Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 223
Filter
1.
Life Sci ; 240: 117110, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31786191

ABSTRACT

AIMS: Thymic carcinoma is a rare epithelial tumor, for which, optimal pharmacotherapeutic methods have not yet been established. To develop new drug treatments for thymic carcinoma, we investigated the effects of fluvastatin-mediated pharmacological inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) on thymic carcinoma. MAIN METHODS: Thymic carcinoma tissue was surgically excised and HMGCR expression was assessed by immunohistochemistry. Ty82 human thymic carcinoma cells were treated with fluvastatin (1-10 µM) and their growth was monitored. KEY FINDINGS: HMGCR was expressed on carcinoma cells but not on normal epithelial cells in thymic tissue. Inhibition of HMGCR by fluvastatin suppressed cell proliferation and induced the death of Ty-82 human thymic carcinoma cells. Fluvastatin mediated its antitumor effects by blocking the production of geranylgeranyl-pyrophosphate (GGPP), an isoprenoid that is produced from mevalonate and binds to small GTPases, which promotes cell proliferation. SIGNIFICANCE: Fluvastatin showed marked antitumor effects on thymic carcinoma. The results suggest that the statin has clinical benefits in thymic carcinoma management.


Subject(s)
Fluvastatin/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Thymoma/drug therapy , Thymus Neoplasms/drug therapy , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Humans , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Immunohistochemistry , MAP Kinase Signaling System/drug effects , Polyisoprenyl Phosphates/antagonists & inhibitors , Polyisoprenyl Phosphates/biosynthesis , Prenylation/drug effects
2.
Mol Med Rep ; 20(4): 3003-3010, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432128

ABSTRACT

Dysregulations of the mevalonate pathway (MVA) have been previously identified. Our previous study demonstrated that 3­hydroxy­3­methylglutaryl­coenzyme A reductase (HMGCR), the rate­limiting enzyme of the MVA pathway, was upregulated in esophageal squamous cell carcinoma (ESCC) and statin­inhibited ESCC tumorigenesis. However, the underlying mechanism of HMGCR regulation in ESCC remains unknown. In the present study, western blotting and immunohistochemistry analysis demonstrated that sterol regulatory element­binding protein 2 (SREBP2), the master regulator for HMGCR, was upregulated in ESCC clinical samples. Overexpression of SREBP2 expression in ESCC cell lines promoted the growth, migration and colony formation of cancer cells in the MTT, Boyden chamber and soft agar assays, respectively, which was inhibited by lovastatin. Downregulation of SREBP2 expression in ESCC cell lines inhibited the viability, and migration and colony formation abilities of cancer cells. Assessment of the molecular mechanism demonstrated that SREBP2 interacted with c­Myc and cooperated with c­Myc to activate HMGCR expression. Collectively, the present study identified SREBP2 as an oncogene associated with the tumorigenesis of ESCC and further demonstrated the therapeutic effects of statins in ESCC.


Subject(s)
Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , Sterol Regulatory Element Binding Protein 2/biosynthesis , Up-Regulation , Cell Line, Tumor , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Proto-Oncogene Proteins c-myc/genetics , Sterol Regulatory Element Binding Protein 2/genetics
3.
Medicine (Baltimore) ; 98(13): e14968, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30921201

ABSTRACT

There are many preclinical and epidemiological reports suggesting a correlation between 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoAR) or HMG-CoAR inhibitor (statin) treatment and prognosis in breast cancer. This study aimed to investigate the expression of HMG-CoAR in Korean patients with breast cancer.The expression of HMG-CoAR on tissue microarrays from 191 patients who underwent resection from 2005 to 2006 in the Pusan National University Hospital was assessed by immunohistochemistry (IHC). The IHC assessment by a board-certified pathologist included areas of both carcinoma and peritumoral tissue of the breast. The scores of cancer-specific staining were adjusted by the scores of peritumoral staining.The patients were followed for a median 9.1 years. Disease-free survival (DFS) was shorter in patients with a positive adjusted HMG-CoAR score by log-rank test (not reached vs 11.6 years, P = .011). After adjusting for age, T stage, N stage, pathological grade, perioperational chemotherapy, adjuvant radiotherapy, estrogen receptor positivity, progesterone receptor positivity, human epidermal growth factor receptor-2 positivity, and high Ki-67 (>10%), a positive adjusted HMG-CoAR IHC score was also associated with shorter DFS (hazard ratio = 2.638, 95% confidence interval [CI] 1.112-6.262, P = .028).The expression of HMG-CoAR might be an independent prognostic factor in breast cancer. There are established drugs targeting HMG-CoAR, and further studies on its potential as a predictive marker are needed.


