1
|
The prognosis of lipid reprogramming with the HMG-CoA reductase inhibitor, rosuvastatin, in castrated Egyptian prostate cancer patients: Randomized trial. PLoS One 2022; 17:e0278282. [PMID: 36480560 PMCID: PMC9731457 DOI: 10.1371/journal.pone.0278282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 10/24/2022] [Indexed: 12/13/2022] Open
Abstract
AIM The role of surgical castration and rosuvastatin treatment on lipid profile and lipid metabolism related markers was evaluated for their prognostic significance in metastatic prostate cancer (mPC) patients. METHODS A total of 84 newly diagnosed castrated mPC patients treated with castration were recruited and divided into two groups: Group I served as control (statin non-users) while group II treated with Rosuvastatin (20 mg/day) for 6 months and served as statin users. Prostate specific antigen (PSA), epidermal growth factor receptor (EGFR), Caveolin-1 (CAV1), lipid profile (LDL, HDL, triglycerides (TG) and total cholesterol (TC)) and lipid metabolism related markers (aldoketoreductase (AKR1C4), HMG-CoA reductase (HMGCR), ATP-binding cassette transporter A1 (ABCA1), and soluble low density lipoprotein receptor related protein 1 (SLDLRP1)) were measured at baseline, after 3 and 6 months. Overall survival (OS) was analyzed by Kaplan-Meier and COX regression for prognostic significance. RESULTS Before castration, HMG-CoA reductase was elevated in patients <65 years (P = 0.009). Bone metastasis was associated with high PSA level (P = 0.013), but low HMGCR (P = 0.004). Patients with positive family history for prostate cancer showed high levels of EGFR, TG, TC, LDL, alkaline phosphatase (ALP), but low AKR1C4, SLDLRP1, CAV1 and ABCA-1 levels. Smokers had high CAV1 level (P = 0.017). After 6 months of castration and rosuvastatin administration, PSA, TG, LDL and TC were significantly reduced, while AKR1C4, HMGCR, SLDLRP1, CAV1 and ABCA-1 were significantly increased. Overall survival was reduced in patients with high baseline of SLDLRP1 (>3385 pg/ml, P = 0.001), PSA (>40 ng/ml, P = 0.003) and CAV1 (>4955 pg/ml, P = 0.021). CONCLUSION Results of the current study suggest that the peripheral lipidogenic effects of rosuvastatin may have an impact on the treatment outcome and survival of castrated mPC patients. TRAIL REGISTRATION This trial was registered at the Pan African Clinical Trial Registry with identification number PACTR202102664354163 and at ClinicalTrials.gov with identification number NCT04776889.
Collapse
|
2
|
Nezhadebrahimi A, Sepehri H, Jahanshahi M, Marjani M, Marjani A. The effect of simvastatin on gene expression of low-density lipoprotein receptor, sterol regulatory element-binding proteins, stearoyl-CoA desaturase 1 mRNA in rat hepatic tissues. Arch Physiol Biochem 2022; 128:1383-1390. [PMID: 32643419 DOI: 10.1080/13813455.2020.1772829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The study aimed to assess the effect of simvastatin on gene expression of LDLR, SREBPs, and SCD1 in rat hepatic tissues fed with high-fat diets (HFD) and its association with some biochemical parameters. Thirty-two male Wister albino rats were divided into four equal groups (three test and one control groups). The biochemical parameters were determined by using spectrophotometer techniques and the Elisa method. Low-density lipoprotein receptor, sterol regulatory element-binding proteins, stearoyl-CoA desaturase1, Beta-actin were analysed by real-time quantitative polymerase chain reaction (RT-PCR) method. At the end of study, the livers of the rats were separated and changes of hepatic tissue were determined. LDLR, SREBP2, and SCD1 expression increased significantly when compared G1 versus G4 and G2 versus G4. The expression of LDLR, SREBP2, and SCD1 also increased significantly when compared G2 versus G3, G1versus G3 and G1 versus G3 and G2 versus G3. The serum level of cholesterol, triglyceride, glucose, LDL, and HDL increased significantly when compared G1 versus G3. LDL showed significantly decreased when compared G1 versus G2. Cholesterol, glucose and HDL and triglyceride levels were increased significantly when compared G1 versus G4 and G2. Treatment of rats with HFD and simvastatin 20 mg/kg, triglyceride and LDL were almost the same as a control group and LDLR expression increased 98% in liver tissue. Gene expressions may be up-regulated in liver tissue and they showed different effects on biochemical parameters.
Collapse
Affiliation(s)
- Abbas Nezhadebrahimi
- Department of Biochemistry and Biophysics, Student Research Center, Metabolic Disorders Research Center, Gorgan Faculty of Medicine, Golestan University Medical Sciences, Gorgan, Iran
- Department of Physiology, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Hamid Sepehri
- Department of Physiology, Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Marjani
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus, Turkey
| | - Abdoljalal Marjani
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University Medical Sciences, Gorgan, Iran
| |
Collapse
|
3
|
Mo H, Jeter R, Bachmann A, Yount ST, Shen CL, Yeganehjoo H. The Potential of Isoprenoids in Adjuvant Cancer Therapy to Reduce Adverse Effects of Statins. Front Pharmacol 2019; 9:1515. [PMID: 30662405 PMCID: PMC6328495 DOI: 10.3389/fphar.2018.01515] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/11/2018] [Indexed: 12/11/2022] Open
Abstract
The mevalonate pathway provides sterols for membrane structure and nonsterol intermediates for the post-translational modification and membrane anchorage of growth-related proteins, including the Ras, Rac, and Rho GTPase family. Mevalonate-derived products are also essential for the Hedgehog pathway, steroid hormone signaling, and the nuclear localization of Yes-associated protein and transcriptional co-activator with PDZ-binding motif, all of which playing roles in tumorigenesis and cancer stem cell function. The phosphatidylinositol-4,5-bisphosphate 3-kinase-AKT-mammalian target of rapamycin complex 1 pathway, p53 with gain-of-function mutation, and oncoprotein MYC upregulate the mevalonate pathway, whereas adenosine monophosphate-activated protein kinase and tumor suppressor protein RB are the downregulators. The rate-limiting enzyme, 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR), is under a multivalent regulation. Sterol regulatory element binding protein 2 mediates the sterol-controlled transcriptional downregulation of HMGCR. UbiA prenyltransferase domain-containing protein-1 regulates the ubiquitination and proteasome-mediated degradation of HMGCR, which is accelerated by 24, 25-dihydrolanosterol and the diterpene geranylgeraniol. Statins, competitive inhibitors of HMGCR, deplete cells of mevalonate-derived intermediates and consequently inhibit cell proliferation and induce apoptosis. Clinical application of statins is marred by dose-limiting toxicities and mixed outcomes on cancer risk, survival and mortality, partially resulting from the statin-mediated compensatory upregulation of HMGCR and indiscriminate inhibition of HMGCR in normal and tumor cells. Tumor HMGCR is resistant to the sterol-mediated transcriptional control; consequently, HMGCR is upregulated in cancers derived from adrenal gland, blood and lymph, brain, breast, colon, connective tissue, embryo, esophagus, liver, lung, ovary, pancreas, prostate, skin, and stomach. Nevertheless, tumor HMGCR remains sensitive to isoprenoid-mediated degradation. Isoprenoids including monoterpenes (carvacrol, L-carvone, geraniol, perillyl alcohol), sesquiterpenes (cacalol, farnesol, β-ionone), diterpene (geranylgeranyl acetone), “mixed” isoprenoids (tocotrienols), and their derivatives suppress the growth of tumor cells with little impact on non-malignant cells. In cancer cells derived from breast, colon, liver, mesothelium, prostate, pancreas, and skin, statins and isoprenoids, including tocotrienols, geraniol, limonene, β-ionone and perillyl alcohol, synergistically suppress cell proliferation and associated signaling pathways. A blend of dietary lovastatin and δ-tocotrienol, each at no-effect doses, suppress the growth of implanted murine B16 melanomas in C57BL6 mice. Isoprenoids have potential as adjuvant agents to reduce the toxicities of statins in cancer prevention or therapy.
