Volume 16, Issue 3 (May-Jun 2022)                   mljgoums 2022, 16(3): 7-13 | Back to browse issues page

XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

Eslami Z, Mohammadnajad Panah kandi Y, Norouzi A, Eghbal Moghanlou A, Sheikh arabi M, Kazeminejad V, et al . Changes in Blood Lipids and Enzymatic Reactions in Response to Atorvastatin Administration Following a High-Fat Diet in a NAFLD Rat Model. mljgoums 2022; 16 (3) :7-13
URL: http://mlj.goums.ac.ir/article-1-1404-en.html
1- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Golestan province, Iran
2- Department of Sport PhysiologyIslamic Azad University, Islamshahr Branch, Islamshahr, Iran
3- Golestan Research Center of Gastroenterology and Hepatology, GolestanUniversity of Medical Sciences, Gorgan, Iran
4- farhangian University, Ardebil, Iran
5- Golestan University of Medical Sciences and Health Services, Gorgan, Iran
6- Department of phatology, Golestan University of Medical Sciences and Health Services, Gorgan, Iran
7- Laboratory Science research center, faculty of Paramedicine of Golestan University of medical sciences, Gorgan, Iran
8- Department of Sport Physiotraphy, University of Lahore, Panjab, Pakistan
9- Laboratory Science research center, faculty of Paramedicine of Golestan University of medical sciences, Gorgan, Iran , hr.joshaghani99@gmail.com
Full-Text [PDF 741 kb]   (375 Downloads)     |   Abstract (HTML)  (2858 Views)
Full-Text:   (1065 Views)
INTRODUCTION
The liver is one of the most important organs, which is involved in many functions; however, its role in fat absorbance and defense against microbes and toxins absorbed through food should not be overlooked (1). Non-alcoholic fatty liver disease (NAFLD) is histologically similar to alcoholic hepatitis but not related to alcohol consumption (2). It shows a spectrum of gradual fat accumulation in the liver, which could be life-threatening (3). The disease refers to the deposition of fat, especially triglycerides (TG) in the liver (4), which occurs when the metabolism of fatty acids shifts towards lipogenesis rather than lipolysis. This clinical condition includes diverse liver damages ranging from simple hepatitis to hepatitis steatosis, advanced fibrosis, cirrhosis, and liver cancer (5). Also, the most important hypothesis in the etiology of NAFLD is called the two-stage theory, which includes insulin resistance and oxidative damage (6). In obese patients, NAFLD is mainly associated with other components of metabolic syndrome, such as hypertension, hyperglycemia, and hyperlipidemia (7).
Aspartate aminotransferase (AST), alkaline phosphatase (ALP), gamma glutamyl transferase (GGT), and especially alanine aminotransferase (ALT) are the most important indicators of liver health (8). Serum levels of liver enzymes including ALP, AST, GGT, and ALT are commonly elevated in patients with NAFLD (9). These enzymes are also increased in conditions such as diabetes and alcoholism (10). Factors such as obesity, hyperglycemia, and inactivity increase the risk of developing NAFLD. Of course, some medications can also cause fat build up in the liver. Rapid weight loss in people who are already obese can also lead to this complication. According to reports, NAFLD is one of the leading causes of liver disease-associated morality, and due to the potential progression of this disease to cirrhosis and liver failure, a lot attention has been given to detect this disease and its associated factors (11).
Carbon tetrachloride (CCl4) has been used for many years to induce liver damage and fibrosis in animal models. When combined with a Western diet rich in fat and fructose, it can induce oxidative stress, inflammation, and apoptosis. As fibrosis progresses in hepatocytes, fatty liver leads to steatosis; CCl4 induces NAFLD through this mechanism (12-14). Due to variable doses and durations of CCl4 injection for the induction of NAFLD, besides the possibility of hepatotoxicity rather than NAFLD induction, a diet-based method is needed to develop an optimal model.
Several medications, including atorvastatin, are prescribed for treatment of NAFLD. Atorvastatin is a subset of statin drugs that if administered for long-term and in high-dose, can cause hepatic complications and increased cell apoptosis. It can also reduce blood cholesterol, TG, and low-density lipoprotein-cholesterol (LDL-C) and increase high-density lipoprotein-cholesterol (HDL-C) levels. Atorvastatin is prescribed in different doses to cardiovascular patients and patients with high cholesterol. Atorvastatin have antioxidant, anti-apoptotic, and anti-inflammatory properties (15,16). The anti-inflammatory properties of this drug are mediated via alteration of expression of pro-inflammatory and anti-inflammatory factors (17). Given that atorvastatin is effective for treatment of NAFLD, the present study investigated effects of a high-fat/fructose diet (HFFD) with atorvastatin on liver enzymes and lipid profile of a NAFLD rat model.

