Effect of Simvastatin on Plasma Homocysteine Levels and Its Modification by MTHFR C677T Polymorphism in Chinese Patients with Primary Hyperlipidemia
Shanqun Jiang,Qianru Chen,Scott A. Venners,Guisheng Zhong,Yi‐Hsiang Hsu,Xing Huang,Xiaobin Wang,Xiping Xu
DOI: https://doi.org/10.1111/cdr.12002
2012-01-01
Cardiovascular Therapeutics
Abstract:Cardiovascular TherapeuticsVolume 31, Issue 4 p. e27-e33 Original Research ArticleFree Access Effect of Simvastatin on Plasma Homocysteine Levels and Its Modification by MTHFR C677T Polymorphism in Chinese Patients with Primary Hyperlipidemia Shanqun Jiang, Corresponding Author Shanqun Jiang School of Life Sciences, Anhui University, Hefei, China Institute of Biomedicine, Anhui Medical University, Hefei, China Correspondence Shanqun Jiang, PhD., School of Life Sciences, Anhui University, 3 Feixi Road, PO Box 41, Hefei 230039, China. Tel.: 86-13956059590 Fax: 86-551-5107354 E-mail: shanqun@gmail.comSearch for more papers by this authorQianru Chen, Qianru Chen School of Life Sciences, Anhui University, Hefei, ChinaSearch for more papers by this authorScott A. Venners, Scott A. Venners Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, CanadaSearch for more papers by this authorGuisheng Zhong, Guisheng Zhong Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USASearch for more papers by this authorYi-Hsiang Hsu, Yi-Hsiang Hsu Institute for Aging Research, HSL and Harvard Medical School, Boston, MA, USA Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA, USASearch for more papers by this authorHouxun Xing, Houxun Xing Institute of Biomedicine, Anhui Medical University, Hefei, ChinaSearch for more papers by this authorXiaobin Wang, Xiaobin Wang The Mary Ann and J. Milburn Smith Child Health Research Program, Children's Memorial Hospital and Children's Memorial Research Center, Chicago, IL, USASearch for more papers by this authorXiping Xu, Xiping Xu Institute of Biomedicine, Anhui Medical University, Hefei, China Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL, USASearch for more papers by this author Shanqun Jiang, Corresponding Author Shanqun Jiang School of Life Sciences, Anhui University, Hefei, China Institute of Biomedicine, Anhui Medical University, Hefei, China Correspondence Shanqun Jiang, PhD., School of Life Sciences, Anhui University, 3 Feixi Road, PO Box 41, Hefei 230039, China. Tel.: 86-13956059590 Fax: 86-551-5107354 E-mail: shanqun@gmail.comSearch for more papers by this authorQianru Chen, Qianru Chen School of Life Sciences, Anhui University, Hefei, ChinaSearch for more papers by this authorScott A. Venners, Scott A. Venners Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, CanadaSearch for more papers by this authorGuisheng Zhong, Guisheng Zhong Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USASearch for more papers by this authorYi-Hsiang Hsu, Yi-Hsiang Hsu Institute for Aging Research, HSL and Harvard Medical School, Boston, MA, USA Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA, USASearch for more papers by this authorHouxun Xing, Houxun Xing Institute of Biomedicine, Anhui Medical University, Hefei, ChinaSearch for more papers by this authorXiaobin Wang, Xiaobin Wang The Mary Ann and J. Milburn Smith Child Health Research Program, Children's Memorial Hospital and Children's Memorial Research Center, Chicago, IL, USASearch for more papers by this authorXiping Xu, Xiping Xu Institute of Biomedicine, Anhui Medical University, Hefei, China Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL, USASearch for more papers by this author First published: 18 July 2013 https://doi.org/10.1111/1755-5922.12002Citations: 16AboutSectionsPDF ToolsRequest permissionExport citationAdd to favoritesTrack citation ShareShare Give accessShare full text accessShare full-text accessPlease review our Terms and Conditions of Use and check box below to share full-text version of article.I have read and accept the Wiley Online Library Terms and Conditions of UseShareable LinkUse the link below to share a full-text version of this article with your friends and colleagues. Learn more.Copy URL Share a linkShare onFacebookTwitterLinked InRedditWechat Summary Objective We investigate the effect of simvastatin on plasma homocysteine (Hcy) levels and whether genetic factor affects the effect of simvastatin. Methods A total of 338 patients with hyperlipidemia were enrolled. Simvastatin was orally administered at a dose of 20 mg/day for 8 weeks. Plasma Hcy levels were measured by high-performance liquid chromatography at baseline and after 8 weeks of treatment. Genotyping of methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism was performed by TaqMan probe technique. Results Serum total Hcy levels were positively correlated with serum creatinine (r = 0.332, P < 0.001). Among total subjects, simvastatin treatment resulted in a significant reduction in serum Hcy levels after 8 weeks (−0.37 ± 2.21 μmol/L, P = 0.003), and this effect was dependent on the initial levels of serum Hcy. The individuals with 677TT genotype had a significantly higher baseline Hcy level and a greater change in Hcy levels. After stratification by body mass index (BMI), we observed a significant increase in Hcy levels among the TT genotype group in adjusted model (beta±SE: 2.64 ± 0.84 μmol/L; P = 0.002) among patients with BMI ≥ 25 (kg/m2). Conclusions Simvastatin can cause a marked decrease in plasma Hcy levels. MTHFR C677T genetic variant contributes to simvastatin's effects among Chinese subjects with primary hyperlipidemia. Introduction Homocysteinemia and hyperlipidemia are two major risk factors associated with the occurrence of atherosclerotic vascular disease 1, 2. Plasma levels of Hcy were related to the cholesterol in homocysteinemic patients as well as in HepG2 cells and experimental animals 3. Methionine administration to pathogenic control rats significantly increased the levels of Hcy, total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-C), very low-density lipoprotein-cholesterol (VLDL-C), and triglyceride (TG) and decreased the levels of high-density lipoprotein-cholesterol (HDL-C) in serum 4, 5. In contrast to these studies, most of the epidemiological data indicated that the elevation of plasma Hcy was not associated with a significant change in plasma TC but negatively associated with HDL-C 6-8. These contradictory findings may come from the unclear interactions between Hcy and lipid metabolism, and thus identifying the mechanisms linking Hcy and lipid metabolism is necessary to resolve the controversy. Statins are widely used to prevent cardiovascular disease through the reduction in TC and especially LDL-C. In addition to blocking 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase involved in cholesterol synthesis, statins possess numerous pleiotropic properties including 9 (1) nitric oxide-mediated improvement in endothelial dysfunction and upregulation of endothelin-1 expression; (2) antioxidant effects; (3) anti-inflammatory properties. Atorvastatin may inhibit Hcy-induced reactive oxygen species accumulation and endothelium cell apoptosis through the NADPH oxidase and/or Nox4/p38MAPK-dependent mechanism 10, 11. Simvastatin attenuated Hcy-induced human umbilical vein endothelial cells apoptosis and caspase-3 activation by upregulating c-IAP1 and c-IAP2 expressions 12. These findings suggest that statins can decrease plasma Hcy level by affecting endothelial function. The effects of simvastatin are not consistent. Studies have shown that higher doses of simvastatin (i.e., 80 mg daily) produced a significant reduction in serum Hcy levels in patients with hypercholesterolemia 13, 14. Recently, treatment with statins in women with polycystic ovary syndrome (PCOS) leads to decreases in serum Hcy levels 15. In contrast to these studies, the neutral effect of therapeutic dosages of either atorvastatin or simvastatin was proposed on Hcy levels in multiple studies 16-20. The different effects of statins on Hcy levels may be due to the small number of patients, differences in the drug doses and treatment periods. Thus, studies with large numbers of participants will be necessary to prove this effect for single statin on Hcy levels. The metabolism of Hcy is under genetic control. MTHFR is an enzyme involved in Hcy metabolism. The 677TT genotype is related to a lower MTHFR activity, a reduced remethylation of Hcy to methionine, and a higher Hcy level 21, 22. For individuals with the TT genotype, the effect of folate on the metabolism of Hcy can lead to a larger decrease in Hcy level than for those without TT genotype 23-25. As statins influence Hcy concentrations, there may be an interaction between C677T in the MTHFR gene and statin therapy in the Hcy-lowering efficacy. In this study, we examined the effect of simvastatin on plasma Hcy levels in a cohort of primary hyperlipidemic patients and whether the genetic factor contribute to the changes in Hcy levels after simvastatin treatment. Patients and Methods Study Population Totally 353 patients were recruited for the study from Anqing region, China. Men and women with dyslipidemia were screened for eligibility by medical history, physical examination, and clinical laboratory evaluation, including the lipid profile. No participant had either symptomatic ischemic heart disease or any other vascular disease. Any lipid-lowering medications had to have been discontinued for at least 4 weeks. All participants gave informed consent, and the study protocol was approved by the ethics committee of Institute of Biomedicine, Anhui Medical University. Participants who met the following criteria of fasting serum lipid level were recruited as primary hyperlipidemia: TC: 5.72–8.32 mmol/L and/or