Collapse
Affiliation(s)
- Huanbiao Mo
- Department of Nutrition, Byrdine F. Lewis College of Nursing and Health Professions, Georgia State University, Atlanta, GA, United States
| | - Rayna Jeter
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Andrea Bachmann
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sophie T Yount
- Department of Chemistry, Georgia State University, Atlanta, GA, United States
| | - Chwan-Li Shen
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Hoda Yeganehjoo
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
4
|
Low-density lipoprotein receptors play an important role in the inhibition of prostate cancer cell proliferation by statins. Prostate Int 2016; 4:56-60. [PMID: 27358845 PMCID: PMC4916060 DOI: 10.1016/j.prnil.2016.02.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 01/06/2023] Open
Abstract
Background There are some reports about the antitumor effects of statins in these days. Statins decrease the level of cholesterol in the blood by inhibiting 3-hydroxy-3-methylglutaryl-coenzyme A reductase. Inhibition of this enzyme decreases intracellular cholesterol synthesis. Thus, the expression of low-density lipoprotein receptor (LDLr) is increased to import more cholesterol from the bloodstream. In this study, we assessed the effects of statins on the proliferation of prostate cancer cells, and studied the relationship between the expression of LDLr and the effects of statins. Methods Simvastatin was used in the experiments. We studied the effect of simvastatin on PC-3 and LNCaP cell proliferation using the MTS assay, and evaluated the expression of LDLr after administration of simvastatin by quantitative polymerase chain reaction and Western blotting. Intracellular cholesterol levels in the prostate cancer cells were measured after administration of simvastatin. Furthermore, small interfering RNA (siRNA) was used to knockdown the gene expression of LDLr. Results In PC-3 cells, simvastatin inhibited cell proliferation. In LNCaP cells, only a high concentration of simvastatin (100μM) inhibited cell proliferation. In LNCaP cells, the protein level of LDLr was increased by simvastatin. In PC-3 cells, the protein levels of LDLr were unregulated. In PC-3 cells, but not in LNCaP cells, intracellular cholesterol levels were significantly decreased by simvastatin. After knocking down LDLr expression by siRNA, intracellular cholesterol levels were decreased, and cell proliferation was inhibited by simvastatin in LNCaP cells. Conclusion Simvastatin inhibited prostate cancer cell growth by decreasing cellular cholesterol and could be more effective in androgen-independent prostate cancer, where there is loss of regulation of LDLr expression. LDLr was shown to play an important role in the statin-induced inhibition of prostate cancer cell proliferation. These results suggest that future studies evaluating the cholesterol-lowering effects of statin may lead to new approaches to the prevention and treatment of prostate cancer.
Collapse
|
5
|
Kim J, Lee JN, Ye J, Hao R, DeBose-Boyd R, Ye J. Sufficient production of geranylgeraniol is required to maintain endotoxin tolerance in macrophages. J Lipid Res 2013; 54:3430-7. [PMID: 24062519 PMCID: PMC3826689 DOI: 10.1194/jlr.m042549] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/19/2013] [Indexed: 12/19/2022] Open
Abstract
Endotoxin tolerance allows macrophages to produce large quantities of proinflammatory cytokines immediately after their contact with lipopolysaccharides (LPSs), but prevents their further production after repeated exposure to LPSs. While this response is known to prevent overproduction of proinflammatory cytokines, the mechanism through which endotoxin tolerance is established has not been identified. In the current study, we demonstrate that sufficient production of geranylgeraniol (GGOH) in macrophages is required to maintain endotoxin tolerance. We show that increased synthesis of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) protein following LPS treatment is required to produce enough GGOH to inhibit expression of Malt1, a protein known to stimulate expression of proinflammatory cytokines, in macrophages repeatedly exposed to LPSs. Depletion of GGOH caused by inhibition of HMGCR led to increased Malt1 expression in macrophages subjected to repeated exposure to LPSs. Consequently, endotoxin tolerance was impaired, and production of interleukin 1-β and other proinflammatory cytokines was markedly elevated in these cells. These results suggest that insufficient production of GGOH in macrophages may cause autoinflammatory diseases.
Collapse
Affiliation(s)
| | | | - James Ye
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rosy Hao
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Russell DeBose-Boyd
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jin Ye
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
6
|
Sonawane PJ, Sahu BS, Sasi BK, Geedi P, Lenka G, Mahapatra NR. Functional promoter polymorphisms govern differential expression of HMG-CoA reductase gene in mouse models of essential hypertension. PLoS One 2011; 6:e16661. [PMID: 21304971 PMCID: PMC3031630 DOI: 10.1371/journal.pone.0016661] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 01/10/2011] [Indexed: 11/18/2022] Open
Abstract
3-Hydroxy-3-methylglutaryl-coenzyme A [HMG-CoA] reductase gene (Hmgcr) is a susceptibility gene for essential hypertension. Sequencing of the Hmgcr locus in genetically hypertensive BPH (blood pressure high), genetically hypotensive BPL (blood pressure low) and genetically normotensive BPN (blood pressure normal) mice yielded a number of single nucleotide polymorphisms (SNPs). BPH/BPL/BPN Hmgcr promoter-luciferase reporter constructs were generated and transfected into liver HepG2, ovarian CHO, kidney HEK-293 and neuronal N2A cells for functional characterization of the promoter SNPs. The BPH-Hmgcr promoter showed significantly less activity than the BPL-Hmgcr promoter under basal as well as nicotine/cholesterol-treated conditions. This finding was consistent with lower endogenous Hmgcr expression in liver and lower plasma cholesterol in BPH mice. Transfection experiments using 5′-promoter deletion constructs (strategically made to assess the functional significance of each promoter SNP) and computational analysis predicted lower binding affinities of transcription factors c-Fos, n-Myc and Max with the BPH-promoter as compared to the BPL-promoter. Corroboratively, the BPH promoter-luciferase reporter construct co-transfected with expression plasmids of these transcription factors displayed less pronounced augmentation of luciferase activity than the BPL construct, particularly at lower amounts of transcription factor plasmids. Electrophoretic mobility shift assays also showed diminished interactions of the BPH promoter with HepG2 nuclear proteins. Taken together, this study provides mechanistic basis for the differential Hmgcr expression in these mouse models of human essential hypertension and have implications for better understanding the role of this gene in regulation of blood pressure.
Collapse
Affiliation(s)
- Parshuram J. Sonawane
- Cardiovascular Genetics Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Bhavani S. Sahu
- Cardiovascular Genetics Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Binu K. Sasi
- Cardiovascular Genetics Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Parimala Geedi
- Cardiovascular Genetics Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Govinda Lenka
- Cardiovascular Genetics Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Nitish R. Mahapatra
- Cardiovascular Genetics Laboratory, Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
- * E-mail:
| |
Collapse
|
7
|
Holla ØL, Strøm TB, Cameron J, Berge KE, Leren TP. A chimeric LDL receptor containing the cytoplasmic domain of the transferrin receptor is degraded by PCSK9. Mol Genet Metab 2010; 99:149-56. [PMID: 19828345 DOI: 10.1016/j.ymgme.2009.09.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 09/24/2009] [Accepted: 09/24/2009] [Indexed: 11/28/2022]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the extracellular domain of the low density lipoprotein receptor (LDLR) at the cell surface, and disrupts the normal recycling of the LDLR. However, the exact mechanism by which the LDLR is re-routed for lysosomal degradation remains to be determined. To clarify the role of the cytoplasmic domain of the LDLR for re-routing to the lysosomes, we have studied the ability of PCSK9 to degrade a chimeric receptor which contains the extracellular and transmembrane domains of the LDLR and the cytoplasmic domain of the transferrin receptor. These studies were performed in CHO T-REx cells stably transfected with a plasmid encoding the chimeric receptor and a novel assay was developed to study the effect of PCSK9 on the LDLR in these cells. Localization, function and stability of the chimeric receptor were similar to that of the wild-type LDLR. The addition of purified gain-of-function mutant D374Y-PCSK9 to the culture medium of stably transfected CHO T-REx cells showed that the chimeric receptor was degraded, albeit to a lower extent than the wild-type LDLR. In addition, a mutant LDLR, which has the three lysines in the intracellular domain substituted with arginines, was also degraded by D374Y-PCSK9. Thus, the mechanism for the PCSK9-mediated degradation of the LDLR does not appear to involve an interaction between the endosomal sorting machinery and LDLR-specific motifs in the cytoplasmic domain. Moreover, ubiquitination of lysines in the cytoplasmic domain does not appear to play a critical role in the PCSK9-mediated degradation of the LDLR.
Collapse
Affiliation(s)
- Øystein L Holla
- Medical Genetics Laboratory, Department of Medical Genetics, Oslo University Hospital Rikshospitalet, NO-0027 Oslo, Norway
| | | | | | | | | |
Collapse
|
8
|
Sekine Y, Koike H, Nakano T, Nakajima K, Takahashi S, Suzuki K. Remnant lipoproteins induced proliferation of human prostate cancer cell, PC-3 but not LNCaP, via low density lipoprotein receptor. Cancer Epidemiol 2009; 33:16-23. [PMID: 19679042 DOI: 10.1016/j.canep.2009.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 04/02/2009] [Accepted: 04/07/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND Hypertriglyceridemia has been shown to be one of the risk factors for prostate cancer. In this study, we investigated the effect of remnant lipoproteins on cell growth in prostate cancer cell lines. METHODS Remnant lipoproteins were isolated as remnant like particles (RLP) from human plasma. We used RLP for TG-rich lipoproteins and low density lipoproteins (LDL) for cholesterol-rich lipoproteins respectively and examined the effect of lipoproteins on proliferation of PC-3 and LNCaP cells using MTS assays. Moreover, we studied the effect of RLP and LDL treatment on the regulation of lipoprotein receptors in prostate cancer cells to investigate the relationship between lipoprotein-induced cell proliferation and lipoprotein receptor expression using real-time PCR, Western blotting assays and siRNA. RESULTS RLP effectively induced PC-3 cell proliferation more than LDL, whereas both RLP and LDL could not induce LNCaP cell proliferation except at a higher concentration of RLP. LDL receptor (LDLr) was expressed in both prostate cancer cells but there was a sharp difference of sterol regulation between two cells. In PC-3 cells, LDL decreased the LDLr expression in some degree, but RLP did not. Meanwhile LDLr expression in LNCaP was easily downregulated by RLP and LDL. Blocking LDLr function significantly inhibited both RLP- and LDL-induced PC-3 cell proliferation. CONCLUSIONS This study demonstrated that RLP-induced PC-3 cell proliferation more than LDL; however, both RLP and LDL hardly induced LNCaP cell proliferation. The differences of proliferation by lipoproteins might be involved in the regulation of LDLr expression.