Subject(s)
Breast Neoplasms/physiopathology , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Proportional Hazards Models , Retrospective Studies , Socioeconomic Factors , Tissue Array Analysis
4.
Cell Death Dis ; 10(2): 91, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692522

ABSTRACT

The mevalonate pathway has emerged as a promising target for several solid tumors. Statins are inhibitors of the 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), the rate-limiting enzyme of this pathway, and are commonly used to treat patients with hypercholesterolemia. Pleiotropic antitumor mechanisms of statins have been demonstrated for several human cancer types. However, cancer cells differ in their individual statin sensitivity and some cell lines have shown relative resistance. In this study we demonstrate, that the human breast cancer cell lines MDA-MB-231, MDA-MB-468, MCF-7, and T47D are differentially affected by statins. Whereas the vitality of MDA-MB-231 and MDA-MB-468 cells was reduced by up to 60% using atorvastatin, simvastatin, or rosuvastatin (p < 0.001), only marginal effects were seen in T47D and MCF-7 cells following exposure to statins. Statin treatment led to an upregulation of HMGCR mRNA and protein expression by up to sixfolds in the statin-resistant cells lines (p < 0.001), but no alterations of HMGCR were observed in the statin-sensitive MDA-MB-231 and MDA-MB-468 cells. The knockdown of HMGCR prior to statin treatment sensitized the resistant cell lines, reflected by a 70% reduction in vitality, increased apoptotic DNA fragmentation (sixfold) and by accumulation of the apoptosis marker cleaved poly-ADP ribose polymerase. Statins induced a cleavage of the sterol-regulatory element-binding protein (SREBP)-2, a transcriptional activator of the HMGCR, in T47D and MCF-7 cells. The inhibition of SREBP-2 activation by co-administration of dipyridamole sensitized MCF-7 and T47D cells for statins (loss of vitality by 80%; p < 0.001). Furthermore, assessment of a statin-resistant MDA-MB-231 clone, generated by long-term sublethal statin exposure, revealed a significant induction of HMGCR expression by up to 12-folds (p < 0.001). Knockdown of HMGCR restored statin sensitivity back to levels of the parental cells. In conclusion, these results indicate a resistance of cancer cells against statins, which is in part due to the induction of HMGCR.


Subject(s)
Breast Neoplasms/drug therapy , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Enzyme Induction , Female , Gene Expression , Humans , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , MCF-7 Cells
5.
Lipids Health Dis ; 15(1): 161, 2016 Sep 19.
Article in English | MEDLINE | ID: mdl-27644038

ABSTRACT

BACKGROUND: Previous reports have suggested that advanced glycation end products (AGEs) participate in the pathogenesis of diabetic macroangiopathy. Our previous study have found that AGEs can increase the lipid droplets accumulation in aortas of diabetic rats, but the current understanding of the mechanisms remains incomplete by which AGEs affect lipids accumulation in macrophages and accelerate atherosclerosis. In this study, we investigated the role of AGEs on lipids accumulation in macrophages and the possible molecular mechanisms including cholesterol influx, esterification and efflux of macrophages. METHODS: THP-1 cells were incubated with PMA to differentiate to be macrophages which were treated with AGEs in the concentration of 300 µg/ml and 600 µg/ml with or without anti-RAGE (receptor for AGEs) antibody and then stimulated by oxidized-LDL (oxLDL) or Dil-oxLDL. Lipids accumulation was examined by oil red staining. The cholesterol uptake, esterification and efflux were detected respectively by fluorescence microscope, enzymatic assay kit and fluorescence microplate. Quantitative RT-PCR and Western blot were used to measure expression of the moleculars involved in cholesterol uptake, synthesis/esterification and efflux. RESULTS: AGEs increased lipids accumulation in macrophages in a concentration-dependent manner. 600 µg/ml AGEs obviously upregulated oxLDL uptake, increased levels of cholesterol ester in macrophages, and decreased the HDL-mediated cholesterol efflux by regulating the main molecular expression including CD36, Scavenger receptors (SR) A2, HMG-CoA reductase (HMGCR), ACAT1 and ATP-binding cassette transporter G1 (ABCG1). The changes above were inversed when the cells were pretreated with anti-RAGE antibody. CONCLUSIONS: The current study suggest that AGEs can increase lipids accumulation in macrophages by regulating cholesterol uptake, esterification and efflux mainly through binding with RAGE, which provide a deep understanding of mechanisms how AGEs accelerating diabetic atherogenesis.