LDL: 3.64–6.50 mmol/L and TG ≥ 1.70 mmol/L. To avoid potentially severe adverse effects, patients with (1) impaired hepatic function (aminotransferase levels > 2 × normal and history of chronic liver disease, such as cirrhosis or alcohol abuse), (2) impaired renal function (serum creatinine levels > 1.8 mg/dL, and/or a history of chronic renal disease, such as glomerulonephritis, chronic pyelonephritis, obstructive renal disease, or proteinuria), (3) diabetes mellitus (fasting blood glucose > 126 mg/dL), (4) raised thyroid-stimulating hormone (TSH) levels (>5.0 μU/L), and (5) any medical conditions that might preclude successful completion were excluded from the study. Simvastatin Treatment After a washout period of 7–10 days, all subjects were treated orally with simvastatin at a daily fixed dosage of 20 mg for eight consecutive weeks. Subjects were required to take their simvastatin between 8:00 am and 10:00 am. During treatment, subjects were required to visit our clinical center every 2 weeks for blood lipid examination and to report any adverse effects. Those receiving drugs possibly affecting the laboratory parameters tested were excluded. Blood samples were taken after a 14-h overnight fast for the determination of serum urea, creatinine, glucose, lipids, Hcy levels, as well as liver and muscle enzymes at baseline and at 8 weeks following treatment initiation. Laboratory Determinations Venous blood samples were drawn and collected in ethylenediaminetetraacetic acid (EDTA) tubes after a 14-h fast at 8.00 am to 10.00 am. The samples were then centrifuged at 2500 g for 10 min to obtain the serum. In our analytical center, automated biochemical analysis was used for the laboratory determinations. Levels of TC, HDL-C, LDL-C, and TG were determined with enzymatic colorimetric assays (Roche Diagnostics, Mannheim, Germany). The intra-assay and inter-assay coefficients of variation were < 5% for all assays were performed. Total plasma Hcy levels were measured by high-performance liquid chromatography (HPLC, Agilent HP 1100 type) coupled with a fluorescence detector (HP 1046 type). The chromatographic column was Hypersil C18 (4.6 mm × 250 mm, 5 μm). We used a 0.07 mol/L HAc buffer as the mobile phase with a flow-rate of 0.8 mL/min. The injection volume was 15 μL. The column temperature was 25°C. The analysis time was 15 min. The excitation and emission wavelengths were set at 390 and 470 nm, respectively. The intra-assay and inter-assay coefficients of variation were < 2%. Genotyping of MTHFR C677T TaqMan allelic discrimination technique was used for detecting MTHFR C677T (rs1801133) genotype in our central laboratory. Universal reaction conditions for genotyping were as follows: 4 ng dried DNA, 0.08 μL 40 × assay locus-specific probe, and 2.0 μL TaqMan universal polymerase chain reaction (PCR) master mix made to a final volume of 4 μL with 1.92 μL sterile water. The PCR cycle conditions were made of initial denaturation at 95°C for 10 min, followed by 50 cycles of 92°C for 15 s and 60°C for 1 min. Each sample was measured twice, and the concordance rate was 99.7%. Statistical Analysis Epidata 3.1 Chinese version was used for database design, data entry, and data check. Mean ± SD were calculated for continuous variables. The t-test was used to compare quantitative variables between two groups. The chi-square test was used for categorical variables. Paired t-test was used to compare Hcy levels before and after treatment. "Post-treatment minus baseline" changes in serum total Hcy levels and serum lipid levels (TC, TG, HDL and LDL), renal function (serum creatinine and urea), or liver function (alanine aminotransferase [ALT], aspartate aminotransferase [AST], alkaline phosphatase [ALP], and gamma-glutamyl transpeptidase [GGT]) were defined as the change in clinical parameters. A multivariate linear regression model was used to evaluate the modification effect of baseline Hcy levels, MTHFR C677T polymorphism on the changes in Hcy levels after simvastatin treatment before and after adjusting for possible confounders, including age, gender, and body mass index (BMI). Relationships between variables were assessed by Pearson's correlation coefficient. Differences were considered to be significant at P < 0.05. All the statistical analysis was carried out using the R packages. Results Baseline Characteristics of Participants A total of 353 patients with primary hyperlipidemia were enrolled and prescribed for simvastatin treatment. Of them, 338 subjects with complete information were used in the final data analysis. There were not significant differences for age, BMI, TG, TC, HDL-C, LDL-C, AST, and ALP between the male and female subjects. Males had higher baseline Hcy levels ([10.25 ± 3.21] μmol/L) than females ([8.70 ± 3.60] μmol/L) (P < 0.001). The percentages of hyperhomocysteinemia (Hcy ≥ 10 μmol/L) were 49.0% and 25.8% in male and female patients, respectively (P < 0.001). Significantly higher ALT, GGT, creatinine, and urea levels were observed in males than in females. MTHFR C677T polymorphism did not deviate from Hardy–Weinberg expectation in females (P > 0.05), but not in males. We re-genotyped all the 338 subjects, the genotype distribution was not changed. The distributions of MTHFR C677T genotypes, cigarette smoking, and alcohol drinking were significantly different between males and females (Table 1). Table 1. Baseline characteristics of the subjects Variables Male (n = 146) Female (n = 192) P Age (year) 50.49 ± 8.56 51.13 ± 6.41 0.45 BMI (kg/m2) 23.57 ± 2.53 23.92 ± 3.10 0.26 Height (cm) 163.23 ± 5.84 153.03 ± 5.59 <0.001 Weight (kg) 62.93 ± 8.47 56.09 ± 8.41 <0.001 TG (mmol/L) 1.78 ± 1.23 1.90 ± 0.97 0.37 TC (mmol/L) 6.39 ± 0.52 6.46 ± 0.58 0.27 HDL-C (mmol/L) 1.97 ± 0.53 1.99 ± 0.44 0.72 LDL-C (mmol/L) 3.56 ± 0.78 3.61 ± 0.59 0.94 ALT (mmol/L) 34.63 ± 18.24 29.89 ± 24.42 0.04 AST (mmol/L) 36.37 ± 10.35 34.39 ± 8.96 0.06 ALP (mmol/L) 89.69 ± 20.50 94.82 ± 35.67 0.10 GGT (mmol/L) 42.07 ± 70.80 25.81 ± 28.51 0.01 Creatinine (mmol/L) 79.70 ± 14.19 60.50 ± 9.33 <0.001 Urea (mmol/L) 354.97 ± 74.25 273.74 ± 56.18 <0.001 Baseline Hcy (μmol/L) 10.25 ± 3.21 8.70 ± 3.60 <0.001 <10 74 (51.0) 144 (74.2) <0.001 ≥10 71 (49.0) 50 (25.8) Non-smoker 52 (35.6) 184 (95.8) <0.001 Smoker 94 (64.4) 8 (4.2) Non-drinker 70 (47.9) 181 (94.3) <0.001 Drinker 76 (52.1) 11 (5.7) MTHFR C677T (%) CC 56 (38.4) 62 (32.3) 0.04 CT 80 (54.8) 100 (52.1) TT 10 (6.8) 30 (15.6) TC, total cholesterol; LDL-C, low-density lipoprotein cholesterol; TG, triglycerides; HDL-C, high-density lipoprotein cholesterol; ALT, alanine transaminase; AST, aspartate transaminase; ALP, alkaline phosphatase; GGT, gamma glutamyl transpeptidase. Bold values denotes significant results. Relationships between Variables Assessed by Pearson's Correlation Coefficient At baseline, serum total Hcy levels positively correlated with serum creatinine (r = 0.332, P < 0.001) and inversely with change in Hcy after simvastatin treatment (r = −0.273, P < 0.001) in the whole population. In addition, change in Hcy treated with simvastatin was positively associated with change in creatinine levels (r = 0.127, P = 0.02) (Figure S1). No correlation was found between "post-treatment minus baseline" changes in serum total Hcy levels and serum lipid levels, or liver function (ALT, AST, ALP, and GGT). Effect of Simvastatin Treatment on TG, TC, HDL-C, and LDL-C and Other Clinical Parameters Simvastatin therapy resulted in a significant reduction in TC, LDL-C, and TG concentrations. HDL-C levels were decreased by simvastatin. ALP, GGT, and urea levels were significantly lowered in the simvastatin-treated group. Simvastatin treatment produced significant increases in serum creatinine and AST levels (Table 2). Table 2. Changes in lipid and other clinical parameter levels after simvastatin treatment Group (mmol/L) Before treatment 8 weeks after treatment Change (μmol/L) Change (%) P TG 1.85 ± 1.09 1.23 ± 0.67 −0.62 ± 0.85 −27 ± 30 <0.001 TC 6.43 ± 0.56 4.46 ± 0.67 −1.97 ± 0.69 −31 ± 10 <0.001 HDL-C 2.00 ± 0.48 1.52 ± 0.43 −0.45 ± 0.27 −23 ± 12 <0.001 LDL-C 3.62 ± 0.67 2.38 ± 0.57 −1.25 ± 0.61 −33 ± 24 <0.001 ALT 31.92 ± 22.10 32.56 ± 17.29 0.64 ± 22.20 13 ± 54 0.6 AST 35.24 ± 9.62 38.50 ± 12.43 3.26 ± 11.46 13 ± 35 <0.001 ALP 92.63 ± 30.20 80.36 ± 24.80 −12.26 ± 17.10 −11 ± 28 <0.001 GGT 32.77 ± 51.62 28.45 ± 59.04 −4.32 ± 21.19 −9 ± 32 <0.001 Creatinine 68.71 ± 15.03 70.73 ± 13.97 2.01 ± 5.48 4 ± 8 <0.001 Urea 308.48 ± 75.96 298.45 ± 73.41 −10.03 ± 43.03 −2 ± 15 <0.001 TC, total cholesterol; LDL-C, low-density lipoprotein cholesterol; TG, triglycerides; HDL-C, high-density lipoprotein cholesterol; ALT, alanine transaminase; AST, aspartate transaminase; ALP, alkaline phosphatase; GGT, gamma-glutamyl transpeptidase.Bold values denotes significant results. Effect of Simvastatin Treatment on Hcy Levels Hcy levels were significantly decreased in patients receiving simvastatin in the total population (−0.37 ± 2.21 μmol/L, P = 0.003) (Figure 1). Stratified by baseline Hcy levels, the patients with baseline Hcy < 10 μmol/L had no significant change in plasma Hcy levels (P = 0.93). By contrast, those with baseline Hcy ≥ 10 μmol/L had a markedly reduction in plasma Hcy levels (−1.04 ± 3.05 μmol/L, P < 0.001) (Figure 1). Multivariate linear regression further identified that compared with patients with baseline Hcy < 10 μmol/L, those with baseline Hcy ≥ 10 μmol/L had a significantly lower change in Hcy levels after simvastatin treatment in either unadjusted (beta ± SE: −1.05 ± 0.24; P < 0.001) or adjusted (beta ± SE: −1.08 ± 0.26; P < 0.001) regression model (Table 3). Figure 1Open in figure viewerPowerPoint Effect of Simvastatin treatment on change in Hcy levels. Change in