Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 285
Filter
1.
Cell Death Dis ; 15(5): 325, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724499

ABSTRACT

Cholesterol metabolism reprogramming is one of the significant characteristics of hepatocellular carcinoma (HCC). Cholesterol increases the risk of epithelial-mesenchymal transition (EMT) in cancer. Sterol O-acyltransferases 1 (SOAT1) maintains the cholesterol homeostasis. However, the exact mechanistic contribution of SOAT1 to EMT in HCC remains unclear. Here we demonstrated that SOAT1 positively related to poor prognosis of HCC, EMT markers and promoted cell migration and invasion in vitro, which was mediated by the increased cholesterol in plasmalemma and cholesterol esters accumulation. Furthermore, we reported that SOAT1 disrupted cholesterol metabolism homeostasis to accelerate tumorigenesis and development in HCC xenograft and NAFLD-HCC. Also, we detected that nootkatone, a sesquiterpene ketone, inhibited EMT by targeting SOAT1 in vitro and in vivo. Collectively, our finding indicated that SOAT1 promotes EMT and contributes to hepatocarcinogenesis by increasing cholesterol esterification, which is suppressed efficiently by nootkatone. This study demonstrated that SOAT1 is a potential biomarker and therapeutic target in NAFLD-HCC and SOAT1-targeting inhibitors are expected to be the potential new therapeutic treatment for HCC.


Subject(s)
Carcinoma, Hepatocellular , Cholesterol , Epithelial-Mesenchymal Transition , Liver Neoplasms , Sterol O-Acyltransferase , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Humans , Cholesterol/metabolism , Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase/genetics , Animals , Mice , Male , Mice, Nude , Cell Line, Tumor , Cell Movement , Female , Mice, Inbred BALB C , Sesquiterpenes/pharmacology , Gene Expression Regulation, Neoplastic
2.
J Biochem Mol Toxicol ; 38(6): e23733, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38770938

ABSTRACT

The aim of this investigation was to evaluate the differential expression of the sterol O-acyltransferase 1 (SOAT1) protein in gallbladder cancer tissues and cells, investigate the impact of Avastin on the proliferation, migration, invasion capabilities of gallbladder cancer cells, and its potential to induce cell apoptosis. Immunohistochemical analysis of samples from 145 gallbladder cancer patients was conducted, along with analysis of SOAT1 protein, mRNA expression levels, and cholesterol content in gallbladder cancer cell lines SGC-996, NOZ, and gallbladder cancer (GBC)-SD using Western blot and q-PCR techniques. Furthermore, the effects of Avastin on the proliferation, migration, and invasion capabilities of these gallbladder cancer cell lines were studied, and its ability to induce cell apoptosis was evaluated using flow cytometry, Western blot, and immunohistochemical methods. Additionally, gene expression and pathway analysis were performed, and the synergistic therapeutic effects of Avastin combined with gemcitabine were tested in a gallbladder cancer xenograft model. The study found that SOAT1 expression was significantly upregulated in GBC tissues and positively correlated with lymph node metastasis and TNM staging. In vitro experiments demonstrated that Avastin significantly inhibited the proliferation, migration, and invasion capabilities of SGC-996 and GBC-SD cell lines and induced apoptosis. RNA sequencing analysis revealed multiple differentially expressed genes in cells treated with Avastin, primarily enriched in biological pathways such as signaling transduction, malignant tumors, and the immune system. In vivo, experiments confirmed that Avastin could effectively suppress tumor growth in a gallbladder cancer xenograft model and enhanced the treatment efficacy when used in combination with gemcitabine. Overall, these findings provide new insights and strategies for targeted therapy in gallbladder cancer.


Subject(s)
Gallbladder Neoplasms , Sterol O-Acyltransferase , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/genetics , Humans , Female , Male , Cell Line, Tumor , Animals , Middle Aged , Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase/genetics , Mice , Gemcitabine , Cell Proliferation/drug effects , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Mice, Nude , Apoptosis/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Cell Movement/drug effects , Xenograft Model Antitumor Assays , Aged , Gene Expression Regulation, Neoplastic/drug effects , Mice, Inbred BALB C , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics
3.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38673803

ABSTRACT

Niemann-Pick disease type C1 (NPC1) is a lysosomal disorder due to impaired intracellular cholesterol transport out of the endolysosomal compartment.. Marked heterogeneity has been observed in individuals with the same NPC1 genotype, thus suggesting a significant effect of modifier genes. Prior work demonstrated that decreased SOAT1 activity decreased disease severity in an NPC1 mouse model. Thus, we hypothesized that a polymorphism associated with decreased SOAT1 expression might influence the NPC1 phenotype. Phenotyping and genomic sequencing of 117 individuals with NPC1 was performed as part of a Natural History trial. Phenotyping included determination of disease severity and disease burden. Significant clinical heterogeneity is present in individuals homozygous for the NPC1I1061T variant and in siblings. Analysis of the SOAT1 polymorphism, rs1044925 (A>C), showed a significant association of the C-allele with earlier age of neurological onset. The C-allele may be associated with a higher Annualized Severity Index Score as well as increased frequency of liver disease and seizures. A polymorphism associated with decreased expression of SOAT1 appears to be a genetic modifier of the NPC1 phenotype. This finding is consistent with prior data showing decreased phenotypic severity in Npc1-/-:Soat1-/- mice and supports efforts to investigate the potential of SOAT1 inhibitors as a potential therapy for NPC1.