Subject(s)
Cholesterol/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/metabolism , Lipid Metabolism/genetics , Receptor for Advanced Glycation End Products/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/biosynthesis , Acetyl-CoA C-Acetyltransferase/biosynthesis , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Angiopathies/genetics , Diabetic Angiopathies/pathology , Esterification , Gene Expression Regulation , Glycation End Products, Advanced/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Macrophages/pathology , Rats , Receptor for Advanced Glycation End Products/metabolism
6.
Exp Biol Med (Maywood) ; 241(6): 667-74, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26825354

ABSTRACT

Hawthorn is a berry-like fruit from the species of Crataegus. In China, it has another more famous name, Shan-Zha, which has been used to improve digestion as a traditional Chinese medicine or food for thousands of years. Moreover, during the last decades, hawthorn has received more attention because of its potential to treat cardiovascular diseases. However, currently, only fruits of C. pinnatifida and C. pinnatifida var. major are included as Shan-Zha in the Chinese Pharmacopoeia. In this study, our results showed that the ethanol extract of Zhongtian hawthorn, a novel grafted cultivar of C. cuneata (wild Shan-Zha), could markedly reduce body weight and levels of serum total cholesterol, triglyceride, low-density lipoprotein cholesterol, and liver cholesterol of hyperlipidemia mice. It could suppress the stimulation effect of high-fat diet on the transcription of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) and p65, and counteract the downregulation of CYP7A1 and LDLR. In addition, the results of luciferase reporter assay and Western blot showed that the transcriptional activity of HMGCR promoter was inhibited by Zhongtian hawthorn ethanol extract in a dose-dependent manner, while overexpression of p65 could reverse this transcriptional repression effect. These results suggested that Zhongtian hawthorn could provide health benefits by counteracting the high-fat diet-induced hypercholesteolemic and hyperlipidemic effects in vivo, and the mechanism underlying this event was mainly dependent on the suppressive effect of Zhongtian hawthorn ethanol extract on the transcription of HMGCR via nuclear factor-kappa B (NF-κB) signal pathway. Therefore, this novel cultivar of hawthorn cultivar which has much bigger fruits, early bearing, high yield, cold resistance, and drought resistance, might be considered as a good alternative to Shan-Zha and has great value in the food and medicine industry. In addition, to our best knowledge, this is also the first report that the extract of Crataegus could suppress the transcription of HMGCR via NF-κB signal pathway.


Subject(s)
Anticholesteremic Agents/administration & dosage , Cholesterol/blood , Crataegus/chemistry , Hydroxymethylglutaryl CoA Reductases/biosynthesis , NF-kappa B/antagonists & inhibitors , Plant Extracts/administration & dosage , Transcription, Genetic/drug effects , Animals , Anticholesteremic Agents/isolation & purification , Diet, High-Fat/adverse effects , Disease Models, Animal , Hypercholesterolemia/drug therapy , Male , Mice , Plant Extracts/isolation & purification , Serum/chemistry , Signal Transduction/drug effects
7.
BMC Dev Biol ; 15: 47, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26627605

ABSTRACT

BACKGROUND: Biliary atresia (BA) is a human infant disease with inflammatory fibrous obstructions in the bile ducts and is the most common cause for pediatric liver transplantation. In contrast, the sea lamprey undergoes developmental BA with transient cholestasis and fibrosis during metamorphosis, but emerges as a fecund adult. Therefore, sea lamprey liver metamorphosis may serve as an etiological model for human BA and provide pivotal information for hepatobiliary transformation and possible therapeutics. RESULTS: We hypothesized that liver metamorphosis in sea lamprey is due to transcriptional reprogramming that dictates cellular remodeling during metamorphosis. We determined global gene expressions in liver at several metamorphic landmark stages by integrating mRNA-Seq and gene ontology analyses, and validated the results with real-time quantitative PCR, histological and immunohistochemical staining. These analyses revealed that gene expressions of protein folding chaperones, membrane transporters and extracellular matrices were altered and shifted during liver metamorphosis. HSP90, important in protein folding and invertebrate metamorphosis, was identified as a candidate key factor during liver metamorphosis in sea lamprey. Blocking HSP90 with geldanamycin facilitated liver metamorphosis and decreased the gene expressions of the rate limiting enzyme for cholesterol biosynthesis, HMGCoA reductase (hmgcr), and bile acid biosynthesis, cyp7a1. Injection of hsp90 siRNA for 4 days altered gene expressions of met, hmgcr, cyp27a1, and slc10a1. Bile acid concentrations were increased while bile duct and gall bladder degeneration was facilitated and synchronized after hsp90 siRNA injection. CONCLUSIONS: HSP90 appears to play crucial roles in hepatobiliary transformation during sea lamprey metamorphosis. Sea lamprey is a useful animal model to study postembryonic development and mechanisms for hsp90-induced hepatobiliary transformation.


Subject(s)
Bile Ducts, Intrahepatic/embryology , Biliary Atresia/embryology , Cholestasis/embryology , HSP90 Heat-Shock Proteins/genetics , Metamorphosis, Biological/physiology , Petromyzon/embryology , Animals , Benzoquinones/pharmacology , Bile Acids and Salts/metabolism , Bile Ducts, Intrahepatic/pathology , Biliary Atresia/pathology , Cholesterol 7-alpha-Hydroxylase/biosynthesis , Cholesterol 7-alpha-Hydroxylase/genetics , Enzyme Inhibitors/pharmacology , Extracellular Matrix/metabolism , Fibrosis/embryology , Gallbladder/embryology , Gallbladder/pathology , Gene Expression Regulation, Developmental/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Lactams, Macrocyclic/pharmacology , Liver/embryology , Organic Anion Transporters, Sodium-Dependent/biosynthesis , Proto-Oncogene Proteins c-met/biosynthesis , RNA Interference , RNA, Small Interfering/genetics , Symporters/biosynthesis
8.
J Biol Chem ; 290(48): 28822-33, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26463208