Hcy levels (Mean ± SED) before and after treatment was compared by paired t-test Significantly different from pretreatment **P < 0.01. In total population, Hcy levels were significantly decreased in patients receiving simvastatin (−0.37 ± 2.21 μmol/L, P = 0.003). The patients with baseline Hcy < 10 μmol/L had no significant change in plasma Hcy levels (P = 0.93). The patients with baseline Hcy ≥ 10 μmol/L had a marked reduction in plasma Hcy levels (−1.04 ± 3.05 μmol/L, P < 0.001). Table 3. Multivariate linear regression of the baseline Hcy level modification effect on the changes of Hcy levels after Simvastatin treatment Group (μmol/L) n Hcy change Unadjusted Adjustedaa Adjusted for age, sex, BMI, smoking, and alcohol consumption.Bold values denotes significant results. β SE P β SE P Baseline Hcy < 10 217 0.01 ± 1.44 Ref Ref Baseline Hcy ≥ 10 121 −1.04 ± 3.05 −1.05 0.24 <0.001 −1.08 0.26 <0.001 a Adjusted for age, sex, BMI, smoking, and alcohol consumption.Bold values denotes significant results. Modification Effect of MTHFR C677T Polymorphism on the Changes in Hcy Levels after Simvastatin Treatment Among total subjects, the baseline Hcy levels for CC, CT, and TT genotypes were (8.72 ± 2.95), (9.27 ± 3.17), and (11.59 ± 5.29) μmol/L, respectively. Subjects with the TT genotype not only had higher baseline Hcy levels (P < 0.001), but also had a greater change in Hcy levels after 8 weeks of simvastatin treatment in either unadjusted (beta±SE: 0.79 ± 0.40; P = 0.05) or adjusted (beta±SE: 0.82 ± 0.41; P = 0.04) models. After stratification by BMI, we observed that those with the TT genotype had a more significant increase in Hcy levels after simvastatin treatment in either unadjusted (beta ± SE: 2.30 ± 0.80; P = 0.007) or adjusted (beta±SE: 2.64 ± 0.84; P = 0.002) models among patients with BMI ≥ 25 (kg/m2). However, among the BMI < 25 (kg/m2) group, no effect of MTHFR C677T polymorphism on change in Hcy levels was found (Table 4). Table 4. Modification effect of MTHFR C677T polymorphism on the changes in Hcy levels after Simvastatin treatment stratified by BMI Genotype n Baseline Hcy (μmol/L) Hcy change Unadjusted Adjustedaa Adjusted for age, sex, BMI, smoking, and alcohol consumption.Bold values denotes significant results. β SE P β SE P Total CC 118 8.72 ± 2.95 −0.52 ± 1.98 Ref Ref CT 180 9.27 ± 3.17 −0.41 ± 1.76 0.10 0.26 0.7 0.11 0.26 0.68 TT 40 11.59 ± 5.29 0.28 ± 3.94 0.79 0.4 0.05 0.82 0.41 0.04 BMI<25 CC 78 8.51 ± 2.51 −0.25 ± 1.72 Ref Ref CT 135 9.19 ± 3.10 −0.48 ± 1.63 −0.22 0.28 0.43 −0.27 0.28 0.34 TT 25 11.56 ± 4.77 −0.27 ± 3.57 −0.01 0.44 0.97 −0.06 0.45 0.89 BMI≥25 CC 40 9.15 ± 3.70 −1.04 ± 2.37 Ref Ref CT 45 9.51 ± 3.38 −0.23 ± 2.09 0.81 0.58 0.17 0.94 0.60 0.12 TT 15 11.64 ± 6.27 1.22 ± 4.49 2.300 0.804 0.007 2.64 0.84 0.002 a Adjusted for age, sex, BMI, smoking, and alcohol consumption.Bold values denotes significant results. Discussion This is the first study to detect the change in Hcy levels after simvastatin treatment in Chinese primary hyperlipidemia. The relatively large number of subjects is a prominent advantage, due to which our findings had more statistical power than many previous studies of smaller sample sizes 15-22. Nonetheless, these findings need to be replicated and expanded to a larger population of primary hyperlipidemic individuals to understand the effect of lipid-lowering treatment on Hcy levels. Hcy can cause endothelial damage and oxidative stress and thus hyperhomocysteinemia may be an independent risk factor for vascular diseases. Apart from their lipid-lowering capacity, statins exert anti-inflammatory and antioxidant effects. The anti-inflammatory and antioxidant capacity of statins is thought to contribute to the Hcy-lowering beneficial effects in patients 26. In this study, we identified the effects of simvastatin treatment on Hcy levels in patients with primary hyperlipidemia. Our results suggested that simvastatin treatment were effective not only in improving the lipid profile (reducing TC, TG, and LDL-C levels), but also in reducing nonlipid biochemical risk factors such as Hcy levels. Animal experimental studies showed that atorvastatin significantly decreased the levels of Hcy, TC, TG, LDL-C, and VLDL-C and increased the serum levels of HDL-C in hyperhomocysteinemic rats 4, 5. Simvastatin can significantly decrease serum Hcy levels by augmenting renal function, decreasing microalbuminuria, improving renal allograft survival, and a fall in blood pressure 27, 28. Our findings supported the view that the simvastatin treatment interfered with creatinine metabolism, because a hypohomocysteinemic effect of simvastatin was positively associated with a decrease in creatinine levels (r = 0.127, P = 0.02). However, the exact pathophysiological mechanisms underlying the association of serum Hcy change with statins remain to be further elucidated. Lipid-lowering agents are known to modify the concentration of plasma Hcy. In a meta-analysis of 15 studies, statins caused small reductions (−3.5%) in Hcy blood concentrations, but the clinical relevance of these small effects is not clear 29. Milionis et al. 16 proposed the neutral effect of therapeutic dosages of either atorvastatin or simvastatin on Hcy levels in patients with primary hyperlipidemia. The finding is in agreement with the small number of studies evaluating the influence of statin therapy on Hcy levels 17-20. A prospective study designed by Luftjohann et al. 13 demonstrated that a higher dose of simvastatin produced a significant reduction in serum Hcy levels in patients with hypercholesterolemia. Vladimirova-Kitova et al. 14 compared the effects of moderate (40 mg) to high (80 mg) simvastatin doses on total Hcy levels in patients with hypercholesterolemia and found that the therapy with 80 mg, but not 40 mg, simvastatin significantly decreased the total Hcy level. More recently, Kaya et al. 15 randomly divided into two groups for treatment with atorvastatin and simvastatin. After 12 weeks of treatment, serum Hcy levels in the atorvastatin group decreased from 14.3 ± 2.9 to 10.6 ± 1.7 μmol/L; in the simvastatin group, the levels decreased from 13.6 ± 2.1 to 11.1 ± 1.9 μmol/L. Their findings demonstrated that treatment with statins in women with PCOS leads to decreases in serum Hcy levels. Similar to several previous studies 13-15, we found a significant decrease in Hcy levels after simvastatin treatment for 8 weeks (P = 0.003). Importantly, stratified by baseline Hcy levels, we found that those with baseline Hcy ≥ 10 μmol/L had a significant decrease in plasma Hcy levels (P < 0.001), whereas those with baseline Hcy < 10 μmol/L had no change in plasma Hcy levels after treatment. The basis for our study to define hyperhomocysteinemia as Hcy ≥10 μmol/L was from the American Heart Association/American Stroke Association (AHA/ASA) guidelines for the prevention of stroke in patients with ischemic stroke (IS) or transient ischemic attack (TIA) in 2006. Moreover, many epidemiological and clinical studies also revealed that the risk of developing cardiovascular disease increased significantly when plasma Hcy level was more than 10 μmol/L. Further, we did a multivariate linear regression and estimated a modification effect of the baseline Hcy level on the changes in Hcy levels after simvastatin treatment and provided further support for the conclusion that subjects with baseline Hcy ≥ 10 μmol/L had significantly lower changes in Hcy levels (adjusted beta±SE: -1.08 ± 0.26, P < 0.001). Our data showed that if the initial serum Hcy concentration is different, simvastatin will have distinct effects. Indeed, in some previous studies 13-15, the baseline Hcy level was relatively higher (13.0–15.5) μmol/L and this would lead to the efficient therapy of simvastatin. Whereas in other studies 16-20, the baseline Hcy level was lower (around 10.5 μmol/L), and simvastatin treatment was consequently less efficient. Our findings provide important insights to the mechanism of simvastatin on serum Hcy. In the study of Giral et al. 18, it was identified that changes in Hcy levels in response to atorvastatin were heterogeneous, particularly in one patient who showed a 75% increase in Hcy level, which meant that this may be explained by genetic variants. A common C677T polymorphism in the MTHFR gene plays a vital role in regulating serum Hcy levels. The MTHFR C677T can interact with statin therapy to influence the change in Hcy levels and further modify the risk of cardiovascular outcomes. Maitland-van et al. 30 found that the MTHFR 677 CC variant appeared to modify the efficacy of pravastatin in reducing risk of cardiovascular events [0.71 (95% CI: 0.58-0.87)]. Recently, a small sample-sized, placebo-controlled study 31 showed that statin therapy significantly decreased the Hcy concentration, but the decreased Hcy levels were not modified by the MTHFR polymorphism. Fan et al. 32 found that the observed changes in Hcy concentrations with antihypertension medications were independent of MTHFR polymorphisms. However, our study found that the MTHFR C677T polymorphism remarkably modified the changes in Hcy levels among total cohort subjects. After stratification by BMI, we found that the 677TT subjects with BMI ≥ 25 kg/m2 had a more significant increase in Hcy levels after simvastatin treatment compared to the CC genotype. The increase in BMI was associated with elevated Hcy levels and reduced vitamin levels among hypertensives 33. A previous study showed that simvastatin treatment may result in reduction of plasma antioxidant vitamins 34. Our results imply that the reduced vitamin status after simvastatin treatment might explain the elevation in Hcy levels, especia
What problem does this paper attempt to address?
