , Induced Pluripotent Stem Cells , Lipid Metabolism/genetics , Lipidomics , Liver X Receptors/agonists , Mice , Mice, Knockout , Mice, Knockout, ApoE , RNA-Seq
7.
Curr Alzheimer Res ; 16(8): 699-709, 2019.
Article in English | MEDLINE | ID: mdl-31441726

ABSTRACT

BACKGROUND: Alzheimer´s disease (AD) is a chronic and progressive disease which impacts caregivers, families and societies physically, psychologically and economically. Currently available drugs can only improve cognitive symptoms, have no impact on progression and are not curative, so identifying and studying new drug targets is important. There are evidences which indicate disturbances in cholesterol homeostasis can be related with AD pathology, especially the compartmentation of intracellular cholesterol and cytoplasmic cholesterol esters formed by acyl-CoA: cholesterol acyltransferase 1 (ACAT1) can be implicated in the regulation of amyloid-beta (Aß) peptide, involved in AD. Blocking ACAT1 activity, beneficial effects are obtained, so it has been suggested that ACAT1 can be a potential new therapeutic target. The present review discusses the role of cholesterol homeostasis in AD pathology, especially with ACAT inhibitors, and how they have been raised as a therapeutic approach. In addition, the genetic relationship of ACAT and AD is discussed. CONCLUSION: Although there are several lines of evidence from cell-based and animal studies that suggest that ACAT inhibition is an effective way of reducing cerebral Aß, there is still an information gap in terms of mechanisms and concerns to cover before passing to the next level. Additionally, an area of interest that may be useful in understanding AD to subsequently propose new therapeutic approaches is pharmacogenetics; however, there is still a lot of missing information in this area.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Alzheimer Disease/metabolism , Animals , Cholesterol/metabolism , Humans
8.
Life Sci ; 232: 116592, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31228515

ABSTRACT

Acetoacetyl-CoA thiolase also known as acetyl-CoA acetyltransferase (ACAT) corresponds to two enzymes, one cytosolic (ACAT2) and one mitochondrial (ACAT1), which is thought to catalyse reversible formation of acetoacetyl-CoA from two molecules of acetyl-CoA during ketogenesis and ketolysis respectively. In addition to this activity, ACAT1 is also involved in isoleucine degradation pathway. Deficiency of ACAT1 is an inherited metabolic disorder, which results from a defect in mitochondrial acetoacetyl-CoA thiolase activity and is clinically characterized with patients presenting ketoacidosis. In this review I discuss the recent findings, which unexpectedly expand the known functions of ACAT1, indicating a role for ACAT1 well beyond its classical activity. Indeed ACAT1 has recently been shown to possess an acetyltransferase activity capable of specifically acetylating Pyruvate DeHydrogenase (PDH), an enzyme involved in producing acetyl-CoA. ACAT1-dependent acetylation of PDH was shown to negatively regulate this enzyme with a consequence in Warburg effect and tumor growth. Finally, the elevated ACAT1 enzyme activity in diverse human cancer cell lines was recently reported. These important novel findings on ACAT1's function and expression in cancer cell proliferation point to ACAT1 as a potential new anti-cancer target.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Neoplasms/enzymology , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Cytosol/enzymology , Humans , Mitochondria/enzymology , Molecular Targeted Therapy , Neoplasms/drug therapy , Protein Processing, Post-Translational , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase 2
9.
Arch Biochem Biophys ; 671: 103-110, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31251920