ABSTRACT

Plasmalogen biosynthesis is regulated by modulating fatty acyl-CoA reductase 1 stability in a manner dependent on cellular plasmalogen level. However, physiological significance of the regulation of plasmalogen biosynthesis remains unknown. Here we show that elevation of the cellular plasmalogen level reduces cholesterol biosynthesis without affecting the isoprenylation of proteins such as Rab and Pex19p. Analysis of intermediate metabolites in cholesterol biosynthesis suggests that the first oxidative step in cholesterol biosynthesis catalyzed by squalene monooxygenase (SQLE), an important regulator downstream HMG-CoA reductase in cholesterol synthesis, is reduced by degradation of SQLE upon elevation of cellular plasmalogen level. By contrast, the defect of plasmalogen synthesis causes elevation of SQLE expression, resulting in the reduction of 2,3-epoxysqualene required for cholesterol synthesis, hence implying a novel physiological consequence of the regulation of plasmalogen biosynthesis.


Subject(s)
Cholesterol/biosynthesis , Homeostasis/physiology , Plasmalogens/biosynthesis , Animals , CHO Cells , Cholesterol/genetics , Cricetinae , Cricetulus , Gene Expression Regulation, Enzymologic/physiology , HEK293 Cells , HeLa Cells , Humans , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmalogens/genetics , Protein Prenylation/physiology , Squalene Monooxygenase/biosynthesis , Squalene Monooxygenase/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
9.
PLoS One ; 10(8): e0135637, 2015.
Article in English | MEDLINE | ID: mdl-26302339

ABSTRACT

High blood cholesterol has been associated with cardiovascular diseases. The enzyme HMG CoA reductase (HMGCR) is responsible for cholesterol synthesis, and inhibitors of this enzyme (statins) have been used clinically to control blood cholesterol. Sterol regulatory element binding protein (SREBP) -2 is a key transcription factor in cholesterol metabolism, and HMGCR is a target gene of SREBP-2. Attenuating SREBP-2 activity could potentially minimize the expression of HMGCR. Luteolin is a flavone that is commonly detected in plant foods. In the present study, Luteolin suppressed the expression of SREBP-2 at concentrations as low as 1 µM in the hepatic cell lines WRL and HepG2. This flavone also prevented the nuclear translocation of SREBP-2. Post-translational processing of SREBP-2 protein was required for nuclear translocation. Luteolin partially blocked this activation route through increased AMP kinase (AMPK) activation. At the transcriptional level, the mRNA and protein expression of SREBP-2 were reduced through luteolin. A reporter gene assay also verified that the transcription of SREBF2 was weakened in response to this flavone. The reduced expression and protein processing of SREBP-2 resulted in decreased nuclear translocation. Thus, the transcription of HMGCR was also decreased after luteolin treatment. In summary, the results of the present study showed that luteolin modulates HMGCR transcription by decreasing the expression and nuclear translocation of SREBP-2.


Subject(s)
Cardiovascular Diseases/drug therapy , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Luteolin/administration & dosage , Sterol Regulatory Element Binding Protein 2/biosynthesis , Adenylate Kinase/genetics , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Lipid Metabolism/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Messenger/biosynthesis , Sterol Regulatory Element Binding Protein 2/genetics
10.
Biochem J ; 471(3): 369-79, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26310456

ABSTRACT

Pcyt2 (CTP:phosphoethanolamine cytidylyltransferase) is the rate-limiting enzyme in mammalian PE (phosphatidylethanolamine) biosynthesis. Previously, we reported that Pcyt2 mRNA levels increased in several types of cells after serum starvation, an effect that could be suppressed by supplementation with low-density lipoprotein or 25-HC (25-hydroxycholesterol). Transcription of Hmgcr, which encodes 3-hydroxy-3-methylglutaryl-CoA reductase, is also suppressed by 25-HC in the same dose-dependent manner. Nevertheless, a sterol-regulatory element was not detected in the Pcyt2 promoter region. The important element for transcriptional control of Pcyt2 by 25-HC (1.25 µM) was determined to reside between -56 and -36 on the basis of analysis with several Pcyt2 promoter deletion-luciferase reporters in NIH 3T3 cells. Using the yeast one-hybrid system, we found that NF-Y (nuclear factor-Y) binds at C(-37)CAAT(-41) and YY1 (Yin Yang1) binds at C(-42)AT(-40) in the Pcyt2 promoter. Endogenous NF-Y and YY1 bind clearly and competitively to these sites and are important for basal Pcyt2 transcription. Moreover, NF-Y binds to the Hmgcr promoter at C(-14)CA(-12) in gel-shift analysis, and suppression of the basal luciferase activity of the Hmgcr promoter-reporter construct (-30/+61) by 25-HC was abolished when C(-14)CA(-12) was mutated. Furthermore, transcriptional suppression of Pcyt2 by 25-HC was reduced following knockdown targeting of NF-YA or YY1. ChIP analysis revealed that 25-HC inhibited the interaction between NF-Y and RNA polymerase II on the Pcyt2 and Hmgcr promoters. On the basis of these results, we conclude that NF-Y and YY1 are important for the basal transcription of Pcyt2 and that NF-Y is involved in the inhibitory effects of 25-HC on Pcyt2 transcription.