-
Interference in mevalonate pathway ameliorates homocysteine-induced endothelium-dysfunction.
Biqi Zhang,Lihong Qiu,Michael Fu,Shenjiang Hu
DOI: https://doi.org/10.1016/j.ejphar.2012.07.014
IF: 5.195
2012-01-01
European Journal of Pharmacology
Abstract:Homocysteine is a risk factor for atherosclerosis and hypertension and induces endothelium-dysfunction. Accumulation of cholesterol and reactive oxygen species plays a key role in the endothelium-dysfunction. This study investigated the hypothesis of an involvement of mevalonate pathway and oxidative pathway in homocysteine-induced endothelial damage. Homocysteine induced impairment of the endothelium-dependent vasorelaxation of rat aortic rings by isometric tension, while it also reduced the nitric oxide level and the nitric oxide synthase activity in human umbilical vein endothelial cells, followed by accumulation of superoxide anion and cholesterol. However, the level of asymmetric dimethylarginine remained unaffected by homocysteine. The adverse effect of homocysteine on endothelial function was found to be partially enhanced either by squalestatin-reducing cholesterol or by superoxide dismutase-reducing superoxide anion. Moreover, this effect of homocysteine could be completely ameliorated by simvastatin, very similar to that of cotreatment of squalestatin and superoxide dismutase. Respectively, mevalonolactone partly or squalene fully attenuated the effect of simvastatin or squalestatin on homocysteine-induced endothelial dysfunction. In conclusion, our results suggested that the mevalonate pathway mediates homocysteine-induced endothelium dysfunction besides the oxidative pathway. Interference in the mevalonate pathway and oxidative pathway provides effective protection of endothelial function.
-
The effect of simvastatin on C reactive protein in patients with coronary heart disease
王文标,郑伟英,钟忆周,梁亚非,付国胜
DOI: https://doi.org/10.3969/j.issn.1006-2785.2004.08.007
2004-01-01
Abstract:Objective To investigate the changes of C reactive protein (CRP) in coronary heart disease (CHD) and the effect of simvastatin on CRP. Methods The plasma CRP levels were evaluated in patients with CHD and age-matched normal control group. Patients with CHD were randomized to simvastatin group and non-simvastatin group, and the CRP levels were measured before and after treatment. Results The plasma CRP levels in patients with CHD were significantly higher than those of normal controls (9.87±2.32)mg/ml vs (4.28±1.23)mg/ml(P0.01) . In simvastatin group the plasma CRP levels were significantly decreased after treatment (7.31±2.87)mg/ml, while those of non-simvastatin group remained higher level (9.22±2.54)mg/ml. The latter was significantly higher than former (t=2.34, P0.05). Conclusion Patients with CHD have significantly higher plasma CRP. Simvastatin can decrease the CRP level in patients with CHD.
-
Associations of Two Common Polymorphisms in MTHFR Gene with Blood Lipids and Therapeutic Efficacy of Simvastatin.
Shanqun Jiang,Yu Liu,Cuiping Lu,Yajie Li,Scott A. Venners
DOI: https://doi.org/10.2174/1381612828666220623102537
IF: 3.31
2022-06-26
Current Pharmaceutical Design
Abstract:Background and purpose: Cardio-cerebrovascular disease is an important public health challenge worldwide, and its complex etiology has not been elucidated fully. The study was to investigate the relationship between two common polymorphisms C677T and A1298C in the methylenetetrahydrofolate reductase (MTHFR) gene, baseline lipids and the lipid-lowering efficacy of simvastatin in a Chinese hyperlipidemic population. Methods: All participants were recruited from Anhui, China. By the extreme sampling method, we selected subjects with low response (n=108) and high response (n=106) based on their adjusted lipid-lowering response to simvastatin administrated for 8 consecutive weeks. Both MTHFR C677T and A1298C loci were genotyped by MALDI-TOF MS platform. Serum triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) levels were measured at baseline and after 8 weeks of oral 20 mg/d tablet of simvastatin. Results: Patients with the 677TT genotype had significantly higher baseline TC, HDL-C, and change in HDL-C (ΔHDL-C) levels after treatment than those with 677CC+CT genotypes (β = 0.207, P = 0.045; β = 0.182, P = 0.026; and β = 0.16, P = 0.002, respectively). Patients with 1298AC+CC genotypes had significantly higher baseline LDL-C and change in LDL-C (ΔLDL-C) levels (β = 0.276, P =0.043; β = 0.359, P = 0.025, respectively) than those with 1298AA genotype. We found statistical interactions between the two SNPs in association with baseline HDL-C (P for interaction = 0.034), TC (P for interaction = 0.069), and TG (P for interaction = 0.034). Baseline TC (P = 0.027) and HDL-C (P = 0.046) and change in HDL-C (P = 0.019) were different among those with the MTHFR A-T haplotype compared with A-C. Conclusions: Our major findings suggest that both MTHFR C677T and A1298C polymorphisms could be important genetic determinants of lipid traits and drug efficacy of simvastatin. This will contribute to a better understanding of strategies for personalized medication in Chinese patients with dyslipidemia. Keywords: MTHFR, polymorphism, haplotype, dyslipidemia, efficacy, simvastatin.
pharmacology & pharmacy
-
Effects of simvastatin on apolipoprotein M in vivo and in vitro
Xiaoying Zhang,Shubing Mao,Guanghua Luo,Jiang Wei,Maria Berggren-Söderlund,Peter Nilsson-Ehle,Ning Xu
DOI: https://doi.org/10.1186/1476-511X-10-112
2011-07-05
Abstract:Objective: To investigate effects of lipid lowering drug, simvastatin, on apolipoprotein M expression in the hyperlipidemic mice and in hepatic cell line, HepG2 cells. Methods: Swiss male mice were randomly divided into the high fat group and control group, and were intragastrically fed with 0.9% saline (control group) or lipid emulsion (high fat group) at the daily dosage of 15 ml/kg body weight, respectively. After 8 weeks feeding, the hyperlipidemic model was successfully induced and these hyperlipidemic mice were then randomly divided into three experimental groups: vehicle control group, high-dose simvastatin-treated group (100 mg/kg body weight), and low-dose simvastatin-treated group (10 mg/kg body weight). Mice were dosed daily for 6 weeks of simvastatin before mice were sacrificed for determining serum lipid profile and apoM protein levels that was determined by using dot blotting analysis. Effects of simvastatin on apoM mRNA expression in the HepG2 cells were determined by real-time RT-PCR. Results: Comparing to high fat model mice without simvastatin treatment, 100 mg/kg simvastatin could significantly increase serum total cholesterol (P < 0.05). Serum apoM levels, in all mice, were significantly lower in the mice at the age of 26 weeks than the mice at 12 weeks old (P < 0.05), which indicated that serum apoM levels were significantly correlated to the mice age. It demonstrated also that treatment of simvastatin did not influence serum apoM levels in these mouse model, although serum apoM levels were increased by about 13% in the 10 mg/kg simvastatin group than in the vehicle control group without simvastatin. In HepG2 cell cultures, simvastatin could significantly decrease apoM mRNA levels with dose- and time-dependent manners. At 10 μM simvastatin treatment, apoM mRNA decreased by 52% compared to the controls. Conclusion: The present study suggested that simvastatin, in vivo, had no effect on apoM levels in the hyperlipidemic mouse model. ApoM serum levels in mice were significantly correlated to the animal's age, whereas in cell cultures simvastatin does inhibit apoM expression in the HepG2 cells. The mechanism behind it is not known yet.