Collapse
Affiliation(s)
- Yoshitaka Sekine
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Gunma-pref, Japan
| | | | | | | | | | | |
Collapse
|
9
|
Hall EA, Ren S, Hylemon PB, Redford K, del Castillo A, Gil G, Pandak WM. Mitochondrial cholesterol transport: A possible target in the management of hyperlipidemia. Lipids 2005; 40:1237-44. [PMID: 16477808 DOI: 10.1007/s11745-005-1491-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sterol 27-hydroxylase (CYP27A1) may defend cells against accumulation of excess cholesterol, making this enzyme a possible target in the management of hyperlipidemia. The study objective was to analyze cholesterol homeostatic responses to increases in CYP27A1 activity in HepG2 cells and primary human hepatocytes. Increasing CYP27A1 activity by increasing enzyme expression led to significant increases in bile acid synthesis with compensatory increases in HMG-CoA reductase (HMGR) activity/protein, LDL receptor (LDLR) mRNA, and LDLR-mediated cholesterol uptake. Under these conditions, only a small increase in cellular 27-hydroxycholesterol (27OH-Chol) concentration was observed. No changes were detected in mature sterol regulatory element-binding proteins (SREBP) 1 or 2. Increasing CYP27A1 activity by increasing mitochondrial cholesterol transport (i.e., substrate availability) led to greater increases in bile acid synthesis with significant increases in cellular 27OH-Chol concentration. Mature SREBP 2 protein decreased significantly with compensatory decreases in HMGR protein. No change was detected in mature SREBP 1 protein. Despite increasing 27OH-Chol and lowering SREBP 2 protein concentrations, LDLR mRNA increased significantly, suggesting alternative mechanisms of LDLR transcriptional regulation. These findings suggest that regulation of liver mitochondrial cholesterol transport represents a potential therapeutic strategy in the treatment of hyperlipidemia and atherosclerosis.
Collapse
Affiliation(s)
- E A Hall
- Department of Internal Medicine, Virginia Commonwealth University, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Patalay M, Lofgren IE, Freake HC, Koo SI, Fernandez ML. The lowering of plasma lipids following a weight reduction program is related to increased expression of the LDL receptor and lipoprotein lipase. J Nutr 2005; 135:735-9. [PMID: 15795426 DOI: 10.1093/jn/135.4.735] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To determine whether changes in plasma lipids following a weight loss program were related to modifications in gene expression of the LDL receptor (LDL-R), lipoprotein lipase (LPL), and 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, overweight/obese premenopausal women were recruited. The 10-wk, randomized, double-blind intervention consisted of a hypoenergetic diet, high in protein (30% energy) and low in carbohydrate (40% energy), increased physical activity (number of steps taken per day), and intake of a supplement (carnitine or placebo). Our initial hypothesis was that carnitine would enhance the beneficial effects of weight loss on plasma lipids and anthropometrics. Because the carnitine and placebo groups did not differ in any of the measured variables, data for all subjects were pooled and comparisons were made between baseline and postintervention. Mean weight loss was 4.4 kg (P < 0.001), and plasma triglycerides (TG), total, and LDL cholesterol (LDL-C) were reduced by 31.8, 9.9, and 11.9%, respectively (P < 0.001). The expression of the genes of interest was measured in RNA extracted from mononuclear cells at baseline and postintervention using a semiquantitative RT-PCR method. Glyceraldehyde-3-phosphate dehydrogenase was used as an internal control. After 10 wk, there was a 25.7% increase in the abundance of LPL mRNA (P < 0.01) and a 27.7% increase in that of LDL-R mRNA (P < 0.01). The expression of HMG-CoA reductase was not altered by weight loss. The results suggest that the increased expression of the LDL-R and LPL after the intervention might have contributed to the lower plasma LDL-C and TG observed in these women.
Collapse
Affiliation(s)
- Madhu Patalay
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA
| | | | | | | | | |
Collapse
|
11
|
Begg MJ, Sturrock ED, van der Westhuyzen DR. Soluble LDL-R are formed by cell surface cleavage in response to phorbol esters. ACTA ACUST UNITED AC 2004; 271:524-33. [PMID: 14728679 DOI: 10.1046/j.1432-1033.2003.03953.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A 140-kDa soluble form of the low density lipoprotein (LDL) receptor has been isolated from the culture medium of HepG2 cells and a number of other cell types. It is produced from the 160-kDa mature LDL receptor by a proteolytic cleavage, which is stimulated in the presence of 4beta-phorbol 12-myristate 13-acetate (PMA), leading to the release of a soluble fragment that constitutes the bulk of the extracellular domain of the LDL receptor. By labeling HepG2 cells with [35S]methionine and chasing in the presence of PMA, we demonstrated that up to 20% of LDL-receptors were released into the medium in a 2-h period. Simultaneously, the level of labeled cellular receptors was reduced by 30% in those cells treated with PMA compared to untreated cells, as was the total number of cell surface LDL-receptors assayed by the binding of 125I-labeled antibody to whole cells. To determine if endocytosis was required for cleavage, internalization-defective LDL-receptors were created by mutagenesis or deletion of the NPXY internalization signal, transfected into Chinese hamster ovary cells, and assayed for cleavage in the presence and absence of PMA. Cleavage was significantly greater in the case of the mutant receptors than for wild-type receptors, both in the absence and presence of PMA. Similar results were seen in human skin fibroblasts homozygous for each of the internalization-defective LDL receptor phenotypes. LDL receptor cleavage was inhibited by the hydoxamate-based inhibitor TAPI, indicating the resemblance of the LDL receptor cleavage mechanism to that of other surface released membrane proteins.
Collapse
Affiliation(s)
- Michael J Begg
- Division of Medical Biochemistry, University of Cape Town, South Africa
| | | | | |
Collapse
|
12
|
Vega-López S, Freake HC, Fernandez ML. Sex and hormonal status modulate the effects of psyllium on plasma lipids and monocyte gene expression in humans. J Nutr 2003; 133:67-70. [PMID: 12514268 DOI: 10.1093/jn/133.1.67] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Psyllium (PSY) intake decreases plasma LDL cholesterol (LDL-C) in men and pre- and post-menopausal women while PSY effects on plasma triglycerides (TG) are sex related. A significant decrease in plasma TG was observed in men while postmenopausal women experienced an increase in plasma TG concentrations following PSY supplementation. To further explore the mechanisms by which sex and hormonal status influence the effects of PSY on plasma lipids, HMG-CoA reductase, LDL receptor and lipoprotein lipase (LPL) mRNA abundance were measured in mononuclear cells isolated from these subjects. The intervention followed a randomized crossover design in which participants were allocated to either 15 or 0 g (control) of PSY/d for 30 d. Compared to the control period, PSY intake induced a 20% increase in HMG-CoA reductase mRNA abundance (P < 0.05) while no significant changes in LDL receptor mRNA abundance were observed. In contrast, LPL mRNA abundance was 24% higher in men and 23% lower in postmenopausal women (P < 0.05) when comparing PSY with the control period. These results suggest that the LDL-C lowering induced by PSY was related to changes in HMG-CoA reductase gene expression in monocytes while the expression of LPL in this system was affected by sex and hormonal status.
Collapse
Affiliation(s)
- Sonia Vega-López
- Department of Nutritional Sciences, University of Connecticut, Storrs 06269, USA
| | | | | |
Collapse
|
13
|
Chen Y, Hughes-Fulford M. Human prostate cancer cells lack feedback regulation of low-density lipoprotein receptor and its regulator, SREBP2. Int J Cancer 2001; 91:41-5. [PMID: 11149418 DOI: 10.1002/1097-0215(20010101)91:1<41::aid-ijc1009>3.0.co;2-2] [Citation(s) in RCA: 136] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The low-density lipoprotein receptor (LDLR) pathway provides cells with essential fatty acids for prostaglandin E2 (PGE2) synthesis. Regulation of LDLR expression by LDL was compared between the human normal and cancer prostate cells using semi-quantitative RT-PCR and LDL uptake assays. LDLR mRNA expression and LDL uptake by LDLR were down-regulated in the presence of exogenous LDL or whole serum in the normal prostate cells, but not in the prostate cancer cells. Addition of exogenous cholesterol down-regulated both LDLR and a potent regulator of the ldlr promoter, sterol regulatory element binding protein 2 (SREBP2), in normal cells but not in cancer cells. PGE2 synthesis in prostate cancer cells was significantly increased in response to LDL. Our study suggests that over-production of LDLR is an important mechanism in cancer cells for obtaining more essential fatty acids through LDLR endocytosis, allowing increased synthesis of prostaglandins, which subsequently stimulate cell growth. The data also suggest that the sterol regulatory element and SREBP2 play a role in the loss of sterol feedback regulation in cancer cells.