Subject(s)
CCAAT-Binding Factor/metabolism , RNA Nucleotidyltransferases/genetics , Transcription, Genetic/drug effects , YY1 Transcription Factor/metabolism , Animals , CCAAT-Binding Factor/genetics , DNA-Binding Proteins/genetics , Humans , Hydroxycholesterols/administration & dosage , Hydroxycholesterols/metabolism , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Mice , NIH 3T3 Cells , Promoter Regions, Genetic , YY1 Transcription Factor/genetics
11.
Cell Signal ; 27(11): 2182-90, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26208883

ABSTRACT

Glyoxalase 1 (GLO1) and HMG-CoA reductase (HMGCR) are highly expressed in most tumor cells and little in normal cells. In this study, treatment of HL-60 cells with lovastatin induced characteristic apoptosis in a dose-dependent manner. We demonstrated that lovastatin treatment inhibited Ras and Raf protein translocation to cell membrane and eliminated the phosphorylation of the downstream effectors Akt and ERK, and the subsequent NF-κB translocation into nucleus. Specific inhibitors and γ-tocotrienol confirmed the Ras/Raf/ERK/NF-κB/GLO1 and Ras/Akt/NF-κB/GLO1 pathways. Data revealed that lovastatin induced HL-60 cell death was attenuated by mevalonate treatment. We demonstrated also that γ-tocotrienol showed its apoptotic effect on the HL-60 cell through the same pathway. γ-Tocotrienol enhanced the apoptotic effect of lovastatin through the down-regulation of GLO1 and HMGCR resulting in an increase of methylglyoxal and a decrease of cholesterol and led to the apoptosis of HL-60 cells. Data also revealed that both lovastatin and gamma-tocotrienol induced significant HL-60 cell differentiation. These results suggest that both lovastatin and gamma-tocotrienol could induce differentiation and followed by apoptosis.


Subject(s)
Apoptosis/drug effects , Chromans/pharmacology , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lactoylglutathione Lyase/biosynthesis , Lovastatin/pharmacology , Vitamin E/analogs & derivatives , Active Transport, Cell Nucleus/physiology , Cell Differentiation , Cell Line, Tumor , Cholesterol/metabolism , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , HL-60 Cells , Humans , Mevalonic Acid/pharmacology , NF-kappa B/metabolism , Phosphorylation , Protein Transport/physiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Pyruvaldehyde/metabolism , Signal Transduction/physiology , Vitamin E/pharmacology , raf Kinases/metabolism
12.
Int J Clin Exp Pathol ; 8(3): 2876-87, 2015.
Article in English | MEDLINE | ID: mdl-26045796

ABSTRACT

Malignant fibrous histiocytoma (MFH) of the breast and visceral organs is extremely rare. There is an incomplete understanding of the clinical pathology of the primary MFH originating from the breast and visceral organs, especially in comparison with other soft tissue sarcomas. As a consequence we searched and analyzed the clinical and pathological records of all the nine patients with diagnosed breast and visceral MFH in our hospital. Immunohistochemical staining was performed for ezrin and HMG-CoA reductase in these MFH cases and relevant mesenchymal sarcomas. The 9 MFH cases presented with nonspecific symptoms and imaging manifestations. 6 cases were classified as storiform-pleomorphic MFH, 2 cases as inflammatory MFH, and the remaining 1 case as giant cell MFH. The results showed that ezrin expression, as well as HMG-CoA reductase expression, was significantly stronger in MFH cases than other non-MFH sarcomas. Poor prognosis seemed to be associated with younger age. Certain characteristics and clinicopathologic features can help us making the diagnosis of MFH. In conclusion, our study provided the potential value of ezrin and HMG-CoA reductase for diagnosis and differential diagnosis of MFH located in the breast and visceral organs. More accurate prognostic information of this rare disease needed to be further investigated.