-
The association between the SLCO1B1, apolipoprotein E, and CYP2C9 genes and lipid response to fluvastatin: a meta-analysis.
Qian Xiang,Xiaodan Zhang,Lingyue Ma,Kun Hu,Zhuo Zhang,Guangyan Mu,Qiufen Xie,Shuqing Chen,Yimin Cui
DOI: https://doi.org/10.1097/FPC.0000000000000356
2018-01-01
Pharmacogenetics and Genomics
Abstract:Objective The aim of this study was to determine the impact of the SLCO1B1, apolipoprotein E (ApoE), and CYP2C9 genotypes on the lipid-lowering efficacy of fluvastatin. Methods We performed electronic searches on the PubMed, Embase, and Cochrane Library databases to identify studies published through October 2017. Studies that reported the effect estimates with 95% confidence intervals (CIs) of total cholesterol (TC), triglyceride, low-density lipoprotein (LDL), and high-density lipoprotein were included so that the different genotype categories could be compared. Weighted mean difference (WMD) was used to summarize the effect estimates. Results Six studies, involving a total of 1171 individuals, were included in the final analysis. We noted that the patient carrier SLCO1B1 521TT was associated with greater change in TC (WMD: -2.98; 95% CI: -5.12 to -0.84; P=0.006) and LDL (WMD: -5.58; 95% CI: -10.64 to -0.52; P=0.031) compared with 521TC or CC. Furthermore, the patient carrier ApoE*2/*3 showed more change in high-density lipoprotein compared with ApoE*3/*3 (WMD: 18.76; 95% CI: 8.97-28.55; P<0.001) and ApoE*3/*4 or *4/*4 (WMD: 22.51; 95% CI: 0.98-44.04; P=0.040). Finally, the CYP2C9 genotypes showed no correlation with the effects of fluvastatin on TC, triglyceride, and LDL. Conclusion The findings of this study suggested that the SLCO1B1 and ApoE polymorphisms could influence the lipid-lowering effect of fluvastatin, whereas the CYP2C9 genotypes were not associated with the therapeutic effects of fluvastatin.
-
Short-term Effect of Simvastatin on Serum C-reactive Protein of Hyperlipidemia Patients
曾辉,张福春,郭静萱
DOI: https://doi.org/10.3969/j.issn.1001-1439.2004.03.010
2004-01-01
Journal of Clinical Cardiology
Abstract:Objective:To study the effect of simvastatin on serum C-reactive protein (CRP).Method:Ninety hyperlipidemic patients were randomly divided into two groups: treatment group (treated with Jing Bi Shu Xin, 20 mg QN×4 weeks) and control group (without any lipid lowering medication). Serum levels of total cholesterol, LDL-cholesterol, HDL-cholesterol, triglycerides and high sensitivity CRP (hsCRP) at the beginning and after 1 month of treatment were monitored.Result:After 1 month of treatment the decrease of serum total cholesterol, LDL-cholesterol and hsCRP (P 0.05 to 0.01) were 20.8%, 21.4%and 7.6% respectively; however the increase of HDL-cholesterol was 6.3%. But no significant changes were found in the control group. The change of hsCRP was significantly associated with the change of HDL-cholesterol (r=- 0.22; P 0.01), and was independent of TC and LDL-C.Conclusion:Simvastatin treatment not only lowered the lipid level but also reduced serum CRP. The change of CRP was significantly associated with the change of HDL-C but not with TC or LDL-C.
-
Impact of common genetic variation on response to simvastatin therapy among 18 705 participants in the Heart Protection Study
Jemma C. Hopewell,Sarah Parish,Alison Offer,Emma Link,Robert Clarke,Mark Lathrop,Jane Armitage,Rory Collins,,
DOI: https://doi.org/10.1093/eurheartj/ehs344
IF: 39.3
2012-10-24
European Heart Journal
Abstract:AIMS: Statins reduce LDL cholesterol (LDL-C) and the risk of vascular events, but it remains uncertain whether there is clinically relevant genetic variation in their efficacy. This study of 18 705 individuals aims to identify genetic variants related to the lipid response to simvastatin and assess their impact on vascular risk response.METHODS AND RESULTS: A genome-wide study of the LDL-C and apolipoprotein B (ApoB) response to 40 mg simvastatin daily was performed in 3895 participants in the Heart Protection Study, and the nine strongest associations were tested in 14 810 additional participants. Selected candidate genes were also tested in up to 18 705 individuals. There was 90% power to detect differences of 2.5% in LDL-C response (e.g. 42.5 vs. 40% reduction) in the genome-wide study and of 1% in the candidate gene study. None of the associations from the genome-wide study was replicated, and nor were significant associations found for 26 of 36 candidates tested. Novel lipid response associations with variants in LPA, CELSR2/PSRC1/SORT1, and ABCC2 were found, as well as confirmatory evidence for published associations in LPA, APOE, and SLCO1B1. The largest and most significant effects were with LPA and APOE, but were only 2-3% per allele. Reductions in the risk of major vascular events during 5 years of statin therapy among 18 705 high-risk patients did not differ significantly across genotypes associated with the lipid response.CONCLUSIONS: Common genetic variants do not appear to alter the lipid response to statin therapy by more than a few per cent, and there were similar large reductions in vascular risk with simvastatin irrespective of genotypes associated with the lipid response to simvastatin. Consequently, their value for informing clinical decisions related to maximizing statin efficacy appears to be limited.
cardiac & cardiovascular systems
-
EFFECTS OF CHANGE OF STATINS ON ENDOTHELIAL NITRIC OXIDE PRODUCTION IN VASCULAR ENDOTHELIAL CELLS
Xing Yan,Chen Hong,Hu Dayi
DOI: https://doi.org/10.1136/heartjnl-2012-302920a.208
IF: 5.7
2012-01-01
Heart
Abstract:ObjectivesTo investigate the effects of change of statins on endothelial nitric oxide production in human umbilical vein endothelial cells (HUVECs).MethodsHUVECs were exposed to simvastatin (10−6 mmol/l) for 24 h. After wash, HUVECs were exposed to lovastatin with similar concentration, or continue to use the primary statin for another 24 h, then the NO production was menstruated by Griess reagents. Levels of eNOS mRNA after 24 h of change of statins were examined by RT-PCR.ResultsEffect of change of statins: Compared with the control levels, continue to use simvastatin increased the NO production by 103%±31% p<0.05). Compared with continue use of statin, change of simvastatin by lovastatin the production of NO was reduced by 68%±7% p<0.05), but there was no significant statistical difference with control levels. When change of simvastatin by lovastatin HUVECs eNOS mRNA expression was decreased by 53%±11% p<0.05) compared with continue use of simvastatin.ConclusionsChange of statins can decrease the production of nitric oxide and the expression of eNOSmRNA, which may account for high cardiovascular events rate in clinical study when statins treatment were changed.
-
Effects of long-time simvastatin-treated on bile acids homeostasis and relative gene expressions in hypercholesterolemia mice
Yu-qi HE,Yi-mei DU,Lei LING,Fei-fei MA,Chun-yuan ZHU,Rui-jie MAO,Xu-mei ZHOU,Li YANG,Zheng-tao WANG,Yu-he WANG,Yan-liu LU
DOI: https://doi.org/10.14109/j.cnki.xyylc.2017.09.011
2017-01-01
Abstract:AIM To investigate the role of simvastatin on bile acids regulation in high fat diet induced hypercholesterolemia mice.METHODS Three experimental groups were set up including normal diet group,high fat diet group,and simvastatin (20 mg·kg-1.d-1) group.Mice were feed with high fat diet for 16 weeks to establish the hypercholesterolemia model.Meanwhile,the normal diet group mice were feed with normal diet.Simvastatin were treated after week 16 for 22 weeks.Mice serum was collected for total cholesterol (TC),low density lipoprotein cholesterol (LDL-C),aspartate transaminase (AST) and alanine transaminase (ALT)assays.Mice livers were used for HE staining and mRNA expression analysis of genes involved in bile acids regulation.Hepatic bile acids assay was done by using LC-MS technology.RESULTS Simvastatin reduced the serum TC and LDL-C,showing the expected role on hypercholesterolemia treatment (P < 0.05);however,accompanied by the abnormally induced ALT levels.Simvastatin attenuated repressed gene expression of Cyp 7al induced by high fat diet (P < 0.05).In addition,simvastatin also induced the mRNA level of Cyp7al regulators in livers such as Fxr,Fgf15,Jnk1/2 and Erk1/2 (P < 0.05).In particular,simvastatin also regulated bile acids regulators in small intestines such as Fgf1S.LC-MS analysis showed that most bile acids in liver tissues were significantly changed by simvastatin.CONCLUSION Simvastatin significantly changes expression levels of genes involved in bile acids homeostasis regulation.