Collapse
Affiliation(s)
- Y Chen
- Laboratory of Cell Growth, University of California San Francisco and Veterans Affairs Medical Center, USA
| | | |
Collapse
|
14
|
Truong TQ, Auger A, Denizeau F, Brissette L. Analysis of low-density lipoprotein catabolism by primary cultures of hepatic cells from normal and low-density lipoprotein receptor knockout mice. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1484:307-15. [PMID: 10760479 DOI: 10.1016/s1388-1981(00)00023-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Low-density lipoproteins (LDL) are taken up by LDL receptor (LDLr)-dependent and -independent pathways; the role and importance of the latest being less well defined. We analyzed the importance of these pathways in the mouse by comparing LDL binding to primary cultures of hepatocytes from LDLr knockout (LDLr KO) and normal C57BL/6J mice. Saturation curve analysis shows that (125)I-LDL bind specifically to normal and LDLr KO mouse hepatocytes with similar dissociation constants (K(d)) (31.2 and 22.9 microg LDL-protein/ml, respectively). The maximal binding capacity (B(max)) is, however, reduced by 48% in LDLr KO mouse hepatocytes in comparison to normal hepatocytes. Conducting the assay in the presence of a 200-fold excess of high-density lipoprotein-3 (HDL3) reduced by 39% the binding of (125)I-LDL to normal hepatocytes and abolished the binding to the LDLr KO mouse hepatocytes. These data indicate that in normal mouse hepatocytes, the LDLr is responsible for approximately half of the LDL binding while a lipoprotein binding site (LBS), interacting with both LDL and HDL3, is responsible for the other half. It can also be deduced that both receptors/sites have a similar affinity for LDL. The metabolism of LDL-protein and cholesteryl esters (CE) was analyzed in both types of cells. (125)I-LDL-protein degradation was reduced by 95% in LDLr KO hepatocytes compared to normal hepatocytes. Comparing the association of (125)I-LDL and (3)H-CE-LDL revealed a CE-selective uptake of 35.6- and 22-fold for normal and LDLr KO mouse hepatocytes, respectively. Adding a 200-fold excess of HDL3 in the assay reduced by 71% the CE-selective uptake in LDLr KO hepatocytes and by 96% in normal hepatocytes. This indicates that mouse hepatocytes are able to selectively take up CE from LDL by the LBS. The comparison of LDL-CE association also showed that the LBS pathway provides 5-fold more LDL-CE to the cell than the LDLr. Overall, our results indicate that in mouse hepatocytes, LDLr is almost completely responsible for LDL-protein degradation while the LBS is responsible for the major part of LDL-CE entry by a CE-selective uptake pathway.
Collapse
Affiliation(s)
- T Q Truong
- Département des Sciences Biologiques, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montréal, Que., Canada
| | | | | | | |
Collapse
|
15
|
Abstract
Oxygenated derivatives of cholesterol (oxysterols) present a remarkably diverse profile of biological activities, including effects on sphingolipid metabolism, platelet aggregation, apoptosis, and protein prenylation. The most notable oxysterol activities center around the regulation of cholesterol homeostasis, which appears to be controlled in part by a complex series of interactions of oxysterol ligands with various receptors, such as the oxysterol binding protein, the cellular nucleic acid binding protein, the sterol regulatory element binding protein, the LXR nuclear orphan receptors, and the low-density lipoprotein receptor. Identification of the endogenous oxysterol ligands and elucidation of their enzymatic origins are topics of active investigation. Except for 24, 25-epoxysterols, most oxysterols arise from cholesterol by autoxidation or by specific microsomal or mitochondrial oxidations, usually involving cytochrome P-450 species. Oxysterols are variously metabolized to esters, bile acids, steroid hormones, cholesterol, or other sterols through pathways that may differ according to the type of cell and mode of experimentation (in vitro, in vivo, cell culture). Reliable measurements of oxysterol levels and activities are hampered by low physiological concentrations (approximately 0.01-0.1 microM plasma) relative to cholesterol (approximately 5,000 microM) and by the susceptibility of cholesterol to autoxidation, which produces artifactual oxysterols that may also have potent activities. Reports describing the occurrence and levels of oxysterols in plasma, low-density lipoproteins, various tissues, and food products include many unrealistic data resulting from inattention to autoxidation and to limitations of the analytical methodology. Because of the widespread lack of appreciation for the technical difficulties involved in oxysterol research, a rigorous evaluation of the chromatographic and spectroscopic methods used in the isolation, characterization, and quantitation of oxysterols has been included. This review comprises a detailed and critical assessment of current knowledge regarding the formation, occurrence, metabolism, regulatory properties, and other activities of oxysterols in mammalian systems.
Collapse
Affiliation(s)
- G J Schroepfer
- Departments of Biochemistry, Rice University, Houston, Texas, USA.
| |
Collapse
|
16
|
Di Croce L, Vicent GP, Pecci A, Bruscalupi G, Trentalance A, Beato M. The promoter of the rat 3-hydroxy-3-methylglutaryl coenzyme A reductase gene contains a tissue-specific estrogen-responsive region. Mol Endocrinol 1999; 13:1225-36. [PMID: 10446899 DOI: 10.1210/mend.13.8.0333] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The isoprenoid metabolic pathway is mainly regulated at the level of conversion of 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) to mevalonate, catalyzed by HMG CoA reductase. As estrogens are known to influence cholesterol metabolism, we have explored the potential regulation of the HMG CoA reductase gene promoter by estrogens. The promoter contains an estrogen-responsive element-like sequence at position -93 (termed Red-ERE), which differs from the ERE consensus by one mismatch in each half of the palindrome. A Red-ERE oligonucleotide specifically bound estrogen receptor in vitro and conferred receptor-dependent estrogen responsiveness to a heterologous promoter in all cell lines tested. However, expression of a reporter driven by the rat HMG CoA reductase promoter was induced by estrogen treatment after transient transfection into the breast cancer cell line MCF-7 cells but not in hepatic cell lines expressing estrogen receptor. Estrogen induction in MCF-7 cells was dependent on the Red-ERE and was strongly inhibited by the antiestrogen ICI 164,384. A functional cAMP-responsive element is located immediately upstream of the Red-ERE, but cAMP and estrogens inhibit each other in terms of transactivation of the promoter. Similarly, induction by estrogens was inhibited by micromolar concentrations of cholesterol, likely acting via changes in occupancy of the sterol-responsive element located 70 bp upstream of the Red-ERE. Thus, within its natural context, Red-ERE is able to mediate hormonal regulation of the HMG CoA reductase gene in tissues that respond to estrogens with enhanced cell proliferation, while it is not operative in liver cells. We postulate that this tissue-specific regulation of HMG CoA reductase by estrogens could partially explain the protective effect of estrogens against heart disease.
Collapse
Affiliation(s)
- L Di Croce
- Institute for Molecular Biology and Tumor Research (IMT), Marburg, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Charest MC, Rhainds D, Falstrault L, Matzouranis T, Brissette L. Selective uptake of cholesteryl ester from low density lipoprotein is involved in HepG2 cell cholesterol homeostasis. EUROPEAN JOURNAL OF BIOCHEMISTRY 1999; 263:402-9. [PMID: 10406948 DOI: 10.1046/j.1432-1327.1999.00527.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Low density lipoprotein (LDL) can follow either a holoparticle uptake pathway, initiated by the LDL receptor (LDLr), and be completely degraded, or it can deliver its cholesteryl esters (CE) selectively to HepG2 cells. Although high density lipoprotein-CE selective uptake has been shown to be linked to cell cholesterol homeostasis in nonhepatic cells, there is no available information on the effect of LDL-CE selective uptake on hepatic cell cholesterol homeostasis. In order to define the role of the LDL-CE selective uptake pathway in hepatic cell cholesterol homeostasis, we used a cellular model that expresses constitutively a LDLr antisense mRNA and that shows LDLr activity at 31% the normal level (HepG2-all cells). The addition of a specific antibody anti-LDLr (IgG-C7) reduces LDL protein degradation (LDLr activity) to 7%. This cellular model therefore reflects, above all, LDL-CE selective uptake activity when incubated with LDL. The inactivation of LDLr reduces LDL-protein association by 78% and LDL-CE association by only 43%. The LDL-CE selective uptake was not reduced by the inactivation of LDLr. The activities of the various enzymes involved in cell cholesterol homeostasis were measured in normal and LDLr-deficient cells during incubation in the absence or presence of LDL as a cholesterol source. Essentially, 3-hydroxy-3-methylglutaryl coenzyme A reductase and acyl coenzyme A:cholesterol acyltransferase (ACAT) activities responded to LDL in LDLr-deficient cells as well as in normal HepG2 cells. Inhibition of lysosomal hydrolysis with chloroquine abolished the effect measured on ACAT activity in the presence of LDL, suggesting that CE of LDL, but not free cholesterol, maintains cell cholesterol homeostasis. Thus, in HepG2 cells, when LDLr function is virtually abolished, LDL-CE selective uptake is coupled to cell cholesterol homeostasis.