Subject(s)
Niemann-Pick Disease, Type C , Sterol O-Acyltransferase , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/metabolism , Humans , Male , Female , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Niemann-Pick C1 Protein , Child , Polymorphism, Single Nucleotide , Animals , Mice , Phenotype , Adolescent , Child, Preschool , Genes, Modifier , Adult , Alleles , Severity of Illness Index , Genotype , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Young Adult
4.
Org Lett ; 26(9): 1807-1812, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38393343

ABSTRACT

We have identified the biosynthetic gene cluster (hvm) for the sterol O-acyltransferase inhibitor helvamide (1) from the genome of Aspergillus rugulosus MST-FP2007. Heterologous expression of hvm in A. nidulans produced a previously unreported analog helvamide B (5). An α-ketoglutarate-dependent oxygenase Hvm1 was shown to catalyze intramolecular cyclization of 1 to yield 5. The biosynthetic branch to the related hancockiamides and helvamides was found to be controlled by the substrate selectivity of monomodular nonribosomal peptide synthetases.


Subject(s)
Ketoglutaric Acids , Oxygenases , Oxygenases/genetics , Oxygenases/metabolism , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Cyclization , Multigene Family , Peptide Synthases/metabolism
5.
Int J Mol Sci ; 23(24)2022 Dec 07.
Article in English | MEDLINE | ID: mdl-36555105

ABSTRACT

Metabolic syndrome is associated with the development of chronic kidney disease (CKD). We previously demonstrated that aged kidneys are prone to developing tertiary lymphoid tissues (TLTs) and sustain inflammation after injury, leading to CKD progression; however, the relationship between renal TLT and metabolic syndrome is unknown. In this study, we demonstrated that a high-fat diet (HFD) promoted renal TLT formation and inflammation via sterol O-acyltransferase (SOAT) 1-dependent mechanism. Mice fed a HFD prior to ischemic reperfusion injury (IRI) exhibited pronounced renal TLT formation and sustained inflammation compared to the controls. Untargeted lipidomics revealed the increased levels of cholesteryl esters (CEs) in aged kidneys with TLT formation after IRI, and, consistently, the Soat1 gene expression increased. Treatment with avasimibe, a SOAT inhibitor, attenuated TLT maturation and renal inflammation in HFD-fed mice subjected to IRI. Our findings suggest the importance of SOAT1-dependent CE accumulation in the pathophysiology of CKDs associated with TLT.


Subject(s)
Metabolic Diseases , Metabolic Syndrome , Renal Insufficiency, Chronic , Reperfusion Injury , Animals , Mice , Metabolic Syndrome/metabolism , Diet, High-Fat/adverse effects , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Kidney/metabolism , Lymphoid Tissue/metabolism , Inflammation/metabolism , Fibrosis , Renal Insufficiency, Chronic/metabolism , Metabolic Diseases/metabolism , Reperfusion Injury/metabolism , Mice, Inbred C57BL
6.
J Pharmacol Exp Ther ; 382(3): 356-365, 2022 09.
Article in English | MEDLINE | ID: mdl-35970601