Subject(s)
Biomarkers, Tumor/analysis , Cytoskeletal Proteins/biosynthesis , Histiocytoma, Malignant Fibrous/diagnosis , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Adult , Aged , Cytoskeletal Proteins/analysis , Female , Humans , Hydroxymethylglutaryl CoA Reductases/analysis , Immunohistochemistry , Male , Middle Aged , Viscera/pathology
13.
PLoS Genet ; 11(3): e1005038, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25774983

ABSTRACT

Corpus allatum (CA) ablation results in juvenile hormone (JH) deficiency and pupal lethality in Drosophila. The fly CA produces and releases three sesquiterpenoid hormones: JH III bisepoxide (JHB3), JH III, and methyl farnesoate (MF). In the whole body extracts, MF is the most abundant sesquiterpenoid, followed by JHB3 and JH III. Knockout of JH acid methyl transferase (jhamt) did not result in lethality; it decreased biosynthesis of JHB3, but MF biosynthesis was not affected. RNAi-mediated reduction of 3-hydroxy-3-methylglutaryl CoA reductase (hmgcr) expression in the CA decreased biosynthesis and titers of the three sesquiterpenoids, resulting in partial lethality. Reducing hmgcr expression in the CA of the jhamt mutant further decreased MF titer to a very low level, and caused complete lethality. JH III, JHB3, and MF function through Met and Gce, the two JH receptors, and induce expression of Kr-h1, a JH primary-response gene. As well, a portion of MF is converted to JHB3 in the hemolymph or peripheral tissues. Topical application of JHB3, JH III, or MF precluded lethality in JH-deficient animals, but not in the Met gce double mutant. Taken together, these experiments show that MF is produced by the larval CA and released into the hemolymph, from where it exerts its anti-metamorphic effects indirectly after conversion to JHB3, as well as acting as a hormone itself through the two JH receptors, Met and Gce.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Drosophila Proteins/genetics , Fatty Acids, Unsaturated/genetics , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Metamorphosis, Biological/genetics , Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Corpora Allata/growth & development , Corpora Allata/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Fatty Acids, Monounsaturated/metabolism , Fatty Acids, Unsaturated/biosynthesis , Fatty Acids, Unsaturated/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Larva , Methyltransferases/biosynthesis , Methyltransferases/genetics , Pupa , Transcription Factors/metabolism
14.
Inflammation ; 38(3): 959-71, 2015.
Article in English | MEDLINE | ID: mdl-25387652

ABSTRACT

Inflammation and lipids play significant roles in the progression of chronic kidney disease. This study was designed to investigate whether inflammation disrupts cellular cholesterol homeostasis and causes the lipid nephrotoxicity in vitro and in vivo, and explored its underlying mechanisms. Inflammatory stress was induced by cytokines (interleukin-1ß (IL-1ß); tumor necrosis factor α (TNF-α)) to human mesangial cells (HMCs) in vitro and by subcutaneous casein injection in C57BL/6J mice in vivo. The data showed that inflammatory stress exacerbated renal cholesterol ester accumulation in vitro and in vivo. Inflammation increased cellular cholesterol uptake and synthesis via upregulating the expression of low-density lipoprotein receptor (LDLr) and 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCoA-R), while it decreased cholesterol efflux via downregulating the expression of liver X receptor alpha and ATP-binding cassette transporter A1. The increased lipid accumulation by inflammatory stress induced reactive oxygen species (ROS) and increased levels of endoplasmic reticulum (ER) stress markers (inositol-requiring protein 1 and activating transcription factor 6) in HMCs and kidneys of C57BL/6J mice. This study implied that inflammation promoted renal lipid accumulation and foam cell formation by disrupting cellular cholesterol homeostasis. Increased intracellular lipids under inflammatory stress caused oxidative stress and ER stress in vitro and in vivo which may contribute to renal injury and progression of chronic kidney disease.


Subject(s)
Cholesterol/metabolism , Inflammation/pathology , Kidney/pathology , Lipid Metabolism/physiology , Renal Insufficiency, Chronic/pathology , ATP Binding Cassette Transporter 1/biosynthesis , Activating Transcription Factor 6/metabolism , Animals , Biological Transport , Caseins/pharmacology , Cell Line , Creatinine/blood , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Foam Cells/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Interleukin-1beta/pharmacology , Liver X Receptors , Male , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Receptors, LDL/biosynthesis , Serum Amyloid A Protein/metabolism , Tumor Necrosis Factor-alpha/pharmacology
15.
Arterioscler Thromb Vasc Biol ; 34(9): 1917-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25035345