ABSTRACT

Cholesterol is an important lipid molecule and is needed for all mammalian cells. In various cell types, excess cholesterol is stored as cholesteryl esters; acyl-CoA:cholesterol acyltransferase 1 (ACAT1) plays an essential role in this storage process. ACAT1 is located at the endoplasmic reticulum and has nine transmembrane domains (TMDs). It is a member of the membrane-bound O-acyltransferase (MBOAT) family, in which members contain multiple TMDs and participate in a variety of biological functions. When solubilized in the zwitterionic detergent CHAPS, ACAT1 can be purified to homogeneity with full enzyme activity and behaves as a homotetrameric protein. ACAT1 contains two dimerization motifs. The first motif is located near the N-terminus and is not conserved in MBOATs. Deletion of the N-terminal dimerization domain converts ACAT1 to a dimer with full catalytic activity; therefore, ACAT1 is a two-fold dimer. The second dimerization domain, located near the C-terminus, is conserved in MBOATs; however, it was not known whether the C-terminal dimerization domain is required for enzyme activity. Here we show that treating ACAT1 with non-ionic detergent, Triton X-100 or octyl glucoside, causes the enzyme to become a two-fold monomer without any enzymatic activity. Detergent exchange of Triton X-100 with CHAPS restores ACAT1 to a two-fold dimer but fails to restore its enzymatic activity. These results implicate that ACAT1 requires hydrophobic subunit interactions near the C-terminus in order to remain active as a two-fold dimer. Our results also caution the use of Triton X-100 or octyl glucoside to purify other MBOATs.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Detergents/chemistry , Enzyme Inhibitors/chemistry , Glucosides/chemistry , Octoxynol/chemistry , Protein Multimerization/drug effects , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , CHO Cells , Cholic Acids/chemistry , Cricetulus , Goats , HEK293 Cells , Humans , Mice , Rabbits
10.
J Med Chem ; 61(23): 10635-10650, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30433781

ABSTRACT

2-(4-(2-((1 H-Benzo[ d]imidazol-2-yl)thio)ethyl)piperazin-1-yl)- N-(6-methyl-2,4-bis(methylthio)pyridin-3-yl)acetamide hydrochloride (K-604, 2) has been identified as an aqueous-soluble potent inhibitor of human acyl-coenzyme A:cholesterol O-acyltransferase (ACAT, also known as SOAT)-1 that exhibits 229-fold selectivity for human ACAT-1 over human ACAT-2. In our molecular design, the insertion of a piperazine unit in place of a 6-methylene chain in the linker between the head (pyridylacetamide) and tail (benzimidazole) moieties led to a marked enhancement of the aqueous solubility (up to 19 mg/mL at pH 1.2) and a significant improvement of the oral absorption (the Cmax of 2 was 1100-fold higher than that of 1 in fasted dogs) compared with those of the previously selected compound, 1. After ensuring the pharmacological effects and safety, we designated 2 as a clinical candidate, named K-604. Considering the therapeutic results of ACAT inhibitors in past clinical trials, we believe that K-604 will be useful for the treatment of incurable diseases involving ACAT-1 overexpression.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Drug Discovery , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Water/chemistry , Animals , Benzimidazoles/pharmacokinetics , Cell Line , Enzyme Inhibitors/pharmacokinetics , Humans , Rabbits , Solubility , Structure-Activity Relationship , Tissue Distribution
11.
Nanomedicine ; 14(8): 2541-2550, 2018 11.
Article in English | MEDLINE | ID: mdl-30193815

ABSTRACT

Cholesterol facilitated the formation of T cell receptor on cytotoxic CD8+ T lymphocytes (CTLs). However, the activation of CD8+ T cells always resulted in the upregulation of acetyl-CoA acetyltransferase-1 (ACAT-1) and enhanced the esterification of cholesterol. To relieve the suppression on CD8+ T cells, an ACAT-1 inhibitor avasimibe was combined with chemo-immunotherapy. Paclitaxel and immunoadjuvant αGC were co-encapsulated in liposomes modified with pH sensitive TH peptide (PTX/αGC-TH-Lip). After intravenous injections, the combination of avasimibe significantly elevated the free cholesterol level and relieved the inhibition of CD8+ T cells caused by PTX/αGC-TH-Lip, leading to enhanced CTL responses and anti-tumor effects of PTX/αGC-TH-Lip in B16F10 melanoma xenograft and lung metastasis models. The adoptive immunotherapy further confirmed the enhanced anti-tumor immune responses of the combined strategy. The combination of avasimibe and PTX/αGC-TH-Lip was proven as a feasible approach to enhance the antitumor effects of chemo-immunotherapy by relieving the inhibition of CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cholesterol/metabolism , Immunotherapy, Adoptive , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Paclitaxel/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Acetamides , Acetates/pharmacology , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/chemistry , Cells, Cultured , Cholesterol/chemistry , Cytochrome P-450 CYP3A Inducers/pharmacology , Esterification , Female , Humans , Liposomes/administration & dosage , Liposomes/chemistry , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Sulfonamides , Sulfonic Acids/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/pathology
12.
Lipids Health Dis ; 17(1): 122, 2018 May 24.
Article in English | MEDLINE | ID: mdl-29793481