ABSTRACT

Acute lung injury (ALI) is characterized by epithelial damage, barrier dysfunction, and pulmonary edema. Macrophage activation and failure to resolve play a role in ALI; thus, macrophage phenotype modulation is a rational target for therapeutic intervention. Large, lipid-laden macrophages have been observed in various injury models, including intratracheal bleomycin (ITB), suggesting that lipid storage may play a role in ALI severity. The endoplasmic reticulum-associated enzyme acyl coenzyme A acyltransferase-1 (Acat-1/Soat1) is highly expressed in macrophages, where it catalyzes the esterification of cholesterol, leading to intracellular lipid accumulation. We hypothesize that inhibition of Acat-1 will reduce macrophage activation and improve outcomes of lung injury in ITB. K-604, a selective inhibitor of Acat-1, was used to reduce cholesterol esterification and hence lipid accumulation in response to ITB. Male and female C57BL6/J mice (n = 16-21/group) were administered control, control + K-604, ITB, or ITB + K-604 on d0, control or K-604 on d3, and were sacrificed on day 7. ITB caused significant body weight loss and an increase in cholesterol accumulation in bronchoalveolar lavage cells. These changes were mitigated by Acat-1 inhibition. K-604 also significantly reduced ITB-induced alveolar thickening. Surfactant composition was normalized as indicated by a significant decrease in phospholipid: SP-B ratio in ITB+K-604 compared with ITB. K-604 administration preserved mature alveolar macrophages, decreased activation in response to ITB, and decreased the percentage mature and pro-fibrotic interstitial macrophages. These results show that inhibition of Acat-1 in the lung is associated with reduced inflammatory response to ITB-mediated lung injury. SIGNIFICANCE STATEMENT: Acyl coenzyme A acyltransferase-1 (Acat-1) is critical to lipid droplet formation, and thus inhibition of Acat-1 presents as a pharmacological target. Intratracheal administration of K-604, an Acat-1 inhibitor, reduces intracellular cholesterol ester accumulation in lung macrophages, attenuates inflammation and macrophage activation, and normalizes mediators of surface-active function after intratracheal bleomycin administration in a rodent model. The data presented within suggest that inhibition of Acat-1 in the lung improves acute lung injury outcomes.


Subject(s)
Acute Lung Injury , Pneumonia , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acyl Coenzyme A , Acyltransferases , Animals , Benzimidazoles , Bleomycin , Cholesterol , Female , Male , Mice , Mice, Inbred C57BL , Sterol O-Acyltransferase/genetics
7.
Lipids Health Dis ; 21(1): 22, 2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35172832

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most common cancers worldwide characterized by disparities in age, gender, race and anatomic sites. The mechanism underlying pathogenesis, progression and disparities of CRC remains unclear. This study aims to reveal the association of expression levels of enzymes related to cholesteryl ester (CE) metabolism with pathogenesis, progression and disparities of CRC. METHODS: The differences in gene expression levels were analyzed for enzymes in CE synthesis (acyl CoA: cholesterol acyltransferase 1 and 2, ACAT1, and ACAT2), and in CE hydrolysis (neutral cholesterol ester hydrolase, NCEH1 and lysosomal acid lipase, LAL) on TNMplot platform between CRC and normal colorectal tissues (NCT) in a large cohort. The differences in protein expression levels for these enzymes were determined by Immunochemistry (IHC) performed on tissue microarray containing 96 pairs of CRC and benign colorectal tissues (BCT) from different patient populations. The expression level represented as IHC score of each enzyme was compared between CRC and BCT in entire population and populations stratified by race, gender and anatomic sites. Student's t-test, Fisher exact test and ANOVA were used for data analysis. Significant p value was set at P<0.05. RESULTS: The gene expression level of ACAT1 was significantly lower in CRC than in NCT (P = 2.15e-119). The gene expression level of ACAT2 was not statistically different between CRC and NCT. The gene expression level of LIPA (encoding LAL) was significantly higher in CRC than in NCT (P = 2.01e-14). No data was found for the gene expression level of NCEH1. The IHC score of ACAT1was significantly lower in CRC than in BCT in all studied populations and in sub site of colon, but not in that of rectum. The IHC score of ACAT2 was not statistically different between CRC and BCT. IHC score of NCEH1 was significantly higher in CRC than in BCT only in African American (AA) population. The IHC score of LAL was significantly higher in CRC than in BCT in all studied populations and in all sub sites. In addition, decreased ACAT1 in CRC significantly correlated to progression of CRC: the lower IHC score of ACAT1, the more advanced clinical stage of CRC will be. CONCLUSIONS: This study revealed that altered expression levels in enzymes related to CE metabolism highly correlate to the pathogenesis, clinical progression and disparities of CRC. The results will add revenue in elucidating mechanisms underlying progression of CRC, and shed light on seeking biomarkers and exploring therapeutic targets for CRC in a new direction.


Subject(s)
Cholesterol Esters , Colorectal Neoplasms , Cholesterol Esters/metabolism , Colorectal Neoplasms/genetics , Humans , Sterol Esterase/genetics , Sterol Esterase/metabolism , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism
8.
J Intern Med ; 292(2): 296-307, 2022 08.
Article in English | MEDLINE | ID: mdl-34982494