-
Simvastatin rescues homocysteine-induced apoptosis of osteocytic MLO-Y4 cells by decreasing the expressions of NADPH oxidase 1 and 2
Ayumu Takeno,Ippei Kanazawa,Ken-Ichiro Tanaka,Masakazu Notsu,Maki Yokomoto-Umakoshi,Toshitsugu Sugimoto
DOI: https://doi.org/10.1507/endocrj.EJ15-0480
2016-04-25
Abstract:Clinical studies have shown that hyperhomocysteinemia is associated with bone fragility. Homocysteine (Hcy) induces apoptosis of osteoblastic cell lineage by increasing oxidative stress, which may contribute to Hcy-induced bone fragility. Statins, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, ameliorate oxidative stress by regulating oxidant and anti-oxidant enzymes. However, the effects of statins on Hcy-induced apoptosis of osteocytes are unknown. This study was thus aimed to investigate whether or not statins prevent Hcy-induced apoptosis of osteocytic MLO-Y4 cells and regulate NADPH oxidase (Nox) expression. TUNEL staining showed that 5 mM Hcy induced apoptosis of MLO-Y4 cells, and that co-incubation of 10(-9) or 10(-8) M simvastatin significantly suppressed the apoptotic effect. Moreover, we confirmed the beneficial effect of simvastatin against Hcy's apoptotic effect by using a DNA fragment ELISA assay. However, TUNEL staining showed no significant effects of pravastatin, a hydrophilic statin, on the Hcy-induced apoptosis. Real-time PCR showed that Hcy increased the mRNA expressions of Nox1 and Nox2, whereas simvastatin inhibited the stimulation of Nox1 and Nox2 expressions by Hcy. In contrast, neither Hcy nor simvastatin had any effect on Nox4 expression. These findings indicate that simvastatin prevents the detrimental effects of Hcy on the apoptosis of osteocytes by regulating the expressions of Nox1 and Nox2, suggesting that statins may be beneficial for preventing Hcy-induced osteocyte apoptosis and the resulting bone fragility.
-
Effect of Methylfolate, Pyridoxal-5′-Phosphate, and Methylcobalamin (SolowaysTM) Supplementation on Homocysteine and Low-Density Lipoprotein Cholesterol Levels in Patients with Methylenetetrahydrofolate Reductase, Methionine Synthase, and Methionine Synthase Reductase Polymorphisms: A Randomized Controlled Trial
Evgeny Pokushalov,Andrey Ponomarenko,Sevda Bayramova,Claire Garcia,Inessa Pak,Evgenya Shrainer,Marina Ermolaeva,Dmitry Kudlay,Michael Johnson,Richard Miller
DOI: https://doi.org/10.3390/nu16111550
IF: 5.9
2024-05-22
Nutrients
Abstract:Exploring the link between genetic polymorphisms in folate metabolism genes (MTHFR, MTR, and MTRR) and cardiovascular disease (CVD), this study evaluates the effect of B vitamin supplements (methylfolate, pyridoxal-5′-phosphate, and methylcobalamin) on homocysteine and lipid levels, potentially guiding personalized CVD risk management. In a randomized, double-blind, placebo-controlled trial, 54 patients aged 40–75 with elevated homocysteine and moderate LDL-C levels were divided based on MTHFR, MTR, and MTRR genetic polymorphisms. Over six months, they received either a combination of methylfolate, P5P, and methylcobalamin, or a placebo. At the 6 months follow-up, the treatment group demonstrated a significant reduction in homocysteine levels by 30.0% (95% CI: −39.7% to −20.3%) and LDL-C by 7.5% (95% CI: −10.3% to −4.7%), compared to the placebo (p < 0.01 for all). In the subgroup analysis, Homozygous Minor Allele Carriers showed a more significant reduction in homocysteine levels (48.3%, 95% CI: −62.3% to −34.3%, p < 0.01) compared to mixed allele carriers (18.6%, 95% CI: −25.6% to −11.6%, p < 0.01), with a notable intergroup difference (29.7%, 95% CI: −50.7% to −8.7%, p < 0.01). LDL-C levels decreased by 11.8% in homozygous carriers (95% CI: −15.8% to −7.8%, p < 0.01) and 4.8% in mixed allele carriers (95% CI: −6.8% to −2.8%, p < 0.01), with a significant between-group difference (7.0%, 95% CI: −13.0% to −1.0%, p < 0.01). Methylfolate, P5P, and methylcobalamin supplementation tailored to genetic profiles effectively reduced homocysteine and LDL-C levels in patients with specific MTHFR, MTR, and MTRR polymorphisms, particularly with homozygous minor allele polymorphisms.
nutrition & dietetics
-
The effect of APOE, CETP, and PCSK9 polymorphisms on simvastatin response in Thai hypercholesterolemic patients
Smith Wanmasae,Wisant Sirintronsopon,Sureerut Porntadavity,Nutjaree Jeenduang
DOI: https://doi.org/10.1111/1755-5922.12302
Abstract:Aim: To investigate the effect of apolipoprotein E (APOE), cholesteryl ester transfer protein (CETP) and proprotein convertase subtilisin kexin type 9 (PCSK9) polymorphisms on the lipid-lowering response to simvastatin therapy in Thai hypercholesterolemic patients. Method: Two hundred and twenty-five hypercholesterolemic patients in southern Thailand were enrolled and treated with simvastatin 20 or 40 mg per day for 3 months. Serum lipids were measured before and after the therapy. APOE, CETP TaqIB, and PCSK9 (R46L, I474V, and E670G) polymorphisms were analyzed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Results: After 3 months of simvastatin therapy, subjects with APOE2 (Total cholesterol [TC]: -30.89% vs-13.56%, P < .05, LDL-C: -45.00% vs -17.73%, P < .05) and APOE3 carriers (TC: -26.22% vs -13.56%, P < .05, LDL-C: -37.14% vs -17.73%, P < .05) had greater TC and LDL-C reduction compared to APOE4 carriers, whereas CETP TaqIB B2B2 genotype showed lower TC (-16.37% vs -24.92%, P = .016) and LDL-C (-22.54% vs -35.19%, P = .028) reduction compared to CETP TaqIB B1 carriers. In addition, PCSK9 474IV carriers showed greater LDL-C (-50.57% vs -32.99%) reduction compared to PCSK9 474II carriers. Combined effect analyses showed that individuals carrying more risk alleles tended to have lower TC and LDL-C (P for trend = .000 and .000, respectively) reduction in response to simvastatin therapy. Conclusion: APOE4 carriers and the CETP TaqIB B2B2 genotype were associated with a decreased response, but PCSK9 474IV carriers tended to be associated with an increased response to simvastatin therapy in Thai hypercholesterolemic patients.
-
Homocysteine and Stroke Risk
Min Zhao,Xiaobin Wang,Mingli He,Xianhui Qin,Genfu Tang,Yong Huo,Jianping Li,Jia Fu,Xiao Huang,Xiaoshu Cheng,Binyan Wang,Fan,Ningling Sun,Yefeng Cai
DOI: https://doi.org/10.1161/strokeaha.116.015324
IF: 10.17
2017-01-01
Stroke
Abstract:Background and Purpose— Elevated blood homocysteine concentration increases the risk of stroke, especially among hypertensive individuals. Homocysteine is largely affected by the methylenetetrahydrofolate reductase C677T polymorphism and folate status. Among hypertensive patients, we aimed to test the hypothesis that the association between homocysteine and stroke can be modified by the methylenetetrahydrofolate reductase C677T polymorphism and folic acid intervention. Methods— We analyzed the data of 20 424 hypertensive adults enrolled in the China Stroke Primary Prevention Trial. The participants, first stratified by methylenetetrahydrofolate reductase genotype, were randomly assigned to receive double-blind treatments of 10-mg enalapril and 0.8-mg folic acid or 10-mg enalapril only. The participants were followed up for a median of 4.5 years. Results— In the control group, baseline log-transformed homocysteine was associated with an increased risk of first stroke among participants with the CC/CT genotype (hazard ratio, 3.1; 1.1–9.2), but not among participants with the TT genotype (hazard ratio, 0.7; 0.2–2.1), indicating a significant gene–homocysteine interaction ( P =0.008). In the folic acid intervention group, homocysteine showed no significant effect on stroke regardless of genotype. Consistently, folic acid intervention significantly reduced stroke risk in participants with CC/CT genotypes and high homocysteine levels (tertile 3; hazard ratio, 0.73; 0.55–0.97). Conclusions— In Chinese hypertensive patients, the effect of homocysteine on the first stroke was significantly modified by the methylenetetrahydrofolate reductase C677T genotype and folic acid supplementation. Such information may help to more precisely predict stroke risk and develop folic acid interventions tailored to individual genetic background and nutritional status. Clinical Trial Registration— URL: http://www.clinicaltrials.gov . Unique identifier: NCT00794885.
-
Homocysteine and All-Cause Mortality in Hypertensive Adults Without Pre-Existing Cardiovascular Conditions: Effect Modification by MTHFR C677T Polymorphism.