Collapse
Affiliation(s)
- M C Charest
- Département des Sciences Biologiques, Université du Québec à Montréal, Canada
| | | | | | | | | |
Collapse
|
18
|
Truong TQ, Falstrault L, Tremblay C, Brissette L. Low density lipoprotein-receptor plays a major role in the binding of very low density lipoproteins and their remnants on HepG2 cells. Int J Biochem Cell Biol 1999; 31:695-705. [PMID: 10404642 DOI: 10.1016/s1357-2725(99)00014-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The binding to HepG2 cells of very low density lipoproteins (VLDL) and their remnants (IDL) was alternatively, in the past, attributed to the low density lipoprotein receptor (LDLr) or to an apoE-specific receptor. In order to resolve this issue, we have compared the binding of those lipoproteins labelled with iodine-125 to normal and LDLr deficient HepG2 cells. Those deficient cells were obtained by a constitutive antisense strategy and their LDLr level is 14% the level of normal HepG2 cells. By saturation curve analysis, we show that VLDL and IDL bind to high and low affinity sites on cells. The low affinity binding was eliminated by conducting the assay in presence of a 200-fold excess of HDL3 respective to the concentrations of 125I-labelled VLDL and IDL. For 125I-VLDL high affinity binding to normal HepG2 cells, we found a dissociation constant (Kd) of 21.2 +/- 3.7 micrograms prot./ml (S.E., N = 5) and a maximal binding capacity (Bmax) of 0.0312 +/- 0.0063 microgram prot./mg cell prot, while we have measured a Kd of 5.3 +/- 0.8 and a Bmax of 0.0081 +/- 0.0014 with LDLr deficient cells. This indicates that LDLr is responsible for 74% of VLDL binding to HepG2 cells and that the non-LDLr high affinity receptor has a higher affinity for VLDL than LDLr. A 53% loss of 125I-IDL binding capacity was measured with LDLr deficient cells compared with normal cells (Bmax: 0.028 +/- 0.005 versus 0.059 +/- 0.006), while no significant statistical difference was found between affinities. The study shows that the LDLr is almost the only contributor in VLDL binding, while it shares IDL binding capacity with another high affinity receptor. The physiological importance of LDLr is confirmed by an almost equivalent loss of IDL and VLDL degradation in LDLr deficient cells.
Collapse
Affiliation(s)
- T Q Truong
- Département des Sciences Biologiques, Université du Québec à Montréal, Canada
| | | | | | | |
Collapse
|
19
|
Allen S, Khan S, Tam SP, Koschinsky M, Taylor P, Yacoub M. Expression of adhesion molecules by lp(a): a potential novel mechanism for its atherogenicity. FASEB J 1998; 12:1765-76. [PMID: 9837867 DOI: 10.1096/fasebj.12.15.1765] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Lp(a) is a major inherited risk factor for premature atherosclerosis. The mechanism of Lp(a) atherogenicity has not been elucidated, but likely involves both its ability to interfere with plasminogen activation and its atherogenic potential as a lipoprotein particle after receptor-mediated uptake. We demonstrate that Lp(a) stimulates production of vascular cell adhesion molecule 1 (VCAM-1) and E-selectin in cultured human coronary artery endothelial cells (HCAEC). This effect resulted from a rise in intracellular free calcium induced by Lp(a) and could be inhibited by the intracellular calcium chelator, BAPTA/AM. The involvement of the LDL and VLDL receptors in Lp(a) activation of HCAEC were ruled out since Lp(a) induction of adhesion molecules was not prevented by an antibody (IgGC7) to the LDL receptor or by receptor-activating protein, an antagonist of ligand binding to the VLDL receptor. Addition of alpha2-macroglobulin as well as treatment with heparinase, chondroitinase ABC, and sodium chlorate did not decrease levels of VCAM-1 and E-selectin stimulated by Lp(a), suggesting that neither the low density lipoprotein receptor-related protein nor cell-surface proteoglycans are involved in Lp(a)-induced adhesion molecule production. Neither does the binding site on HCAEC responsible for adhesion molecule production by Lp(a) appear to involve plasminogen receptors, as levels of VCAM-1 and E-selectin were not significantly decreased by the addition of glu-plasminogen, the lysine analog epsilon-aminocaproic acid, or by trans-4-(aminomethyl)-cyclohexanecarboxymethylic acid (tranexamic acid), which acts by binding to the lysine binding sites carried on the kringle structures in plasminogen. In contrast, recombinant apolipoprotein (a) [r-apo(a)] competed with Lp(a) and attenuated the expression of VCAM-1 and E-selectin. In summary, we have identified a calcium-dependent interaction of Lp(a) with HCAEC capable of inducing potent surface expression of VCAM-1 and E-selectin that does not appear to involve any of the known potential Lp(a) binding sites. Because leukocyte recruitment to the vessel wall appears to represent one of the important early events in atherogenesis, this newly described endothelial cell-activating effect of Lp(a) places it at a crucial juncture in the initiation of atherogenic disease and may lead to a better understanding of the role of Lp(a) in the vascular biology of atherosclerosis.
Collapse
Affiliation(s)
- S Allen
- Department of Cardiothoracic Surgery, Imperial College of Science, Technology and Medicine, Royal Brompton and Harefield NHS Trust Hospital, Harefield Hospital, Harefield, Middlesex UB9 6JH, United Kingdom
| | | | | | | | | | | |
Collapse
|
20
|
Peffley DM, Gayen AK, Morand OH. Down-regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase mRNA levels and synthesis in syrian hamster C100 cells by the oxidosqualene cyclase inhibitor [4'-(6-allyl-ethyl-amino-hexyloxy)-2'-fluoro-phenyl]-(4-bromophenyl)-me thanone (Ro 48-8071): comparison to simvastatin. Biochem Pharmacol 1998; 56:439-49. [PMID: 9763219 DOI: 10.1016/s0006-2952(98)00083-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In vivo inhibition of 2,3-oxidosqualene:lanosterol cyclase (OSC, E.C. 5.4.99.7)--the enzyme which catalyzes the cyclization of monooxidosqualene to lanosterol--does not result in elevated 3-hydroxy-3-methylglutaryl CoA reductase (HMGR) activity. This trait is attributed to increased levels of oxysterols, produced upon partial inhibition of OSC, that suppress HMGR and other sterol-responsive genes. The OSC inhibitor [4'-(6-allyl-ethyl-amino-hexyloxy)-2'-fluoro-phenyl]-(4-bromopheny l)-methanone (Ro 48-8071) was shown earlier to lower low-density lipoprotein (LDL) cholesterol in hamsters with no increase in hepatic HMGR, in contrast to simvastatin. To delineate the regulatory mechanism(s) by which Ro 48-8071 reduces cholesterol synthesis without raising HMGR levels, Syrian hamster C100 cells were incubated with either Ro 48-8071 or simvastatin, and their effects on cholesterol synthesis and LDL uptake, as well as on HMGR mRNA levels and rates of synthesis, were determined. Using RNase protection and radioimmunoprecipitation assays, we found that, in the absence of LDL in the culture medium, both HMGR mRNA levels and synthesis were reduced with concentrations of Ro 48-8071 inhibiting cholesterol synthesis by 50-75%, whereas LDL uptake was either reduced or unchanged. In contrast, simvastatin, at concentrations inhibiting cholesterol synthesis by the same 50-75%, increased both HMGR mRNA levels and synthesis, as well as LDL uptake. In the presence of LDL, HMGR mRNA levels and synthesis along with LDL uptake were little affected after incubation with Ro 48-8071. Still, simvastatin markedly increased both HMGR mRNA levels and synthesis in cells incubated in the presence of LDL, leaving LDL uptake unaffected. These data suggest that inhibition of OSC by Ro 48-8071 results in an indirect down-regulation of HMGR mRNA levels and synthesis.
Collapse
Affiliation(s)
- D M Peffley
- Department of Pharmacology and Molecular Biology, Finch University of Health Sciences-The Chicago Medical School, IL, USA
| | | | | |
Collapse
|
21
|
Gauthier BR. Characterization of a novel liver-specific protein/DNA binding site in the human HMG CoA reductase promoter. Biochem Biophys Res Commun 1998; 247:280-6. [PMID: 9642117 DOI: 10.1006/bbrc.1998.8777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
These studies define a novel binding element (site C) within the human HMG CoA reductase promoter using a combination of in vitro DNase I footprinting and gel mobility shift assays. The factor interacting with site C appears to be restricted to the liver, indicating a possible role for this protein in regulating hepatic expression of the gene. Studies based on competitive gel mobility shift assays and transient co-transfection experiments performed using a reporter construct harbouring the promoter of HMG CoA reductase suggest that the protein binding to site C may belong to the C/EBP family of transcription factors. A factor interacting with this binding element was also identified in human liver nuclear protein extracts.