ABSTRACT

BACKGROUND: Sterol O-acyltransferase 2 (Soat2) encodes acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2), which synthesizes cholesteryl esters in hepatocytes and enterocytes fated either to storage or to secretion into nascent triglyceride-rich lipoproteins. OBJECTIVES: We aimed to unravel the molecular mechanisms leading to reduced hepatic steatosis when Soat2 is depleted in mice. METHODS: Soat2-/- and wild-type mice were fed a high-fat, a high-carbohydrate, or a chow diet, and parameters of lipid and glucose metabolism were assessed. RESULTS: Glucose, insulin, homeostatic model assessment for insulin resistance (HOMA-IR), oral glucose tolerance (OGTT), and insulin tolerance tests significantly improved in Soat2-/- mice, irrespective of the dietary regimes (2-way ANOVA). The significant positive correlations between area under the curve (AUC) OGTT (r = 0.66, p < 0.05), serum fasting insulin (r = 0.86, p < 0.05), HOMA-IR (r = 0.86, p < 0.05), Adipo-IR (0.87, p < 0.05), hepatic triglycerides (TGs) (r = 0.89, p < 0.05), very-low-density lipoprotein (VLDL)-TG (r = 0.87, p < 0.05) and the hepatic cholesteryl esters in wild-type mice disappeared in Soat2-/- mice. Genetic depletion of Soat2 also increased whole-body oxidation by 30% (p < 0.05) compared to wild-type mice. CONCLUSION: Our data demonstrate that ACAT2-generated cholesteryl esters negatively affect the metabolic control by retaining TG in the liver and that genetic inhibition of Soat2 improves liver steatosis via partitioning of lipids into secretory (VLDL-TG) and oxidative (fatty acids) pathways.


Subject(s)
Fatty Liver , Insulins , Sterol O-Acyltransferase , Animals , Cholesterol Esters/metabolism , Fatty Liver/metabolism , Glucose/metabolism , Insulins/metabolism , Lipoproteins, VLDL/metabolism , Liver/metabolism , Male , Mice , Mice, Knockout , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Triglycerides , Sterol O-Acyltransferase 2
9.
Ir J Med Sci ; 191(4): 1549-1554, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34460058

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignant gastrointestinal cancers. Metastasis is the major leading cause of death in patients with CRC, and many patients treated with radical surgery were diagnosed with metastasis during follow-up. However, the underlying molecular mechanisms regulating CRC metastasis are still elusive. Sterol o-acyltransferase 1 (SOAT1) is a critical participant in maintaining intracellular cholesterol balance. Here, by analyzing the clinical specimens and in vitro cell line experiments, we evaluated the clinical relevance and role of SOAT1 in regulating CRC metastasis. The results revealed that SOAT1 was overexpressed in colon cancer tissues compared to peritumor tissues at mRNA and protein levels. High intratumor SOAT1 expression correlates to lymph node metastasis and indicates poor patient disease-free survival and overall survival. The silencing of SOAT1 strongly inhibited the migration and invasion ability of CRC tumor cells. These results demonstrated that SOAT1 was upregulated in colon cancer. Upregulation of SOAT1 expression may promote CRC progression by enhancing the migration and invasion ability of CRC. Our results indicate that targeting SOAT1 activity may be applied as a promising therapeutic strategy for preventing the metastasis of CRC after radical surgical treatment.


Subject(s)
Colorectal Neoplasms , Sterol O-Acyltransferase , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/surgery , Humans , Prognosis , Sterol O-Acyltransferase/genetics
10.
Prostate Cancer Prostatic Dis ; 25(3): 484-490, 2022 09.
Article in English | MEDLINE | ID: mdl-34326474

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is the most frequent cancer in men. The prognosis of PCa is heterogeneous with many clinically indolent tumors and rare highly aggressive cases. Reliable tissue markers of prognosis are lacking. Active cholesteryl ester synthesis has been associated with prostate cancer aggressiveness. Sterol-O-Acyl transferases (SOAT) 1 and 2 catalyze cholesterol esterification in humans. OBJECTIVE: To investigate the value of SOAT1 and SOAT2 tissue expression as prognostic markers in high risk PCa. PATIENTS AND METHODS: Formalin-fixed paraffin-embedded tissue samples from 305 high risk PCa cases treated with radical prostatectomy were analyzed for SOAT1 and SOAT2 protein expression by semi-quantitative immunohistochemistry. The Kaplan-Meier method and Cox proportional hazards modeling were used to compare outcome. MAIN OUTCOME MEASURE: Biochemical recurrence (BCR) free survival. RESULTS: SOAT1 expression was high in 73 (25%) and low in 219 (75%; not evaluable: 13) tumors. SOAT2 was highly expressed in 40 (14%) and at low levels in 249 (86%) samples (not evaluable: 16). By Kaplan-Meier analysis, we found significantly shorter median BCR free survival of 93 months (95% confidence interval 23.6-123.1) in patients with high SOAT1 vs. 134 months (112.6-220.2, Log-rank p < 0.001) with low SOAT1. SOAT2 expression was not significantly associated with BCR. After adjustment for age, preoperative PSA, tumor stage, Gleason score, resection status, lymph node involvement and year of surgery, high SOAT1 but not SOAT2 expression was associated with shorter BCR free survival with a hazard ratio of 2.40 (95% CI 1.57-3.68, p < 0.001). Time to clinical recurrence and overall survival were not significantly associated with SOAT1 and SOAT2 expression CONCLUSIONS: SOAT1 expression is strongly associated with BCR free survival alone and after multivariable adjustment in high risk PCa. SOAT1 may serve as a histologic marker of prognosis and holds promise as a future treatment target.