ABSTRACT

OBJECTIVE: Interindividual variation in pathways affecting cellular cholesterol metabolism can influence levels of plasma cholesterol, a well-established risk factor for cardiovascular disease. Inherent variation among immortalized lymphoblastoid cell lines from different donors can be leveraged to discover novel genes that modulate cellular cholesterol metabolism. The objective of this study was to identify novel genes that regulate cholesterol metabolism by testing for evidence of correlated gene expression with cellular levels of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) mRNA, a marker for cellular cholesterol homeostasis, in a large panel of lymphoblastoid cell lines. APPROACH AND RESULTS: Expression array profiling was performed on 480 lymphoblastoid cell lines established from participants of the Cholesterol and Pharmacogenetics (CAP) statin clinical trial, and transcripts were tested for evidence of correlated expression with HMGCR as a marker of intracellular cholesterol homeostasis. Of these, transmembrane protein 55b (TMEM55B) showed the strongest correlation (r=0.29; P=4.0E-08) of all genes not previously implicated in cholesterol metabolism and was found to be sterol regulated. TMEM55B knockdown in human hepatoma cell lines promoted the decay rate of the low-density lipoprotein receptor, reduced cell surface low-density lipoprotein receptor protein, impaired low-density lipoprotein uptake, and reduced intracellular cholesterol. CONCLUSIONS: Here, we report identification of TMEM55B as a novel regulator of cellular cholesterol metabolism through the combination of gene expression profiling and functional studies. The findings highlight the value of an integrated genomic approach for identifying genes that influence cholesterol homeostasis.


Subject(s)
Cholesterol/metabolism , Lymphocytes/metabolism , Receptors, LDL/metabolism , Biological Transport , Cell Membrane/metabolism , Gene Expression Profiling , Hep G2 Cells , Hepatocytes/metabolism , Homeostasis , Humans , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Intracellular Fluid/metabolism , Lipid Metabolism/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism
16.
Funct Integr Genomics ; 14(3): 603-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24913677

ABSTRACT

Tanshinone is widely used for treatment of cardio-cerebrovascular diseases with increasing demand. Herein, key enzyme genes SmHMGR (3-hydroxy-3-methylglutaryl CoA reductase) and SmDXR (1-deoxy-D-xylulose 5-phosphate reductoisomerase) involved in the tanshinone biosynthetic pathway were introduced into Salvia miltiorrhiza (Sm) hairy roots to enhance tanshinone production. Over-expression of SmHMGR or SmDXR in hairy root lines can significantly enhance the yield of tanshinone. Transgenic hairy root lines co-expressing HMGR and DXR (HD lines) produced evidently higher levels of total tanshinone (TT) compared with the control and single gene transformed lines. The highest tanshinone production was observed in HD42 with the concentration of 3.25 mg g(-1) DW. Furthermore, the transgenic hairy roots showed higher antioxidant activity than control. In addition, transgenic hairy root harboring HMGR and DXR (HD42) exhibited higher tanshinone content after elicitation by yeast extract and/or Ag(+) than before. Tanshinone can be significantly enhanced to 5.858, 6.716, and 4.426 mg g(-1) DW by YE, Ag(+), and YE-Ag(+) treatment compared with non-induced HD42, respectively. The content of cryptotanshinone and dihydrotanshinone was effectively elevated upon elicitor treatments, whereas there was no obvious promotion effect for the other two compounds tanshinone I and tanshinone IIA. Our results provide a useful strategy to improve tanshinone content as well as other natural active products by combination of genetic engineering with elicitors.


Subject(s)
Abietanes/biosynthesis , Aldose-Ketose Isomerases/genetics , Hydroxymethylglutaryl CoA Reductases/genetics , Salvia miltiorrhiza/genetics , Abietanes/chemistry , Aldose-Ketose Isomerases/biosynthesis , Biphenyl Compounds/chemistry , Cardiovascular Agents/chemistry , Cardiovascular Agents/metabolism , Free Radical Scavengers/chemistry , Free Radical Scavengers/metabolism , Gene Expression , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Oxidation-Reduction , Picrates/chemistry , Plant Proteins/biosynthesis , Plant Proteins/genetics , Plant Roots/enzymology , Plant Roots/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Salvia miltiorrhiza/enzymology
18.
BMC Microbiol ; 14: 93, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24731286

ABSTRACT

BACKGROUND: Precursors of sterols, carotenoids, the prenyl groups of several proteins and other terpenoid compounds are synthesised via the acetate-mevalonate pathway. One of the key enzyme of this pathway is the 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, which catalyses the conversion of HMG-CoA to mevalonate. HMG-CoA reductase therefore affects many biological processes, such as morphogenesis, synthesis of different metabolites or adaptation to environmental changes. In this study, transcription of the three HMG-CoA reductase genes (designated as hmgR1, hmgR2 and hmgR3) of the ß-carotene producing Mucor circinelloides has been analysed under various culturing conditions; effect of the elevation of their copy number on the carotenoid and ergosterol content as well as on the sensitivity to statins has also been examined. RESULTS: Transcripts of each gene were detected and their relative levels varied under the tested conditions. Transcripts of hmgR1 were detected only in the mycelium and its relative transcript level seems to be strongly controlled by the temperature and the oxygen level of the environment. Transcripts of hmgR2 and hmgR3 are already present in the germinating spores and the latter is also strongly regulated by oxygen. Overexpression of hmgR2 and hmgR3 by elevating their copy numbers increased the carotenoid content of the fungus and decreased their sensitivity to statins. CONCLUSIONS: The three HMG-CoA reductase genes of M. circinelloides displayed different relative transcript levels under the tested conditions suggesting differences in their regulation. They seem to be especially involved in the adaptation to the changing oxygen tension and osmotic conditions of the environment as well as to statin treatment. Overexpression of hmgR2 and hmgR3 may be used to improve the carotenoid content.