ABSTRACT

BACKGROUND: Insulin resistant and the progression of cancer is closely related. The aim of this study was to  investigate the effect of insulin on the proliferation and migration of colon cancer cells and its underlying mechanism. METHODS: Colon carcinoma tissues from the 80 cases of colon cancer patients were collected. Immunohistochemistry was used to detect the expression of acyl coenzyme A: cholesterol acyltransferase1 (ACAT1), and we analyzed the correlation between hyperglycemia and ACAT1, hyperglycemia and metastasis. CCK8 assay and transwell assay were used to investigate the effect of different concentrations of insulin and ACAT1siRNA on human colon cancer cell line HT-29. ACAT1 mRNA expression and protein level in HT-29 cells were determined by real-time quantitative PCR and western blotting, respectively. RESULTS: Biopsies from patients with colon carcinoma showed hyperglycemia links ACAT1, lymph nodes metastasis and distant metastasis. Insulin markedly promoted cell proliferation and migration in human colon cancer HT-29 cells. Moreover, ACAT1mRNA expression and protein level were increased by insulin. ACAT1siRNA resulted in a complete inhibition of the ACAT1 mRNA expression. Consequently insulin-triggered cell proliferation and migration on colon cancer cells were inhibited. CONCLUSION: The progression of colon cancer has a positive correlation with hyperinsulinemia. Insulin-triggered cell proliferation and metastatic effects on colorectal cancer cells are mediated by ACAT1. Therefore, insulin could promote colon cancer progression by upregulation of ACAT1, which maybe is a potential therapeutic target for colon cancer.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hyperglycemia/genetics , Hyperinsulinism/genetics , Insulin/pharmacology , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/metabolism , Cell Movement , Cell Proliferation , Cholesterol/metabolism , Colonic Neoplasms/complications , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Disease Progression , Female , HT29 Cells , Humans , Hyperglycemia/complications , Hyperglycemia/enzymology , Hyperglycemia/pathology , Hyperinsulinism/complications , Hyperinsulinism/enzymology , Hyperinsulinism/pathology , Insulin/metabolism , Lymphatic Metastasis , Male , Middle Aged , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
13.
BMC Endocr Disord ; 18(1): 24, 2018 May 02.
Article in English | MEDLINE | ID: mdl-29720169