Subject(s)
Neoplasm Recurrence, Local , Prostatic Neoplasms , Sterol O-Acyltransferase , Cholesterol/metabolism , Disease-Free Survival , Humans , Male , Neoplasm Grading , Neoplasm Recurrence, Local/genetics , Prostate-Specific Antigen , Prostatectomy , Prostatic Neoplasms/pathology , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism
11.
JCI Insight ; 7(3)2022 02 08.
Article in English | MEDLINE | ID: mdl-34914638

ABSTRACT

Colorectal cancer (CRC) severely threatens human health and life span. An effective therapeutic strategy has not been established because we do not clearly know its pathogenesis. Here, we report that ceramide and sterol O-acyltransferase 1 (SOAT1) have roles in both spontaneous and chemical-induced intestinal cancers. We first found that miRNA-148a deficiency dramatically increased mouse gut dysbiosis through upregulating ceramide synthase 5 (Cers5) expression, which promoted ceramide synthesis afterward. The newly generated ceramide further promoted both azoxymethane/dextran sodium sulfate-induced (AOM/DSS-induced) and ApcMin/+ spontaneous intestinal tumorigenesis via increasing mouse gut dysbiosis. Meanwhile, increased level of ceramide correlated with the significant enhancements of both ß-catenin activity and colorectal tumorigenesis in a TLR4-dependent fashion. Next, we found a direct binding of ß-catenin to SOAT1 promoter to activate transcriptional expression of SOAT1, which further induced cholesterol esterification and colorectal tumorigenesis. In human patients with CRC, the same CERS5/TLR4/ß-catenin/SOAT1 axis was also found to be dysregulated. Finally, the SOAT1 inhibitor (avasimibe) showed significant levels of therapeutic effects on both AOM/DSS-induced and ApcMin/+ spontaneous intestinal cancer. Our study clarified that ceramide promoted CRC development through increasing gut dysbiosis, further resulting in the increase of cholesterol esterification in a SOAT1-dependent way. Treatment with avasimibe to specifically decrease cholesterol esterification could be considered as a clinical strategy for effective CRC therapy in a future study.


Subject(s)
Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Cholesterol/metabolism , Colorectal Neoplasms/genetics , Dysbiosis/complications , Gene Expression Regulation, Neoplastic , Sterol O-Acyltransferase/genetics , Animals , Ceramides/toxicity , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Dysbiosis/chemically induced , Dysbiosis/pathology , Esterification/genetics , Humans , Mice , Mice, Inbred C57BL , Neoplasms, Experimental , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Sterol O-Acyltransferase/biosynthesis
12.
J Exp Med ; 218(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34424266

ABSTRACT

Upon demyelinating injury, microglia orchestrate a regenerative response that promotes myelin repair, thereby restoring rapid signal propagation and protecting axons from further damage. Whereas the essential phagocytic function of microglia for remyelination is well known, the underlying metabolic pathways required for myelin debris clearance are poorly understood. Here, we show that cholesterol esterification in male mouse microglia/macrophages is a necessary adaptive response to myelin debris uptake and required for the generation of lipid droplets upon demyelinating injury. When lipid droplet biogenesis is defective, innate immune cells do not resolve, and the regenerative response fails. We found that triggering receptor expressed on myeloid cells 2 (TREM2)-deficient mice are unable to adapt to excess cholesterol exposure, form fewer lipid droplets, and build up endoplasmic reticulum (ER) stress. Alleviating ER stress in TREM2-deficient mice restores lipid droplet biogenesis and resolves the innate immune response. Thus, we conclude that TREM2-dependent formation of lipid droplets constitute a protective response required for remyelination to occur.


Subject(s)
Lipid Droplets/metabolism , Membrane Glycoproteins/metabolism , Phagocytes/physiology , Receptors, Immunologic/metabolism , Remyelination/physiology , Animals , Cholesterol/metabolism , Endoplasmic Reticulum Stress , Esterification , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Protein Biosynthesis/physiology , Receptors, Immunologic/genetics , Sterol O-Acyltransferase/genetics
13.
Biomolecules ; 11(6)2021 06 08.
Article in English | MEDLINE | ID: mdl-34201030

ABSTRACT

The association between intratumoral cholesteryl ester (CE) and tumor progression has been reported previously. The objective of our study was to investigate a causal effect of CE on mammary tumor progression. Using MMTV-PyMT (MMTV-polyoma virus middle T) transgenic mice and breast tumor cell MCF-7, we show that both exogenous and endogenous CE can increase mammary tumor growth, that CE upregulates the AKT/mTOR pathway, and that CE synthesis blockade suppresses this signaling pathway. Our data suggest that SOAT1, a sterol O-acyltransferase, may be a potential target for the treatment of breast cancer.