Collapse
Affiliation(s)
- B R Gauthier
- Cancer Research Laboratories and Department of Biochemistry, Queen's University, Kingston, Ontario, K7L 3N6, Canada.
| |
Collapse
|
22
|
Wilson G, Vasa M, Deeley R. Stabilization and cytoskeletal-association of LDL receptor mRNA are mediated by distinct domains in its 3′ untranslated region. J Lipid Res 1998. [DOI: 10.1016/s0022-2275(20)33870-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
23
|
Tam SP, Zhang X, Cuthbert C, Wang Z, Ellis T. Effects of dimethyl sulfoxide on apolipoprotein A-I in the human hepatoma cell line, HepG2. J Lipid Res 1997. [DOI: 10.1016/s0022-2275(20)37139-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
24
|
Wilson GM, Roberts EA, Deeley RG. Modulation of LDL receptor mRNA stability by phorbol esters in human liver cell culture models. J Lipid Res 1997. [DOI: 10.1016/s0022-2275(20)37252-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
25
|
Yu L, Qiu S, Genest J. Abnormal regulation of the LDL-R and HMG CoA reductase genes in subjects with familial hypercholesterolemia with the "French Canadian mutation". Atherosclerosis 1996; 124:103-17. [PMID: 8800498 DOI: 10.1016/0021-9150(96)05828-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Familial hypercholesterolemia (FH) is seen with high frequency in the province of Québec, Canada. A large deletion (> 10 kb) of the 5'-end of the low density lipoprotein receptor (LDL-R) gene is the major mutation of the LDL-R in FH subjects in Québec (approximately 60% of FH subjects). No mRNA is produced from the allele bearing the mutation, and cellular cholesterol obtained by receptor-mediated endocytosis is under the control of the non-deletion allele. We have previously reported that some patients with the 10-kb deletion (approximately 9%) fail to respond to the hydroxymethylglutaryl coenzyme A reductase (HMG CoA reductase) inhibitor class of medications. We studied mRNA levels of the LDL-R and HMG CoA reductase genes in response to the HMG CoA reductase inhibitor lovastatin in a time- and dose-dependent fashion in cultured human skin fibroblasts and we devised an in vitro model to study the response to drug therapy in subjects with FH. We determined mRNA levels by RNase protection assay in skin fibroblasts obtained from controls (n = 3) and FH subjects with the > 10-kb deletion (responders, n = 3; non responders, n = 3; to drug therapy). We measured 125I-LDL binding on skin fibroblasts grown in the presence of lipoprotein-deficient serum with or without 1 microM lovastatin, using 10 micrograms/mL of 125I-LDL protein. Control subjects exhibited coordinate regulation of the LDL-R and HMG CoA reductase genes in response to lovastatin, 0.1-25 microM, for 0-24 h. Correlation coefficients between mRNA levels of both genes were > 0.9 in controls and FH subjects. However, by linear regression analysis, the corresponding slopes for the correlation between both genes were 0.98 (controls), 3.36 and 3.63 (FH responders and non-responders), indicating a pattern of dissociated but still coordinate regulation in FH subjects. The magnitude of increase of mRNA levels of the LDL-R gene was approximately five-fold over LPDS in controls, two-fold in FH responders and two-fold in non-responders. Binding studies using 125I-LDL reveal that a control subject and all responders had a 2-2.5-fold increase in binding to cell surface receptors but two out of three FH non-responders showed no increase in binding in response to 1 microM lovastatin. The LDL-R and HMG CoA reductase genes are expressed in coordinate regulation in fibroblasts from subjects with FH due to the > 10-kb deletion, but with a proportionately greater up-regulation of the HMG CoA reductase gene. Some subjects, with FH caused by the > 10-kb deletion of the LDL-R gene, who fail to respond to HMG CoA reductase inhibitors have abnormal LDL receptor binding activity at the cell surface in response to lovastatin in vitro.
Collapse
Affiliation(s)
- L Yu
- Cardiovascular Genetics Laboratory, Clinical Research Institute of Montréal, Québec Canada
| | | | | |
Collapse
|
26
|
Fujioka T, Nara F, Shimada Y, Fukushige J, Shimotsu H, Shigehara E, Fukami M, Tsujita Y. The mechanism of comparable serum cholesterol lowering effects of pravastatin sodium, a 3-hydroxy-3-methylglutaryl coenzyme A inhibitor, between once- and twice-daily treatment regimens in beagle dogs and rabbits. JAPANESE JOURNAL OF PHARMACOLOGY 1996; 70:329-35. [PMID: 8774761 DOI: 10.1254/jjp.70.329] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In dogs, no significant difference in the reduction of serum cholesterol was observed among three dosing regimens of pravastatin: once in the morning (3 mg/kg), once in the evening (3 mg/kg), and twice-daily (1.5 mg/kg x 2) for 21 days. In rabbits, pravastatin was administered at a dose of 50 mg/kg once-daily given in the evening or 25 mg/kg twice-daily for 14 days; the respective serum and liver cholesterol levels were decreased by 41% and 7% in the once-daily dosing and by 51% and 11% in the twice-daily dosing. The amount of low density lipoprotein (LDL) receptor protein was increased 1.2-1.3-fold (P < 0.05) by both treatments, and no significant difference was noted between these treatment regimens. In addition, there was no significant difference in the extent of up-regulated LDL receptor protein between once-daily dosing in the evening and once-daily dosing in the morning. In the experiments with rabbit hepatocytes, the up-regulated LDL receptor activity induced by preincubation with pravastatin was retained even 24 hr after the removal of pravastatin. These results suggest that the comparable efficacy of pravastatin between once- and twice-daily treatment could be explained by retention of up-regulated LDL receptor activity for more than 24 hr in vitro and in vivo.
Collapse
Affiliation(s)
- T Fujioka
- Pharmacology Research Laboratory, Sankyo Co., Ltd., Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Interaction of a recombinant form of apolipoprotein[a] with human fibroblasts and with the human hepatoma cell line HepG2. J Lipid Res 1996. [DOI: 10.1016/s0022-2275(20)37595-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
28
|
Srivastava RA, Ito H, Hess M, Srivastava N, Schonfeld G. Regulation of low density lipoprotein receptor gene expression in HepG2 and Caco2 cells by palmitate, oleate, and 25-hydroxycholesterol. J Lipid Res 1995. [DOI: 10.1016/s0022-2275(20)39730-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
29
|
Ramharack R, Spahr M, Hicks G, Kieft K, Brammer D, Minton L, Newton R. Gemfibrozil significantly lowers cynomolgus monkey plasma lipoprotein[a]-protein and liver apolipoprotein[a] mRNA levels. J Lipid Res 1995. [DOI: 10.1016/s0022-2275(20)41137-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
30
|
Peffley DM, Gayen AK. Mevalonate regulates polysome distribution and blocks translation-dependent suppression of 3-hydroxy-3-methylglutaryl coenzyme A reductase mRNA: relationship to translational control. SOMATIC CELL AND MOLECULAR GENETICS 1995; 21:189-204. [PMID: 7482033 DOI: 10.1007/bf02254770] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We reported previously that 3-hydroxy-3-methylglutaryl coenzyme A reductase synthesis is regulated at the translational level by mevalonate. To determine at what stage mevalonate affects reductase synthesis, we examined the distribution of reductase mRNA in polysomes from cells treated with lovastatin alone; lovastatin and 25-hydroxycholesterol; or lovastatin, 25-hydroxycholesterol, and mevalonate. In lovastatin-treated cells, reductase mRNA was primarily associated with heavy polysome fractions. When 25-hydroxycholesterol was added to lovastatin-treated cells, reductase mRNA levels were reduced approximately fourfold in all polysome fractions, with no accompanying redistribution of reductase mRNA into lighter polysome fractions. However, addition of both 25-hydroxycholesterol and mevalonate to lovastatin-treated cells shifted reductase mRNA from heavier to lighter polysome fractions. No change in the distribution of control beta-actin or ribosomal protein S17 mRNA occurred with any of the treatments. These results suggest that mevalonate suppresses reductase synthesis at the level of initiation. When the translation inhibitor cycloheximide was added to all three regimens, reductase mRNA shifted into heavy polysome fractions. Treatment with either lovastatin alone or lovastatin plus 25-hydroxycholesterol resulted in a 50% greater loss of reductase mRNA from the heavy polysome fractions compared to the same fractions from noncycloheximide-treated cells. No loss of reductase mRNA occurred when cycloheximide was added to cells treated with both 25-hydroxycholesterol and mevalonate. beta-Actin mRNA levels and polysome distribution were not significantly changed by cycloheximide under any of these conditions. Translationally mediated suppression of reductase mRNA did not occur when protein synthesis was inhibited with puromycin. Our results indicate that regulation of reductase mRNA levels is translation-dependent and is linked to the rate of elongation.