ABSTRACT

BACKGROUND: Cushing's syndrome in humans shares many similarities with its counterpart in dogs in terms of etiology (pituitary versus adrenal causes), clinical signs, and pathophysiologic sequelae. In both species, treatment of pituitary- and adrenal-dependent disease is met with limitations. ATR-101, a selective inhibitor of ACAT1 (acyl coenzyme A:cholesterol acyltransferase 1), is a novel small molecule therapeutic currently in clinical development for the treatment of adrenocortical carcinoma, congenital adrenal hyperplasia, and Cushing's syndrome in humans. Previous studies in healthy dogs have shown that ATR-101 treatment led to rapid, dose-dependent decreases in adrenocorticotropic hormone (ACTH) stimulated cortisol levels. The purpose of this clinical study was to investigate the effects of ATR-101 in dogs with Cushing's syndrome. METHODS: ATR-101 pharmacokinetics and activity were assessed in 10 dogs with naturally-occurring Cushing's syndrome, including 7 dogs with pituitary-dependent disease and 3 dogs with adrenal-dependent disease. ATR-101 was administered at 3 mg/kg PO once daily for one week, followed by 30 mg/kg PO once daily for one (n = 4) or three (n = 6) weeks. Clinical, biochemical, adrenal hormonal, and pharmacokinetic data were obtained weekly for study duration. RESULTS: ATR-101 exposure increased with increasing dose. ACTH-stimulated cortisol concentrations, the primary endpoint for the study, were significantly decreased with responders (9 of 10 dogs) experiencing a mean ± standard deviation reduction in cortisol levels of 50 ± 17% at study completion. Decreases in pre-ACTH-stimulated cortisol concentrations were observed in some dogs although overall changes in pre-ACTH cortisol concentrations were not significant. The compound was well-tolerated and no serious drug-related adverse effects were reported. CONCLUSIONS: This study highlights the potential utility of naturally occurring canine Cushing's syndrome as a model for human disease and provides proof of concept for ATR-101 as a novel agent for the treatment of endocrine disorders like Cushing's syndrome in humans.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Adrenocorticotropic Hormone/metabolism , Cushing Syndrome/veterinary , Dog Diseases/metabolism , Hydrocortisone/metabolism , Phenylurea Compounds/pharmacology , Animals , Cushing Syndrome/drug therapy , Cushing Syndrome/metabolism , Cushing Syndrome/pathology , Dogs , Female , Male , Phenylurea Compounds/pharmacokinetics , Tissue Distribution
14.
J Immunother ; 41(2): 45-52, 2018.
Article in English | MEDLINE | ID: mdl-29252915

ABSTRACT

Chimeric antigen receptor-modified T cell (CART) therapy has been demonstrated to have significant effect on hematologic tumor in patients. However, many persistent obstacles and challenges still limit the application. It is known that CD8 T cells are a key component of antitumor immunity. An avasimibe-induced inhibition of cholesterol esterification has been shown to improve the antitumor response of CD8 T cells in mice. In this study, using human CD19-directed CART cells as effector cells and CD19-overexpressing K562 cells as target cells, we detected whether cholesterol acyltransferase inhibition by avasimibe can enhance the antitumor effect of human CART cells. After avasimibe treatment, the infection rate was dropped by up to 50% (P<0.05). The cytotoxic effect of CART cells was significantly increased than the control group in a dose-dependent manner. Moreover, the level of secreted interferon-γ increased in almost half of the cases (P<0.05); the ratio of CD8CD4 T cells was increased among the total T cells and the CART cells in some of cases (P<0.05). Our study suggests that inhibition of cholesterol acyltransferase can promote the antitumor effect of CART cells, and provides a new option for a combination therapy by regulating T-cell metabolism to enhance antitumor effects.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Animals , Antigens, CD19/immunology , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Disease Models, Animal , Humans , Immunotherapy, Adoptive , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays
15.
Med Sci Monit ; 23: 4471-4476, 2017 Sep 17.
Article in English | MEDLINE | ID: mdl-28918429

ABSTRACT

BACKGROUND The main purpose of this study was to explore the antitumor effect and mechanisms of ACAT1 inhibitor combined with CSCs-DC vaccine. MATERIAL AND METHODS We isolated HNSCC CSCs and gained CSCs antigens, then used CSCs antigens to load dendritic cells (DC) and generated a CSCs-DC vaccine. We treated mice after surgical excision of established SCC7 tumors with CSCs-DC vaccine and/or ACAT1 inhibitor, and recorded local tumor relapse and host survival. T cells and B cells were harvested from mice treated with CSCs-DC vaccine and/or ACAT1 inhibitor. We tested antibody production and the death rate of CSCs killed by T cells. RESULTS The tumors in the combined treatment group were smaller than in all other groups (P<0.01). The average survival time of the combined treatment group was 82 days and was the longest of all groups. Analysis of IgG levels secreted by B cell and CTL activity in spleens of mice found that results of the combined treatment group were the highest, and the results of the CSCs-DC group were lower than in the combined treatment group. The ACAT1 inhibitor group results were lower than in the CSCs-DC group and the combined treatment group results, but higher than in the PBS group, and the difference was statistically significant. CONCLUSIONS ACAT1 inhibitor enhanced the therapeutic effect of CSCs-DC vaccine in the treatment of the mouse HNSCC postoperative recurrence model. ACAT1 may play an important role in cancer immunotherapy.