Subject(s)
Cholesterol Esters/metabolism , Mammary Neoplasms, Animal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sterol O-Acyltransferase/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cholesterol Esters/genetics , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/genetics , Sterol O-Acyltransferase/genetics , TOR Serine-Threonine Kinases/genetics
14.
BMC Cancer ; 21(1): 615, 2021 May 26.
Article in English | MEDLINE | ID: mdl-34039309

ABSTRACT

BACKGROUND: Hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) remains a major public health problem and its pathogenesis remains unresolved. A recent proteomics study discovered a lipid enzyme Sterol O-acyltransferase (SOAT1) involvement in the progression of HCC. We aimed to explore the association between SOAT1 genetic variation and HCC. METHODS: We genotyped three exonic SOAT1 variants (rs10753191, V323V; rs3753526, L475L; rs13306731, Q526R) tagging most variations in the gene, in 221 HCC patients and 229 healthy individuals, to assess the impact of SOAT1 gene variation on risk of HCC occurrence. We further conducted immunohistochemistry to compare SOAT1 protein expression levels in 42 paired tumor and adjacent non-tumor tissues. RESULTS: We found that rs10753191 (Odds ratio (OR) = 0.58, P = 0.04) and a haplotype TGA (OR = 0.40, P = 0.01) were associated with reduced HCC risk after adjusting for lipid levels. In the immunohistochemistry experiment, we found that the protein expression of SOAT1 was significantly increased in the tumor compared with adjacent tissue (P < 0.001). CONCLUSION: This study revealed for the first time SOAT1 genetic variation that associates with host susceptibility to HCC occurrence. Our results suggest a role of SOAT1 in the HCC development, which warrants further elucidation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genetic Predisposition to Disease , Hepatitis B, Chronic/pathology , Liver Neoplasms/genetics , Sterol O-Acyltransferase/genetics , Adult , Aged , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Case-Control Studies , Female , Gene Expression Regulation, Neoplastic , Gene Frequency , Healthy Volunteers , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/virology , Humans , Kaplan-Meier Estimate , Liver/pathology , Liver/virology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Middle Aged , Odds Ratio , Polymorphism, Single Nucleotide , RNA-Seq , Sterol O-Acyltransferase/metabolism
15.
Molecules ; 27(1)2021 Dec 27.
Article in English | MEDLINE | ID: mdl-35011377

ABSTRACT

Pachymic acid from Wolfiporia cocos possesses important medicinal values including anti-bacterial, anti-inflammatory, anti-viral, invigorating, anti-rejection, anti-tumor, and antioxidant activities. However, little is known about the biosynthetic pathway from lanostane to pachymic acid. In particular, the associated genes in the biosynthetic pathway have not been characterized, which limits the high-efficiency obtaining and application of pachymic acid. To characterize the synthetic pathway and genes involved in pachymic acid synthesis, in this study, we identified 11 triterpenoids in W. cocos using liquid chromatography tandem mass spectrometry (LC-MS/MS), and inferred the putative biosynthetic pathway from lanostane to pachymic acid based on analyzing the chemical structure of triterpenoids and the transcriptome data. In addition, we identified a key gene in the biosynthetic pathway encoding W. cocos sterol O-acyltransferase (WcSOAT), which catalyzes tumolusic acid to pachymic acid. The results show that silence of WcSOAT gene in W. cocos strain led to reduction of pachymic acid production, whereas overexpression of this gene increased pachymic acid production, indicating that WcSOAT is involved in pachymic acid synthesis in W. cocos and the biosynthesis of W. cocos pachymic acid is closely dependent on the expression of WcSOAT gene. In summary, the biosynthetic pathway of pachymic acid and the associated genes complement our knowledge on the biosynthesis of W. cocos pachymic acid and other triterpenoids, and also provides a reference for target genes modification for exploring high-efficiency obtaining of active components.