Collapse
Affiliation(s)
- D M Peffley
- Department of Pharmacology and Molecular Biology, Finch University of Health Sciences, Chicago Medical School, Illinois 60064, USA
| | | |
Collapse
|
31
|
Rumsey SC, Galeano NF, Lipschitz B, Deckelbaum RJ. Oleate and other long chain fatty acids stimulate low density lipoprotein receptor activity by enhancing acyl coenzyme A:cholesterol acyltransferase activity and altering intracellular regulatory cholesterol pools in cultured cells. J Biol Chem 1995; 270:10008-16. [PMID: 7730302 DOI: 10.1074/jbc.270.17.10008] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Modification of dietary fatty acid composition results in changes in plasma cholesterol levels in man. We examined the effect of in vitro fatty acid supplementation on low density lipoprotein (LDL) receptor activity in cultured cells and questioned whether changes were related to fatty acid-induced alterations in acyl-CoA: cholesterol acyltransferase (ACAT) activity. Preincubation of cultured cells (i.e. human skin fibroblasts, J774 macrophages, and HepG2 cells) with oleic acid (oleic acid:bovine serum albumin molar ratio 2:1) at 37 degrees C for longer than 2 h resulted in a 1.2- to 1.5-fold increase in LDL cell binding at 4 degrees C and LDL cell degradation at 37 degrees C. Scatchard analysis showed that oleic acid increased LDL receptor number but not LDL affinity (Kd). Fatty acid supplementation of J774 macrophages increased both LDL receptor activity and cholesteryl ester accumulation. The ACAT inhibitor, 58-035, eliminated both effects, and increased ACAT activity preceded stimulation of LDL receptor activity by 1-2 h. Supplementation of macrophages with triolein emulsion particles also increased LDL cell binding and degradation, and addition of cholesterol to the emulsions abolished this effect. Among fatty acids tested, oleate (18:1), arachidonate (20:4), and eicosapentanoate (20:5) demonstrated the greatest effects. We hypothesize that certain fatty acids delivered to cells either in free form, or as triglyceride, first increase cellular ACAT activity, which then causes a decrease in an intracellular free cholesterol pool, signaling a need for increased LDL receptor activity. This mechanism may play a role in the effect of certain dietary fatty acids on LDL metabolism in vivo.
Collapse
Affiliation(s)
- S C Rumsey
- Institute of Human Nutrition, Columbia University, New York, New York 10032, USA
| | | | | | | |
Collapse
|
32
|
Hsu H, Nicholson A, Hajjar D. Basic fibroblast growth factor-induced low density lipoprotein receptor transcription and surface expression. Signal transduction pathways mediated by the bFGF receptor tyrosine kinase. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)37096-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
33
|
de Knijff P, van den Maagdenberg AM, Frants RR, Havekes LM. Genetic heterogeneity of apolipoprotein E and its influence on plasma lipid and lipoprotein levels. Hum Mutat 1994; 4:178-94. [PMID: 7833947 DOI: 10.1002/humu.1380040303] [Citation(s) in RCA: 91] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Apolipoprotein E (apoE) is one of the major protein constituents of chylomicron and very-low-density lipoprotein (VLDL) remnants and plays a central role as a ligand in the receptor-mediated uptake of these particles by the liver. Including the most common variant, apoE3, 30 apoE variants have been characterized. At present, 14 apoE variants have been found to be associated with familial dysbetalipoproteinemia, a genetic lipid disorder characterized by elevated plasma cholesterol and triglyceride levels and an increased risk for atherosclerosis. Seven apoE variants were found to be associated with other forms of hyperlipoproteinemia. This report presents an overview of all currently known apoE variants and their effects on lipoprotein metabolism.
Collapse
Affiliation(s)
- P de Knijff
- TNO Institute of Prevention and Health Research, Gaubius Laboratory, Leiden, The Netherlands
| | | | | | | |
Collapse
|
34
|
Kraemer FB, Laane C, Park B, Sztalryd C. Low-density lipoprotein receptors in rat adipocytes: regulation with fasting. THE AMERICAN JOURNAL OF PHYSIOLOGY 1994; 266:E26-32. [PMID: 8304441 DOI: 10.1152/ajpendo.1994.266.1.e26] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Adipose tissue metabolism is exquisitely sensitive to caloric intake. With increasing adiposity more triglyceride and cholesterol are stored within increasingly large adipocytes, whereas less triglyceride and cholesterol are stored as the size of the fat cell decreases. A portion of the uptake of cholesterol by adipocytes is mediated by low-density lipoprotein (LDL) receptors. The present studies addressed whether LDL receptors are differentially regulated in adipose tissue and the liver during fasting in the rat. Two days of fasting caused a reduction in body weight with an approximately 40% decrease in the epididymal fat depot and fat cell size. No changes in serum cholesterol were noted, but serum triglycerides fell approximately 55% with fasting. LDL receptors detected by immunoblotting decreased progressively with fasting to levels that were 95% below controls in adipocytes isolated from epididymal fat pads by 2-3 days. In contrast, hepatic LDL receptor expression was unaltered by fasting. After 2 days of fasting, the rate of synthesis of LDL receptors in isolated adipose cells was decreased approximately 35%, whereas levels of LDL receptor mRNA were diminished approximately 55%. It is concluded that the expression of LDL receptors in rat adipocytes is markedly downregulated during fasting through transcriptional and posttranscriptional mechanisms. Furthermore, LDL receptor expression is differentially regulated in adipose tissue and liver during fasting in the rat.
Collapse
Affiliation(s)
- F B Kraemer
- Veterans Affairs Medical Center, Palo Alto 94304
| | | | | | | |
Collapse
|
35
|
|
36
|
Bennis F, Favre G, Le Gaillard F, Soula G. Importance of mevalonate-derived products in the control of HMG-CoA reductase activity and growth of human lung adenocarcinoma cell line A549. Int J Cancer 1993; 55:640-5. [PMID: 8406993 DOI: 10.1002/ijc.2910550421] [Citation(s) in RCA: 59] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
HMG-CoA reductase catalyzes the synthesis of mevalonate, a crucial intermediate in the biosynthesis of cholesterol and non-sterol isoprenoid compounds essential for cell growth. The HMG-CoA reductase activity of the A549 tumor cell line is higher than that of normal human fibroblasts. This deregulation in mevalonate needs was not due to an alteration in the activated state of the enzyme by short-term regulation. We show that the HMG-CoA reductase in A549 cell line was subject to a multivalent feedback control. A high fraction (40%) of the reductase activity was devoted to non-sterol products. In contrast, normal fibroblasts had only 15-20% of the reductase activity that generated non-sterol products. We also show that cholesterol and at least one of the non-sterol products are necessary for optimal cell growth of A549 cells. Our data strongly suggest that A549 cells produce more non-sterol substances which may be related to increased requirements of mevalonate for upregulated cell growth.
Collapse
Affiliation(s)
- F Bennis
- Laboratoire de Ciblage en Thérapeutique, Biologie de la Cellule Tumorale, UFR des Sciences Pharmaceutiques (Université Paul Sabatier), Toulouse, France
| | | | | | | |
Collapse
|
37
|
Srivastava RA, Baumann D, Schonfeld G. In vivo regulation of low-density lipoprotein receptors by estrogen differs at the post-transcriptional level in rat and mouse. EUROPEAN JOURNAL OF BIOCHEMISTRY 1993; 216:527-38. [PMID: 8375391 DOI: 10.1111/j.1432-1033.1993.tb18171.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Rats and mice are frequently used in studies of the regulation of lipoprotein metabolism. Although the species are closely related, they differ dramatically in the responses of their lipoproteins to estrogen administration. In rats, estrogens produce profound decreases in the levels of all plasma lipoproteins and this is attributed largely to estrogen-induced increases of hepatic low-density lipoprotein receptor (LDL-receptor) activity. Estrogens affect mouse plasma lipoproteins to a much lesser extent. Therefore, one of our aims was to compare the regulation of LDL-receptor gene expression in rats and mice at several potential loci of regulation. To assess the specificity of the estrogen effect, we also compared the responses of apolipoprotein AI (apoAI), apolipoprotein B (apoB), and beta-actin to the response of the LDL-receptor. In male Sprague Dawley rats given 17 beta-estradiol or 17 alpha-ethinyl estradiol at supraphysiological doses of 5 micrograms/g body mass/day, plasma total cholesterol and triacylglycerols fell to approximately 5% and approximately 50%, and, plasma apoAI and apoB fell to approximately 12% and approximately 16% of controls, respectively. By contrast, in male C3H/HeJ mice the above parameters dropped only to approximately 65% of controls and apoB concentrations rose to approximately 200% of controls. In rats, relative rates of LDL-receptor mRNA transcription (nuclear 'run-off' assay) and total hepatic, nuclear and polysomal LDL-receptor mRNA levels (RNase protection assay) increased by 1.5-2-fold, while synthesis of LDL-receptor protein on hepatic polysomes (in a wheat-germ translation system) increased 8-fold and LDL-receptor protein mass in hepatic plasma membranes increased 10-fold (by immunoblotting). In mouse liver, too, LDL-receptor mRNA levels increased 1.5-fold and the LDL-receptor mRNA transcription start sites in rat and mouse were found to be the same, but mouse LDL-receptor protein mass did not change, i.e. LDL-receptors of mice were similar to rat with respect to transcriptional regulation, but differed in their post-transcriptional control mechanisms. In rats, estrogen administration increased apoAI mRNA transcription rates 1.6-fold and also apoAI mRNA levels in total liver homogenates, nuclei and polysomes, (2-fold for each) consistent with transcriptional regulation. However, apoAI synthesis on total RNA increased less than apoAI mRNA, indicating that apoAI translational control mechanisms, at least in part, also regulate hepatic rates of apoAI production. ApoB mRNA transcription rates and levels showed small increases following estrogen administration. Hepatic beta-actin mRNA transcription and levels did not change.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
Affiliation(s)
- R A Srivastava
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO 63110 1093
| | | | | |
Collapse
|
38
|
Mott GE, DeLallo L, Driscoll DM, McMahan CA, Lewis DS. Influence of breast and formula feeding on hepatic concentrations of apolipoprotein and low-density lipoprotein receptor mRNAs. BIOCHIMICA ET BIOPHYSICA ACTA 1993; 1169:59-65. [PMID: 8334151 DOI: 10.1016/0005-2760(93)90082-k] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We tested the hypothesis that breast and formula feeding differentially affect hepatic mRNA concentrations for LDL receptor (LDL-R) and apolipoproteins A-I, B and E in infant baboons during the preweaning period. The mRNA concentrations were measured in liver biopsies obtained prior to weaning at 14 weeks from 43 baboons that were either breast-fed (n = 17) or fed formulas with a high (n = 12) or low (n = 14) polyunsaturated/saturated (P:S) fat ratio. Breast-fed baboons had 99% higher LDL-R mRNA concentrations compared with infants fed formulas, but there were no differences among breast and formula-fed baboons in mRNA concentrations of apolipoproteins A-I, B or E. The fatty acid P:S ratio of the formulas did not affect hepatic LDL-R or apolipoprotein mRNA concentrations. These results suggest that breast-feeding increases LDL-R gene expression even though breast milk is higher in cholesterol and saturated fat compared with formulas.