Subject(s)
Acetates/pharmacology , Head and Neck Neoplasms/drug therapy , Sulfonic Acids/pharmacology , Acetamides , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Cancer Vaccines/pharmacology , Carcinoma, Squamous Cell , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/immunology , Dendritic Cells/physiology , Disease Models, Animal , Female , Head and Neck Neoplasms/prevention & control , Head and Neck Neoplasms/therapy , Immunotherapy/methods , Mice , Mice, Inbred C3H , Neoplasm Recurrence, Local , Squamous Cell Carcinoma of Head and Neck , Sulfonamides , T-Lymphocytes/immunology , Vaccines/pharmacology
16.
Sci Rep ; 6: 32105, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27601313

ABSTRACT

Selective estrogen receptor modulators (SERMs) are widely prescribed drugs that alter cellular and whole-body cholesterol homeostasis. Here we evaluate the effect of SERMs on the macrophage-specific reverse cholesterol transport (M-RCT) pathway, which is mediated by HDL. Treatment of human and mouse macrophages with tamoxifen, raloxifene or toremifene induced the accumulation of cytoplasmic vesicles of acetyl-LDL-derived free cholesterol. The SERMs impaired cholesterol efflux to apolipoprotein A-I and HDL, and lowered ABCA1 and ABCG1 expression. These effects were not altered by the antiestrogen ICI 182,780 nor were they reproduced by 17ß-estradiol. The treatment of mice with tamoxifen or raloxifene accelerated HDL-cholesteryl ester catabolism, thereby reducing HDL-cholesterol concentrations in serum. When [(3)H]cholesterol-loaded macrophages were injected into mice intraperitoneally, tamoxifen, but not raloxifene, decreased the [(3)H]cholesterol levels in serum, liver and feces. Both SERMs downregulated liver ABCG5 and ABCG8 protein expression, but tamoxifen reduced the capacity of HDL and plasma to promote macrophage cholesterol efflux to a greater extent than raloxifene. We conclude that SERMs interfere with intracellular cholesterol trafficking and efflux from macrophages. Tamoxifen, but not raloxifene, impair M-RCT in vivo. This effect is primarily attributable to the tamoxifen-mediated reduction of the capacity of HDL to promote cholesterol mobilization from macrophages.


Subject(s)
Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Macrophages/drug effects , Selective Estrogen Receptor Modulators/pharmacology , ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter, Subfamily G/biosynthesis , ATP Binding Cassette Transporter, Subfamily G/genetics , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Animals , Apolipoprotein A-I/metabolism , Biological Transport/drug effects , Cholesterol/analysis , Cholesterol/blood , Cholesterol Esters/metabolism , Diet, Western , Esterification/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Feces/chemistry , Fulvestrant , Humans , Lipoproteins, LDL/metabolism , Liver/chemistry , Liver/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Raloxifene Hydrochloride/pharmacology , THP-1 Cells , Tamoxifen/pharmacology , Toremifene/pharmacology
17.
Oncogene ; 35(50): 6378-6388, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27132508

ABSTRACT

Cancer cells are known to execute reprogramed metabolism of glucose, amino acids and lipids. Here, we report a significant role of cholesterol metabolism in cancer metastasis. By using label-free Raman spectromicroscopy, we found an aberrant accumulation of cholesteryl ester in human pancreatic cancer specimens and cell lines, mediated by acyl-CoA cholesterol acyltransferase-1 (ACAT-1) enzyme. Expression of ACAT-1 showed a correlation with poor patient survival. Abrogation of cholesterol esterification, either by an ACAT-1 inhibitor or by shRNA knockdown, significantly suppressed tumor growth and metastasis in an orthotopic mouse model of pancreatic cancer. Mechanically, ACAT-1 inhibition increased intracellular free cholesterol level, which was associated with elevated endoplasmic reticulum stress and caused apoptosis. Collectively, our results demonstrate a new strategy for treating metastatic pancreatic cancer by inhibiting cholesterol esterification.