Subject(s)
Fungal Proteins/metabolism , Sterol O-Acyltransferase/metabolism , Triterpenes/metabolism , Wolfiporia/metabolism , Fungal Proteins/genetics , Sterol O-Acyltransferase/genetics , Wolfiporia/enzymology , Wolfiporia/genetics
16.
Mol Neurobiol ; 58(3): 905-912, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33057949

ABSTRACT

One of the hypotheses that have emerged to explain the origin of dementia relates the disease with altered lipid metabolism, particularly cholesterol. To maintain cholesterol homeostasis, the ACAT1 enzyme has an important function to regulate the production of Aß. Moreover, APOE is the main cholesterol carrier in the brain, and it has been reported as a risk factor for this disease. This study evaluates the relationship between ACAT1 and APOE genetic variants with susceptibility for the development of Alzheimer's disease and other dementias. We examined four ACAT1 polymorphisms (rs2247071, rs2862616, rs3753526, rs1044925) and two in the APOE gene (rs7412, rs429358) in a group of 204 controls and 196 cases of dementia. Our results show one protective haplotype: CGCA (OR = 0.34, 95% CI = 0.23-0.46; p < 0.001) and one risk haplotype: CGGA (OR = 1.87, 95% CI = 1.34-2.60; p < 0.001) for the development of dementia. Subjects identified as APOE-ε4 allele carriers had a higher risk of developing dementia compared with non-carriers, OR = 13.33 (95% CI = 3.14-56.31). The results support the hypothesis that the ACAT1 gene, together with the APOE gene, plays an important role in susceptibility to the development of dementia and shows genetic characteristics of the Mexican population that can be used to identify the population at risk.


Subject(s)
Apolipoproteins E/genetics , Dementia/genetics , Genetic Predisposition to Disease , Genetic Variation , Sterol O-Acyltransferase/genetics , Aged , Aged, 80 and over , Case-Control Studies , Female , Haplotypes/genetics , Heterozygote , Humans , Linkage Disequilibrium/genetics , Male , Polymorphism, Genetic
17.
Article in English | MEDLINE | ID: mdl-33069870

ABSTRACT

Although most mammalian fatty acids (FAs) are straight-chain, there also exist branched-chain FAs such as iso- and anteiso-FAs, especially in the meibomian glands. Meibum lipids, which are secreted from the meibomian glands and are important for dry eye prevention, contain abundant branched-chain lipids, such as cholesteryl esters and wax esters with chain-lengths of ≥C21 (very-long-chain; VLC). However, the exact tissue distribution of branched-chain lipids or the enzymes involved in the production of branched-chain VLCFAs has remained poorly understood. Here, we revealed that FA elongases ELOVL1, ELOVL3, and ELOVL7, of the seven mammalian ELOVL isozymes, elongated saturated branched-chain acyl-CoAs. ELOVL3 was highly active toward iso-C17:0 and anteiso-C17:0 acyl-CoAs and elongated them up to iso-C23:0 and anteiso-C25:0 acyl-CoAs, respectively. ELOVL1 elongated both iso- and anteiso-C23:0 acyl-CoAs to C25:0 acyl-CoAs. By establishing a liquid chromatography-tandem mass spectrometry method capable of separating branched- and straight-chain lipids, we showed that essentially all of the cholesteryl esters and 88% of the wax esters in the mouse meibomian glands are branched. In Elovl1 mutant mice, the levels of ≥C24:0 branched-chain cholesteryl esters and ≥C25:0 branched-chain wax esters were decreased, indicating that ELOVL1 indeed elongates branched-chain VLC acyl-CoAs in vivo. In addition, substantial amounts of ceramides containing branched-chain FAs were present in the skin, meibomian glands, and liver. Our findings provide new insights into the molecular mechanisms that create FA and lipid diversity.


Subject(s)
Acyl Coenzyme A/metabolism , Amino Acids, Branched-Chain/metabolism , Fatty Acid Elongases/metabolism , Meibomian Glands/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Amino Acids, Branched-Chain/classification , Animals , Ceramides/classification , Ceramides/metabolism , Cholesterol Esters/classification , Cholesterol Esters/metabolism , Fatty Acid Elongases/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Lipidomics/methods , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Skin/metabolism , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Substrate Specificity
18.
Article in English | MEDLINE | ID: mdl-33031913

ABSTRACT

We previously demonstrated that AKR vs. DBA/2 mouse bone marrow derived macrophages have higher levels of free cholesterol and lower levels of esterified cholesterol after cholesterol loading, and that AKR, but not DBA/2, macrophages induced C/EBP homologous protein (CHOP) expression after cholesterol loading. We earlier determined that the free and esterified cholesterol level effect is due to a truncation in the sterol O-acyltransferase 1 (Soat1) gene, encoding acetyl-coenzyme A acetyltransferase 1 (ACAT1). Here we examined the mechanism for the differential induction of CHOP by cholesterol loading. CHOP was induced in both strains after incubation with tunicamycin, indicating both strains have competent endoplasmic reticulum stress pathways. CHOP was induced when DBA/2 macrophages were cholesterol loaded in the presence of an ACAT inhibitor, indicating that the difference in free cholesterol levels were responsible for this strain effect. This finding was confirmed in macrophages derived from DBA/2 embryonic stem cells. Cholesterol loading of Soat1 gene edited cells, mimicking the AKR allele, led to increased free cholesterol levels and restored CHOP induction. The upstream pathway of free cholesterol induced endoplasmic reticulum stress was investigated; and, RNA-dependent protein kinase-like endoplasmic reticulum kinase (PERK) and inositol-requiring enzyme 1 α protein kinase (IRE1α) pathways were required for maximal CHOP expression.