Subject(s)
Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Mucor/enzymology , Transcription, Genetic , Carotenoids/metabolism , DNA, Fungal/chemistry , DNA, Fungal/genetics , Ergosterol/metabolism , Gene Expression Profiling , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Molecular Sequence Data , Mucor/genetics , Osmotic Pressure , Oxygen/metabolism , Sequence Analysis, DNA
19.
PLoS One ; 9(1): e87594, 2014.
Article in English | MEDLINE | ID: mdl-24489942

ABSTRACT

The mevalonate pathway is essential in eukaryotes and responsible for a diversity of fundamental synthetic activities. 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGR) is the rate-limiting enzyme in the pathway and is targeted by the ubiquitous statin drugs to treat hypercholesterolemia. Independent reports have indicated the cidal effects of statins against the flatworm parasite, S. mansoni, and the possibility that SmHMGR is a useful drug target to develop new statin-based anti-schistosome therapies. For six commercially available statins, we demonstrate concentration- and time-dependent killing of immature (somule) and adult S. mansoni in vitro at sub-micromolar and micromolar concentrations, respectively. Cidal activity trends with statin lipophilicity whereby simvastatin and pravastatin are the most and least active, respectively. Worm death is preventable by excess mevalonate, the product of HMGR. Statin activity against somules was quantified both manually and automatically using a new, machine learning-based automated algorithm with congruent results. In addition, to chemical targeting, RNA interference (RNAi) of HMGR also kills somules in vitro and, again, lethality is blocked by excess mevalonate. Further, RNAi of HMGR of somules in vitro subsequently limits parasite survival in a mouse model of infection by up to 80%. Parasite death, either via statins or specific RNAi of HMGR, is associated with activation of apoptotic caspase activity. Together, our genetic and chemical data confirm that S. mansoni HMGR is an essential gene and the relevant target of statin drugs. We discuss our findings in context of a potential drug development program and the desired product profile for a new schistosomiasis drug.


Subject(s)
Anthelmintics/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Schistosoma mansoni/drug effects , Schistosomiasis mansoni/drug therapy , Simvastatin/pharmacology , Animals , Apoptosis/drug effects , Drug Evaluation, Preclinical , Female , Gene Knockdown Techniques , Helminth Proteins/biosynthesis , Helminth Proteins/genetics , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/genetics , Inhibitory Concentration 50 , Male , Mevalonic Acid/pharmacology , Mice , Molecular Targeted Therapy , Pravastatin/pharmacology , RNA Interference , Schistosoma mansoni/enzymology , Schistosoma mansoni/genetics , Schistosomiasis mansoni/parasitology
20.
Exp Biol Med (Maywood) ; 239(3): 293-301, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24477821

ABSTRACT

The statins competitively inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase activity and consequently the synthesis of mevalonate. The use of statins is associated with insulin resistance, presumably due to the impaired differentiation and diminished glucose utilization of adipocytes. We hypothesize that mevalonate is essential to adipocyte differentiation and adipogenic gene expression. Adipo-Red assay and Oil Red O staining showed that an eight-day incubation with 0-2.5 µmol/L lovastatin dose-dependently reduced the intracellular triglyceride content of murine 3T3-F442A adipocytes. Concomitantly, lovastatin downregulated the expression of peroxisome proliferator-activated receptor γ (Pparγ), leptin (Lep), fatty acid binding protein 4 (Fabp4), and adiponectin (AdipoQ) as measured by quantitative real-time polymerase chain reaction (real-time qPCR). The expression of sterol regulatory element binding protein 1 (Srebp-1), a transcriptional regulator of Pparγ and Lep genes, was also suppressed by lovastatin. Western-blot showed that lovastatin reduced the level of CCAAT/enhancer binding protein α (C/EBPα) while inducing a compensatory over-expression of HMG CoA reductase. The impact of lovastatin on intracellular triglyceride content and expression of the adipogenic genes was reversed by supplemental mevalonate. Mevalonate-derived metabolites have essential roles in promoting adipogenic gene expression and adipocyte differentiation.


Subject(s)
Adipocytes/metabolism , Adipogenesis/drug effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Mevalonic Acid/metabolism , 3T3 Cells , Adipocytes/drug effects , Adiponectin/biosynthesis , Animals , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Survival , Down-Regulation/drug effects , Fatty Acid-Binding Proteins/biosynthesis , Gene Expression , Hydroxymethylglutaryl CoA Reductases/biosynthesis , Hydroxymethylglutaryl CoA Reductases/drug effects , Insulin Resistance , Leptin/biosynthesis , Mice , PPAR gamma/biosynthesis , Sterol Regulatory Element Binding Protein 1/biosynthesis , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...