Collapse
Affiliation(s)
- G E Mott
- Department of Pathology, University of Texas Health Science Center, San Antonio 78284
| | | | | | | | | |
Collapse
|
39
|
Kostner GM. Interaction of Lp(a) and of apo(a) with liver cells. ARTERIOSCLEROSIS AND THROMBOSIS : A JOURNAL OF VASCULAR BIOLOGY 1993; 13:1101-9. [PMID: 8318510 DOI: 10.1161/01.atv.13.7.1101] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Lipoprotein(a) (Lp[a]) is a lipoprotein of high atherogenicity with unknown function. Although it binds in vitro to the low-density lipoprotein (LDL) receptor, it is not clear whether this mechanism also operates in vivo. We studied the interaction of Lp(a) and of apoprotein(a) (apo[a]) with hepatoma cells (HepG2 and Hep3B) with the following results. (1) HepG2 cells exhibited saturable high-affinity binding of LDLs, whereas the majority of Lp(a) binding was of low affinity and nonsaturable. Preincubation of HepG2 cells with LDL markedly reduced cholesterol biosynthesis, but Lp(a) had a much lower effect. (2) When HepG2 cells were preincubated for 48 to 72 hours with Lp(a) or apo(a), 125I-LDL binding was increased by a factor of > 2. During this time, up to approximately 1 microgram of apo(a) per 1 milligram cell protein was found to be cell associated in an undegraded form. Monoclonal antibodies against the LDL receptor did not prevent the increase in LDL binding stimulated by apo(a). (3) Coincubation with LDL caused a significant increase of Lp(a) degradation by HepG2 cells that was probably caused by an increase of Lp(a) uptake in a "hitchhiking"-like process.
Collapse
Affiliation(s)
- G M Kostner
- Institute of Medical Biochemistry, University of Graz, Austria
| |
Collapse
|
40
|
Field FJ, Fujiwara D, Born E, Chappell DA, Mathur SN. Regulation of LDL receptor expression by luminal sterol flux in CaCo-2 cells. ARTERIOSCLEROSIS AND THROMBOSIS : A JOURNAL OF VASCULAR BIOLOGY 1993; 13:729-37. [PMID: 8387332 DOI: 10.1161/01.atv.13.5.729] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The regulation of expression of the intestinal low density lipoprotein (LDL) receptor by luminal (apical) sterol flux was investigated in the human intestinal cell line CaCo-2. Cells were cultured on semipermeable micropore filters, which separated an upper and lower well. To the apical media were added solutions containing either taurocholate micelles alone or micelles containing sterols. Because of an efflux of cholesterol, which occurred from cells incubated with micelles alone, LDL receptor mRNA levels increased threefold. With an influx of micellar sterols, receptor mRNA levels decreased in a dose-dependent manner. Synthesis and degradation of the LDL receptor were addressed by pulse-chase experiments. In cells incubated with micelles containing 25-hydroxycholesterol, the rate of receptor synthesis was significantly decreased, whereas the rate of receptor turnover remained unchanged. As assessed by immunoblots and steady-state labeling of proteins followed by immunoprecipitation of the LDL receptor, cells incubated with micellar 25-hydroxycholesterol contained substantially less receptor protein. These cells also bound and degraded less LDL. In contrast, in cells incubated with micelles alone, the rate of receptor synthesis was increased and cells contained more LDL receptor protein, although this was not reflected in an increased in LDL binding. The results suggest that LDL receptor expression in CaCo-2 cells is regulated by luminal sterol flux and that this regulation occurs at the level of transcription.
Collapse
Affiliation(s)
- F J Field
- Department of Internal Medicine, University of Iowa, Iowa City
| | | | | | | | | |
Collapse
|
41
|
Hoodless PA, Ryan AK, Schrader TJ, Deeley RG. Characterization of liver-enriched proteins binding to a developmentally demethylated site flanking the avian apoVLDLII gene. DNA Cell Biol 1992; 11:755-65. [PMID: 1457044 DOI: 10.1089/dna.1992.11.755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although the avian apoVLDLII gene is normally expressed exclusively in the liver of the laying hen, the gene can be activated by estrogen in birds of either sex beginning between days 7-9 of embryogenesis. Developmentally programmed demethylation of sites in the 5'- and 3'-flanking regions of the gene have been shown to occur during this period of embryogenesis, suggesting that they may reflect changes in protein-DNA interactions that are involved in the acquisition of competence to activate the apoVLDLII gene. We have detected specific protein interactions at one location approximately 2.6 kb upstream from the apoVLDLII gene, that includes an Msp I site whose methylation status changes between days 7 and 9 of embryogenesis. The sequence of this region bears significant similarity to binding sites of members of the bZIP family of liver-enriched or -specific factors such as C/EBP, DBP, and LAP, that are characteristically produced relatively late during liver development. In the studies described here, we demonstrate that proteins binding to the upstream apoVLDLII site do not correspond to previously identified liver-enriched or -specific factors. They also display a pattern of activity during development and in human and avian hepatoma cell lines indicating that their expression is increased in proliferating cells. Southwestern blotting and UV cross-linking studies indicate that two proteins of approximately 60 kD are capable of binding to the site and we describe the purification of these factors from crude nuclear protein extracts obtained from rooster liver.
Collapse
Affiliation(s)
- P A Hoodless
- Cancer Research Laboratories, Queen's University, Kingston, Ontario, Canada
| | | | | | | |
Collapse
|
42
|
Tam SP, Ramharack R. The effect of 25-hydroxycholesterol on the regulation of apolipoprotein E mRNA levels and secretion in the human hepatoma HepG2. Atherosclerosis 1992; 95:137-46. [PMID: 1329783 DOI: 10.1016/0021-9150(92)90017-b] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The human hepatoma cell line, HepG2, was cultured with 25 OH cholesterol, a potent inhibitor of HMG-CoA reductase, in order to examine the effect of the oxysterol on apo E synthesis and secretion. Treatment of cells with oxysterol (2.5 microM) resulted in a greater than 90% inhibition of HMG-CoA reductase activity and a 3-fold reduction in its cognate mRNA level. However, apo E mRNA level and secretion were not affected after 24 h of drug treatment. This drug treatment was associated with a reduction in both cellular free and esterified cholesterol levels by 50% and 40%, respectively. Exposure of HepG2 cells to an ACAT inhibitor, the Sandoz compound (58-035) for 24 h, at a concentration of 5 micrograms/ml, resulted in a 30% increase and 70% decrease in the intracellular levels of free and esterified cholesterol, respectively. Under this regimen of drug treatment, the level of apo E mRNA was increased by approximately 70%, while HMG-CoA reductase mRNA level was decreased by 35%. When the cells were exposed to the combination of the ACAT inhibitor and 25 OH cholesterol, the cellular levels of free and esterified cholesterol were reduced by 30% and 80%, respectively. This combination of drugs had no effect on apo E mRNA; however, the level of HMG-CoA reductase mRNA was decreased by 3.5-fold. Taken together, the data suggested that reduction in the intracellular levels of either free or esterified cholesterol had no effect on apo E mRNA level. By contrast, a small increment in cellular free cholesterol content was associated with a significant induction in apo E mRNA level. Furthermore, 25 OH cholesterol caused a significant redistribution (50%) of apo E from the HDL fraction to the d greater than 1.21 g/ml infranatant. By using high performance liquid chromatography and molecular sieve columns, it was found that the appearance of a lipid-poor apo E particle was not an artifact of ultracentrifugation. This particle contained 85 wt% protein and 15 wt% of free cholesterol and phospholipid. The results suggested that a lipid-poor apo E particle was secreted by the HepG2 cells under certain circumstances.
Collapse
Affiliation(s)
- S P Tam
- Department of Biochemistry, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
43
|
Levy R, Ostlund RE, Schonfeld G, Wong P, Semenkovich CF. Cholesteryl ester storage disease: complex molecular effects of chronic lovastatin therapy. J Lipid Res 1992. [DOI: 10.1016/s0022-2275(20)41416-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|