Benjamin Xu,Xiangyi Kong,Richard Xu,Yun Song,Lishun Liu,Ziyi Zhou,Rui Gu,Xiuli Shi,Min Zhao,Xiao Huang,Mingli He,Jia Fu,Yefeng Cai,Ping Li,Xiaoshu Cheng,Changyan Wu,Fang Chen,Yan Zhang,Genfu Tang,Xianhui Qin,Binyan Wang,Hao Xue,Yundai Chen,Ye Tian,Ningling Sun,Yimin Cui,Fan,Jianping Li,Yong Huo
DOI: https://doi.org/10.1097/md.0000000000005862
IF: 1.6
2017-01-01
Medicine
Abstract:Background: Previous studies support an association between elevated total homocysteine (tHcy) levels and increased all-cause mortality. However, few prospective studies have examined this association in hypertensive patients, and/or tested any effect modification by the methylene tetrahydrofolate reductase (MTHFR) C677T genotype.Methods: This was a post hoc analysis of the China Stroke Primary Prevention Trial. SerumtHcy and folate were measured at baseline. Individual MTHFR C677T genotype (CC, CT, and TT) was determined. Evidence for death included death certificates or home visits. Cumulative hazards of all-causemortality by tHcy quartiles were estimated using the Kaplan-Meier method, and group differences were compared by log-rank tests. Hazard ratios (HRs) and 95% confidence intervals were estimated by Cox proportional-hazard regression models, adjusting for age, sex, baseline folate, vitamin B12, blood pressure, body mass index, smoking and alcohol drinking status, study center, total cholesterol, triglycerides, high-density lipoprotein cholesterol, fasting glucose, creatinine, and treatment group. Potential effect modification by the MTHFR genotype on the relationship between tHcy and all-cause mortality was tested.Results: The analyses included 20,424 hypertensive patients (41% males) without a history of myocardial infarction or stroke. Baseline mean age (SD) was 60 +/- 7.5 years and mean (SD) serum tHcy was 14.5 +/- 8.4 mu mol/L. After a mean follow-up period of 4.5 years, there were 612 (3%) all-cause deaths. Kaplan-Meier survival curves revealed a graded relationship between tHcy quartiles and all-cause mortality. The HRs, using the lowest quartile as the reference, were 1.2, 1.2, and 1.5 in Q2, Q3, and Q4, respectively. A linear trend test, using natural log-transformed tHcy, resulted in an HR of 1.5 (95% confidence interval 1.2-1.9, P<.001) after adjustment for lifestyle and health-related variables. Whereas the MTHFR genotype alone had little effect on mortality, it significantly modified the tHcy-mortality association, which was much stronger in the CC/CT genotype than in the TT genotype (P for interaction < 0.05).Conclusions: Among Chinese hypertensive patients without cardiovascular comorbidities, elevated tHcy was a significant risk marker for death from all causes, and the association was subject to effect modification by MTHFR genotypes. If confirmed that tHcy and MTHFR genotypes may serve as useful biomarkers for mortality risk assessment and targeted intervention.
-
Plasma Homocysteine and Gene Polymorphisms Associated with the Risk of Hyperlipidemia in Northern Chinese Subjects
Lei Huang,Xiao-Ming Song,Wen-Li Zhu,Yong Li
DOI: https://doi.org/10.1016/s0895-3988(09)60011-8
2008-01-01
Abstract:Objective To examine the relationship between occurrence of hyperlipidemia. plasma homocysteine and polymorphisms of methyleneletra hydrotblate reductase (MTHFR) gene and methionine synthase (MS) gene. Methods A total of 192 hyperlipidemia patients were selected and divided into hypercholesterolemia group, hypertriglyceridemia group. and combined hyperlipidemia group. Another 208 normal individuals were selected as control. Total plasma homocysteine (tHcy) concentration was measured by high-performance liquid chromatography (HPLC). Lipid profiles were measured for all Subjects. The polymorphisms of MTHFR gene C677T and MS gene A2756G were analyzed by PCR-RFLP. Results The tHcy concentration in the combined hyperlipidemia patients was significantly higher than that in the control ( 15.95 mu mol/L vs 13.43 mu mol/L, P<0.05). The prevalence of hyperhomocysteinemia (HHcy) in the combined hyperlipidemia group was significantly higher than that in the control (42.2% vs 23.0%, P=0.015), with the odds ratio (OR) of 3.339 (95%Cl: 1.260-8.849). The hyperlipidemia patients with HHcy had a higher concentration of total cholesterol (TC) than that in the normal tHcy patients (5.67 +/- 0.95 mmol/L vs 5.47 +/- 0.92 mmol/L, P=0.034). There was no significant difference in genotype or allele frequencies of MTHFR C677T between the hyperlipidemic and control groups. The hyperlipidemia patients with MTHFR CT/TT genotype had a higher concentration of triglyceride (TG) than those with CC genotype (2.24 +/- 1.75 mmol/L vs 1.87 +/- 0.95 mmol/L. P<0.05). Individuals with CT/TT genotype had a higher concentration of tHcy than those with 677CC genotype both in the hyperlipidemia group (12.61 +/- 1.24 mu mol/L vs 11.20 +/- 1.37 mu mol/L. P<0.05) and in the control group (14.04 +/- 1.48 mu mol/L vs 12.61 +/- 1.24 mu mol/L, P<0.05). The percentage of MS 2756 GG/AG genotype in the combined hyperlipidemia group was significantly higher than that in the control (26.7% vs 13.0%, P=0.012). with the OR of 3.121 (95%Cl: 1.288-7.651). The hyperlipidemia patients with MS 2756AG/GG genotype had a higher concentration of TC (5.87 +/- 0.89 mmol/L vs 5.46 +/- 0.93 mmol/L. P<0.05) and LDL-C (3.29 +/- 0.81mmol/L vs 2.94 +/- 0.85 mmol/L, P<0.05) than those with AA genotype. However. individuals with 2756AG/GG genotype showed no significant difference in tHcy among those with AA genotype. Conclusion HHcy and MS A2756G mutation may be the risk factors for combined hyperlipidemia. Further Study is needed to confirm the role of HHcy and MS A2756G Mutation in the development of hyperlipidemia.
-
Comparative Effectiveness of Anti-Hyperlipidemic Drugs Monotherapy in Primary Prevention of Cardiovascular Disease
Xuechun Li,Dennis Steenhuis,Maarten Bijlsma,Stijn de Vos,Sumaira Mubarik,Jens Bos,Catharina Schuiling-Veninga,Eelko Hak
DOI: https://doi.org/10.2147/ijgm.s479120
IF: 2.145
2024-10-27
International Journal of General Medicine
Abstract:Xuechun Li, 1 Dennis Steenhuis, 1 Maarten J Bijlsma, 1, 2 Stijn de Vos, 1 Sumaira Mubarik, 1 Jens HJ Bos, 1 Catharina CM Schuiling-Veninga, 1 Eelko Hak 1 1 Pharmacotherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands; 2 Laboratory of Population Health, Max Planck Institute for Demographic Research, Rostock, Germany Correspondence: Xuechun Li, PhD Research Fellow, Pharmacotherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, the Netherlands, Tel +31 649019602, Email Purpose: Anti-hyperlipidemic drug treatments are effective in reducing the risk of cardiovascular disease. In a long-term retrospective inception cohort study, we aimed to assess the real-world comparative effectiveness of anti-hyperlipidemic monotherapies for primary prevention of cardiovascular events. Patients and Methods: Patients aged 18 years and older, who initiated primary prevention with anti-hyperlipidemic monotherapy, were selected from the University of Groningen IADB.nl dispensing database. In intention-to-treat (ITT) analysis we included all patients, whereas in per-protocol (PP) analysis we included both all patients independent of adherence (PPIA) and adherent patients (PPA). Study outcome was the time to first prescription of acute cardiac drug therapy measured by valid drug proxies to identify a first major cardiovascular event. We applied inverse probability of treatment-weighted (IPTW) analysis using Cox regression and time-varying Cox regression with simvastatin as the reference category to estimate the average treatment effect hazard ratios (HR) and their corresponding 95% confidence intervals (CI). Results: Atorvastatin users had significantly higher hazards compared to simvastatin users (HR range: 1.27 to 1.47, 95% CI: 1.15 to 1.69). Similarly, Pravastatin users also exhibited increased hazards compared to simvastatin users (HR range: 1.41 to 1.56, 95% CI: 1.14 to 2.04). Similar patterns were observed in patients with diabetes, rheumatoid arthritis, and asthma/COPD. No differences were found in the hazards of rosuvastatin, fluvastatin, fibrates, and simvastatin. Conclusion: Atorvastatin and pravastatin users had higher long-term rates of cardiovascular events compared to simvastatin monotherapy in primary prevention, the difference may be attributed to the confounding by severity, but also possibly due to differences in drug mechanisms or patient response. These findings could influence current guideline recommendations, suggesting a potential preference for simvastatin in primary prevention, underscoring the need for further research to explore long-term impacts and underlying mechanisms, especially in diverse populations. Keywords: acute cardiac drug therapy, time-varying confounding, inverse probability treatment weighting, Cox regression Graphical Cardiovascular diseases (CVD) are the leading cause of mortality worldwide. 1 According to Statistics Netherlands data, 2 from 1950 to 2021, the mortality caused by CVD among all causes showed a trend of initially rising and then slowly decreasing from 2000. However, CVD still account for a substantial proportion of all causes of death in the Netherlands. Hyperlipidemia is a prevalent risk factor for cardiovascular disease. 