Subject(s)
Cholesterol/pharmacology , Endoplasmic Reticulum Stress/genetics , Macrophages/metabolism , Sterol O-Acyltransferase/genetics , Transcription Factor CHOP/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cholesterol/metabolism , Cholesterol Esters/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Endoribonucleases/metabolism , Female , Femur/cytology , Femur/metabolism , Gene Expression Regulation , Macrophages/cytology , Macrophages/drug effects , Male , Mice, Inbred AKR , Mice, Inbred DBA , Mice, Knockout, ApoE , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Species Specificity , Sterol O-Acyltransferase/metabolism , Transcription Factor CHOP/metabolism , Tunicamycin/pharmacology , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
19.
J Mol Endocrinol ; 65(4): 149-161, 2020 11.
Article in English | MEDLINE | ID: mdl-33064661

ABSTRACT

This study aimed to identify circular RNAs differentially expressed in the islets of type 2 diabetes (T2DM) models and clarify their roles in the control of ß-cell functions. Circular RNAs dysregulated in the islets of diabetic db/db mice were identified by high-throughput RNA sequencing. Then, the expression level of the selected circular RNA circ-Tulp4 was confirmed by real-time PCR in the islets of diabetic models and Min6 cells. MTS, EdU, western blot, flow cytometric analysis, and luciferase assay were performed to investigate the impact of circ-Tulp4 on ß-cell functions. This study identified thousands of circular RNAs in mouse pancreatic islets. The circ-Tulp4 level significantly decreased in the diabetic models and altered in the Min6 cells under lipotoxic condition. The modulation of circ-Tulp4 level in Min6 cells regulated cell proliferation. Furthermore, an interaction was demonstrated between circ-Tulp4 and miR-7222-3p, which suppressed the expression of cholesterol esterification-related gene, sterol O-acyltransferase 1 (SOAT1). The accumulation of soat1 activated cyclin D1 expression, thus promoting cell cycle progression. These findings showed that circ-Tulp4 regulated ß-cell proliferation via miR-7222-3p/soat1/cyclin D1 signaling. Our research suggested that circ-Tulp4 might be a potential therapeutic intervention for T2DM. Besides, soat1 might be important for ß-cell adaptation to lipotoxicity.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Intracellular Signaling Peptides and Proteins/genetics , RNA, Circular , Sterol O-Acyltransferase/genetics , Animals , Cell Line , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling , Male , Mice , Mice, Transgenic , RNA, Messenger , Transcriptome
20.
Steroids ; 164: 108725, 2020 12.
Article in English | MEDLINE | ID: mdl-32890578

ABSTRACT

Lipids present in lipoproteins cleared from the circulation are processed sequentially by three major proteins within the late endosomal/lysosomal (E/L) compartment of all cells: lysosomal acid lipase (LAL), Niemann-Pick (NPC) C2 and NPC1. When all three of these proteins are functioning normally, unesterified cholesterol (UC) exits the E/L compartment and is used in plasma membrane maintenance and various pathways in the endoplasmic reticulum including esterification by sterol O-acyltransferase 2 (SOAT2) or SOAT1 depending partly on cell type. Mutations in either NPC2 or NPC1 result in continual entrapment of UC and glycosphingolipids leading to neurodegeneration, pulmonary dysfunction, splenomegaly and liver damage. To date, the most effective agent for promoting release of entrapped UC in nearly all organs of NPC1-deficient mice and cats is 2-hydroxypropyl-ß-cyclodextrin (2HPßCD). The cytotoxic nature of the liberated UC triggers various defenses including suppression of sterol synthesis and increased esterification. The present studies, using the Npc1-/-nih mouse model, measured the comparative quantitative importance of these two responses in the liver versus the spleen of Npc1-/-: Soat2+/+ and Npc1-/-: Soat2-/- mice in the 24 h following a single acute treatment with 2HPßCD. In the liver but not the spleen of both types of mice suppression of synthesis alone or in combination with increased esterification provided the major defense against the rise in unsequestered cellular UC content. These findings have implications for systemic 2HPßCD treatment in NPC1 patients in view of the purportedly low levels of SOAT2 activity in human liver.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Cholesterol/metabolism , Niemann-Pick C1 Protein/genetics , Sterol O-Acyltransferase/metabolism , 2-Hydroxypropyl-beta-cyclodextrin/administration & dosage , Animals , Liver/enzymology , Mice , Mice, Inbred BALB C , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase 2
SELECTION OF CITATIONS
SEARCH DETAIL
...