MATERIALS AND METHODS
Animals and diet
In this study, 32 male Wistar rats weighting 200-250 g were obtained from the Shahid Mirghani Research Institute, Iran. The animals were divided into four groups of eight: 1) normal diet 2) HFFD 3) HFFD + atorvastatin, and 4) Normal + atorvastatin. Atorvastatin was prescribed at the dose of 10 mg/kg (diluted in 6% dimethyl sulfoxide) for eight weeks. The animals were fed ad libitum and housed in pairs in plastic cages, under a controlled temperature (22 ± 2°C) with 12-h light/dark cycles. The normal diet contained 4.30 kcal per gram including 3.87% fat (soy oil), 17.46% casein protein, 68.7% carbohydrates, 8.97% minerals, and 1% vitamins. Hepatosteatosis was induced according to a protocol described previously (18).

Measurement of biochemical indices
The rats were anesthetized by intraperitoneal injection of ketamine (50 mg/kg) and xylazine (5 mg/kg, Merck, Germany). Blood samples were collected and centrifuged at 3,000 rpm for 10 minutes. Levels of AST, ALT, ALP, and GGT were determined by standard enzymatic techniques, and serum cholesterol, TG, LDL, and HDL levels were evaluated using an auto-analyzer (BT-3500, Biotecnica Instruments, Italy).

Statistical analysis
Results are presented as mean ± standard deviation. Data were initially analyzed by one-way analysis of variance (ANOVA). Post hoc Tukey HSD test was applied for group comparisons. A p-value of less than 0.05 was considered to be statistically significant. Statistical analysis of data was carried out in SPSS Statistics (version 22).

Ethical statement
The study was performed in accordance with the “Guide for the Care and Use of Laboratory Animals” published by the US National Institutes of Health (NIH publication No. 85–23, revised 1996). The study protocol was approved by the local ethics committee (IR.GOUMS.REC.1397.274). All efforts were made to minimize animal suffering and reduce the number of animals used.

RESULTS
There was no significant difference in the mean weight of rats before the study, but after 15 weeks of HFFD, a significant difference was observed in the mean weight of animals in all study groups (Table 1).

Table 1. Mean weight of rats before consuming normal diet and HFFD and after 15 weeks of consuming HFFD
After 15 weeks of consuming HFFD
Mean±SD
Before consuming normal diet/HFFD

Mean±SD
Group
390.43±14.32 238.29± 11.52 Normal control
380.44±16.74 243.35±7.05 HFFD control
343.16±12.21 229.66±19.44 HFFD +ATO
256.16±12.21 231.14±18.21 Normal + ATO
0.005 * 0.124 p-value
ATO: Atorvastatin, SD: standard deviation

After 15 weeks of consuming a normal and a high-fat diet, the two groups received atorvastatin for eight weeks, and then, the weight of the rats was measured. The mean weight of rats in the normal control, HFFD control, and normal + atorvastatin groups increased significantly after eight weeks. However, the mean weight of rats in the HFFD + atorvastatin group did not change significantly (Table 2).

Table 2. Mean weight of rats after atorvastatin administration
8th week
Mean±SD
7th week
Mean±SD
6th week
Mean±SD
5th week
Mean±SD
4th week
Mean±SD
3rd week
Mean±SD
Second week
Mean±SD
First week
Mean±SD
Group
454.99±19.27 452.01±19.86 450.41±22.38 427.63±16.23 418.18±26.27 443.53±26.26 420.84±26.98 390.77±23.16 Normal control
461.36±17.81 449.91±17.08 441.47±15.75 427.79±24.85 439.59±12.23 392.36±32.24 378.19±10.01 384.96±25.83 HFFD control
338.84±45.27 343.51±40.36 328.47±46.95 332.99±40.99 330.57±46.53 337.13±37.13 334.30±41.08 343.86±27.84 HFFD + ATO
353.52±23.84 339.65±23.02 342.56±27.77 333.48±21.46 319.35±17.62 302.63±15.39 267.86±18.52 256.23±21.51 Normal + ATO
0.000 * 0.000 * 0.000 * 0.000 * 0.000 * 0.000 * 0.000 * 0.000 * p-value
ATO: Atorvastatin, SD: standard deviation.

There was a significant difference between TG, cholesterol, LDL, HDL, and LDL/HDL ratio between all groups; but, there was no significant difference in the cholesterol/HDL ratio between the groups. The results indicate a significant difference between the liver enzymes of rats in the study groups. Moreover, TG and cholesterol in the HFFD + atorvastatin group were significantly decreased compared with the HFFD control group. In addition, cholesterol and HDL were significantly increased in the normal + atorvastatin group compared with the normal control group. Serum LDL was significantly increased in the HFFD + atorvastatin and the normal + atorvastatin groups compared with other groups.
In liver enzymes, AST was significantly reduced in the HFFD + atorvastatin group compared with the HFFD control. There was a significant difference in the ALT level between the groups taking atorvastatin. In fact, ALT level was lowest in the normal + atorvastatin group. There was a significant increase in the level of ALP and GGT in the normal + atorvastatin group compared with the normal control group. There was also a significant decrease in GGT in the HFFD + atorvastatin group compared with the HFFD control group (Table 3).