3 Anti-hyperlipidemic drug treatments such as statins are effective in regulating plasma Low-Density Lipoprotein (LDL) concentrations, thus reducing the risk of morbidity and mortality associated with cardiovascular disease. 3,4 It is also important to consider the issue of persistence with statin therapy, as studies have indicated poor adherence to this treatment. 5 In clinical practice, statins and fibrates are frequently prescribed as anti-hyperlipidemic drugs, no evidence of a difference between statins and fibrates in reducing cardiovascular events, though statins with fewer adverse effects compared to fibrates. 6 Based on cardiovascular risk management guidelines in the Netherlands, 7 atorvastatin, rosuvastatin, or simvastatin are recommended for patients with a desired LDL cholesterol (LDL-C) reduction of less than 40%, while only atorvastatin or rosuvastatin are advised to achieve reductions greater than 40%. In comparative effectiveness cohort studies, atorvastatin demonstrated superior performance compared to simvastatin in preventing cardiovascular events. 8,9 However, it is important to acknowledge that the observed differences in outcomes between the two statins may not solely be due to the inherent characte -Abstract Truncated-
medicine, general & internal
-
Simvastatin Suppressed HMGB1-RAGE Axis and Atherosclerosis Via Mevalonate Pathway
Liu Ming,Yu Ying,Cao Quan,Jia Jianguo,Zhang Lei,Zou Yunzeng,Ge Junbo
DOI: https://doi.org/10.1136/heartjnl-2011-300867.134
IF: 5.7
2011-01-01
Heart
Abstract:Objective Recent studies suggested that high mobility group box 1 (HMGB1) and receptor for advanced glycation end products (RAGE) contribute to atherosclerosis, and statin may inhibit tissue RAGE or increase serum sRAGE. However, it remains unclear whether statin suppress HMGB1-RAGE axis in atherosclerosis models. Thus, in this study, we tested the hypothesis that simvastatin suppresses HMGB1-RAGE axis and atherosclerosis in apoE deficient mice. Methods Male apoE deficient mice, age five weeks, were offered western diet. At the age of eight weeks, mice were treated with once-daily simvastatin (50 mg/kg/day), simvastatin (50 mg/kg/day)+mevalonic acid (30 mg/kg/day) (SM) or vehicle; all mice were killed at age 11 weeks. Results Compared with apoE deficient mice treated with vehicle or SM, simvastatin-treated mice displayed decreased atherosclerosis lesion area in a lipid-independent manner. In parallel, it was observed that decreased expression of vascular HMGB1, RAGE, VCAM-1, MCP-1 and TF in apoE deficient mice treated with simvastatin versus vehicle or SM. Furthermore, increased sRAGE in serum were observed in simvastatin-treated apoE deficient mice, and the level of sRAGE was negatively correlated with the serum HMGB1 level and atherosclerosis lesion area. More interestingly, consistent with the premise that addition of HMGB1 to HUVECs resulted in increased expression of HMGB1, RAGE and VCAM-1, and simvastatin reverted the effects of HMGB1. The roles of simvastatin were similar to RAGE blockade by anti-RAGE antibody in vitro. Conclusion Simvastatin suppressed HMGB1-RAGE axis and atherosclerosis via mevalonate pathway.
-
Simvastatin Pharmacokinetics in Healthy Chinese Subjects and Its Relations with CYP2C9, CYP3A5, ABCB1, ABCG2 and SLCO1B1 Polymorphisms.
Quan Zhou,Zou-rong Ruan,Bo Jiang,Hong Yuan,Su Zeng
DOI: https://doi.org/10.1691/ph.2013.2676
2013-01-01
Abstract:A pharmacokinetic study of simvastatin (single oral dose, 40 mg) was conducted in 17 healthy Chinese volunteers. Plasma concentrations of simvastatin were determined by an LC-ESI-MS-MS method. The pharmacokinetic parameters of simvastatin were derived with a non-compartmental method. The polymorphisms of CYP2C9, CYP3A5, ABCB1 (encoding P-gp), ABCG2 (encoding BCRP) and SLCO1B1 (encoding OATP1B1) were determined by TaqMan genotyping assay and the impacts of these SNPs on the pharmacokinetics of simvastatin were analyzed. Major pharmacokinetic parameters were as follows: Tmax 1.44 +/- 0.39 h, Cmax 9.83 +/- 2.41 microg x L(-1), t1/2 4.85 +/- 1.23 h and AUC(0-infinity) 40.32 +/- 6.82 microg x h x L(-1). Effect of ABCB1 G2677T/A SNP on dispostion of simvastatin was observed. Significant differences in t1/2, but not Cmax and AUC(0-infinity), were observed between G/A, G/T or G/G carriers and non-G carriers (5.65 +/- 0.50 h vs 4.41 +/- 1.31 h, P < 0.05). There were no significant effects on simvastatin pharmacokinetics by SNPs such as CYP2C9*3 (1075A > C), CYP3A5*3 g.6986A > G, ABCB1 C3435T, ABCG2 c.34G > A, ABCG2 c.421C>A, SLCO1B1 c.388 A > G, SLCO1B1 c.521 T > C, SLCO1B1 g.11187 G > A, SLCO1B1 c.571 T > C and SLCO1B1 c.597 C > T. We conclude here that there is a small inter-subject variation in simvastatin pharmacokinetics in healthy Chinese volunteers. P-gp, OATP1 B1 and BCRP seem unlikely to play an important role in the pharmacokinetics of simvastatin. The gene-dose effects of ABCG2 c.421 C > A and CYP3A5*3 g.6986A > G on simvastatin pharmacokinetics are not strong enough in Chinese subjects.
-
Effectiveness and Safety of Simvastatin 40 mg Daily Use in Treatment of Coronary Heart Disease
Bang-qing Li,Pi-neng LU,Da-yi HU
DOI: https://doi.org/10.3969/j.issn.1672-2531.2005.06.010
2005-01-01
Abstract:Objective To evaluate the effectiveness and safety of simvastatin 40 mg d aily use in treatment of coronary heart disease. Methods The study was designed as before-after study in the same p atients. One hundred and sixty seven patients with coronary heart disease were p rescribed simvastatin 40 mg daily for 3 and 6 months. Total cholestero(TC), lo w-density lipoproteins cholesterol (LDL-C), high-density lipoprotein choleste rol (HDL-C), triglycerldes (TG), ALT and creatine kinase (CK) in serum before therapy and at the end of 3 months and 6 months treatment were dectected. Cont inuous data were analyzed by standard difference of blocked randomization and de scribed by mean±SD. Dunnet-t test was used for multiple compa rison of trial and control groups. Statistical difference was set up at P<0.05. Success rate was assessed by chi square test at the end of 3 and 6 months treatment. Results Simvastatin 40 mg/d significantly decreased the level of TC(P< 0.0 005 ), LDL-C(P< 0.0 005 ), TG (P<0.05), and could elevate HDL-C (P<0.05). There were 39.5% of pati ents whose LDL-C reduced below 70 mg/dl. One patient whose CK raised 5.6 times of upper line of normal range and 4 patients whose ALT raised more than 2 times of upper line of normal range withdrew. The reliability of simvastatin 40 mg/d was relatively good. Conclusions Simvastatin 4 0 mg/d could significantly improve the lipid profile, and is relatively reliable in treatment of coronary heart disease.
-
The anxiolytic effects of atorvastatin and simvastatin on dietary-induced increase in homocysteine levels in rats
Natasa Mijailovic,Dragica Selakovic,Jovana Joksimovic,Vladimir Mihailovic,Jelena Katanic,Vladimir Jakovljevic,Tamara Nikolic,Sergey Bolevich,Vladimir Zivkovic,Milica Pantic,Gvozden Rosic
DOI: https://doi.org/10.1007/s11010-018-3425-6
IF: 3.842
2018-08-17
Molecular and Cellular Biochemistry
Abstract:The aim of this study was to evaluate the effects of atorvastatin and simvastatin on behavioral manifestations that followed hyperhomocysteinemia induced by special dietary protocols enriched in methionine and deficient in B vitamins (B6, B9, B12) by means of alterations in anxiety levels in rats. Simultaneously, we investigated the alterations of oxidative stress markers in rat hippocampus induced by applied dietary protocols. Furthermore, considering the well-known antioxidant properties of statins, we attempted to assess their impact on major markers of oxidative stress and their possible beneficial role on anxiety-like behavior effect in rats. The 4-week-old male Wistar albino rats were divided (eight per group) according to basic dietary protocols: standard chow, methionine-enriched, and methionine-enriched vitamins B (B6, B9, B12) deficient. Each dietary protocol (30 days) included groups with atorvastatin (3 mg/kg/day i.p.) and simvastatin (5 mg/kg/day i.p.). The behavioral testing was performed in the open field and elevated plus maze tests. Parameters of oxidative stress (index of lipid peroxidation, superoxide dismutase, catalase activity, glutathione) were determined in hippocampal tissue samples following decapitation after anesthesia. Methionine-load dietary protocols induced increased oxidative stress in rat hippocampus, which was accompanied by anxiogenic behavioral manifestations. The methionine-enriched diet with restricted vitamins B intake induced more pronounced anxiogenic effect, as well as increased oxidative stress compared to the methionine-load diet with normal vitamins B content. Simultaneous administration of statins showed beneficial effects by means of both decreased parameters of oxidative stress and attenuation of anxiety. The results obtained with simvastatin were more convincible compared to atorvastatin.
cell biology