Subject(s)
Cholesterol Esters/metabolism , Pancreatic Neoplasms/pathology , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/physiology , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Endoplasmic Reticulum Stress , Esterification , Humans , Male , Mice , Neoplasm Metastasis , PTEN Phosphohydrolase/physiology , Pancreatic Neoplasms/metabolism
18.
Nature ; 531(7596): 651-5, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-26982734

ABSTRACT

CD8(+) T cells have a central role in antitumour immunity, but their activity is suppressed in the tumour microenvironment. Reactivating the cytotoxicity of CD8(+) T cells is of great clinical interest in cancer immunotherapy. Here we report a new mechanism by which the antitumour response of mouse CD8(+) T cells can be potentiated by modulating cholesterol metabolism. Inhibiting cholesterol esterification in T cells by genetic ablation or pharmacological inhibition of ACAT1, a key cholesterol esterification enzyme, led to potentiated effector function and enhanced proliferation of CD8(+) but not CD4(+) T cells. This is due to the increase in the plasma membrane cholesterol level of CD8(+) T cells, which causes enhanced T-cell receptor clustering and signalling as well as more efficient formation of the immunological synapse. ACAT1-deficient CD8(+) T cells were better than wild-type CD8(+) T cells at controlling melanoma growth and metastasis in mice. We used the ACAT inhibitor avasimibe, which was previously tested in clinical trials for treating atherosclerosis and showed a good human safety profile, to treat melanoma in mice and observed a good antitumour effect. A combined therapy of avasimibe plus an anti-PD-1 antibody showed better efficacy than monotherapies in controlling tumour progression. ACAT1, an established target for atherosclerosis, is therefore also a potential target for cancer immunotherapy.


Subject(s)
Acetates/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cholesterol/metabolism , Immunotherapy/methods , Melanoma/drug therapy , Melanoma/immunology , Sulfonic Acids/pharmacology , Acetamides , Acetates/therapeutic use , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/deficiency , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Atherosclerosis/drug therapy , CD8-Positive T-Lymphocytes/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Esterification/drug effects , Female , Immunological Synapses/drug effects , Immunological Synapses/immunology , Immunological Synapses/metabolism , Male , Melanoma/metabolism , Melanoma/pathology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , Sulfonamides , Sulfonic Acids/therapeutic use
19.
Future Med Chem ; 7(18): 2451-67, 2015.
Article in English | MEDLINE | ID: mdl-26669800

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia with no cure at present. Cholesterol metabolism is closely associated with AD at several stages. ACAT1 converts free cholesterol to cholesteryl esters, and plays important roles in cellular cholesterol homeostasis. Recent studies show that in a mouse model, blocking ACAT1 provides multiple beneficial effects on AD. Here we review the current evidence that implicates ACAT1 as a therapeutic target for AD. We also discuss the potential usage of various ACAT inhibitors currently available to treat AD.


Subject(s)
Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Alzheimer Disease/drug therapy , Enzyme Inhibitors/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Enzyme Inhibitors/chemistry , Humans , Mice , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism
20.
Mol Med Rep ; 12(4): 6037-42, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26252415

ABSTRACT

Glioblastoma is the most aggressive type of brain tumor and has a poor prognosis. Increased levels of cholesteryl ester and simultaneous expression of acyl­CoA:cholesterol acyltransferase 1 (ACAT1) in tumor cells indicated that cholesterol esterification is critical to tumor growth. The present study confirmed that human glioblastoma tissues as well as the glioblastoma cell line U251­MG showed significant expression of ACAT1. ACAT1 expression in U251­MG cells increased in a cell proliferation­dependent manner. K604, a selective ACAT1 inhibitor, suppressed the proliferation of U251­MG cells and downregulated the activation of Akt and extracellular signal­regulated kinase in proliferating glioblastoma cells. These results suggested that ACAT1 may be a therapeutic target for the treatment of glioblastoma, with K604 as an effective therapeutic agent.


Subject(s)
Acetamides/pharmacology , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Benzimidazoles/pharmacology , Cell Line, Tumor/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...