Table 3. Comparison of lipid profile and liver enzymes between all study groups
p F Normal + ATO
Mean±SD
HFFD + ATO
Mean±SD
HFFD control
Mean±SD
Normal control
Mean±SD
Variable
0.000 * 38.186 85.84±28.27 86.50±6.71 114.42±11.81 55.00±13.11 TG (mg/dl)
0.000 * 50.605 81.50±3.50 44.33±3.72 69.57±6.05 52.33±4.93 CHO (mg/dl)
0.000 * 135.247 20.13±2.05 5.10±1.11 2.45±1.02 13.33±2.05 LDL (mg/dl)
0.001 * 9.492 62.13±7.77 32.20±6.11 38.74±10.99 30.56±5.97 HDL (mg/dl)
0.000 * 62.061 0.32±0.01 0.16±0.05 0.06±0.02 0.44±0.06 LDL/HDL
0.151 2.044 1.32±0.19 1.41±0.27 1.92±0.61 1.75±0.33 CHO /HDL
0.000* 30.539 116.30±4.30 172.48±18.02 249.30±26.48 118.40±41.90 AST (IU/L)
0.006* 6.237 52.00±9.27 107.21±28.24 141.65±56.12 65.50±25.30 ALT (IU/L)
0.000* 11.022 377.00±12.00 369.00±38.97 409.42±77.68 215.00±22.00 ALP (IU/L)
0.000* 50.769 3.50±0.40 1.10±0.36 3.14±0.17 1.66±0.60 GGT (IU/L)
ATO: Atorvastatin, SD: standard deviation, CHO: cholesterol
* Statistically significant. Data were analyzed by one-way analysis of variance.

DISCUSSION
The results of histological and biochemical evaluations showed a significant increase in the effects of atorvastatin in the HFFD group. In addition, a significant reduction in the activity of serum liver enzymes was noted, which indicates the improvement of tissue lesions. This effect was also confirmed by histological findings. Due to the significant increase in the prevalence of NAFLD in the world, exploiting prevention and treatment methods has become essential. The disease is directly related to lifestyle and diet. Dietary changes in developing countries and the consumption of HFFD as well as physical inactivity have led to an increase in the prevalence of NAFLD. Animal models of NAFLD are widely used due to the limitations of obtaining human liver tissue samples.
Cholesterol synthesis in mammalian cells, including the hepatocytes, is governed by negative feedback loop involving the rate-limiting enzyme for cholesterol synthesis 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). Inhibitors of HMGCR, commonly called statins, are widely used for the treatment of hypercholesterolemia (19). Both HMGCR and LDL receptor are of critical importance in cholesterol homeostasis, and their expression is greatly influenced by intracellular cholesterol levels. Early in vitro and animal studies demonstrated that the decreased production of cholesterol induced by statins was accompanied by a marked compensatory increase in HMGCR and LDL receptor expression (20). More recent studies have found a considerable increase in HMGCR mRNA in response to statins in HepG2 cells (21), rats (22), and miniature pig livers (23). It has been also emphasized that the post-transcriptional modifications affect HMGCR levels, at least in rats (19, 22). Despite the importance of oral statin administration in patients with hyperlipidemia, there have been remarkably few studies on the effects of statins on HMGCR or LDL receptor expression in humans (24, 25). These studies have used mononuclear cells, with the regulation of cholesterol metabolism in such cells having been shown to be similar to that of other tissues (26-29).
Statins can influence the kidney in two main pathways. Rhabdomyolysis is the most serious adverse effect of statin use, though it occurs quite rarely (incidence rate of less than 0.1%). Rhabdomyolysis can induce tubular obstruction, causing tubular injury and ischemia. Statin therapy can be associated with a benign proteinuria due to inhibition of the tubular reabsorption of small molecular weight proteins. The clinical significance of this mild proteinuria is unknown, as the protein differs from that of other glomerular diseases. There has been no evidence of long-term renal dysfunction from statin therapy (30, 31).
In the present study, the induction of NAFLD caused an increase in liver enzymes. After administering atorvastatin in the HFFD group, the level of liver enzymes reduced significantly compared with the HFFD control group. It has been reported that NAFLD is associated with cell damage as well as increased serum levels of AST and ALT. The increase in these factors is greater than the increase observed in metabolic syndrome. Atorvastatin acts as a reducer of bad cholesterol by inhibiting the enzyme β-Hydroxy β-methylglutaryl-CoA (HMG-CoA) in the liver, and HMG-COA plays a key role in cholesterol biosynthesis. It is rapidly absorbed through the gastrointestinal tract. Active drug metabolites are responsible for about 70% of the inhibitory effect of HMG-COA reductase (32). Most drugs used to treat various diseases are metabolized in the liver. Inability of the enzymatic systems in refining these metabolites cause acute and chronic damage to the liver tissue and is directly associated with liver toxicity (33).
According to Ji et al., atorvastatin can lower serum fat and liver enzyme profiles, reduce inflammation, and improve liver function (34). In the present study, in addition to liver enzymes, other biochemical markers that are usually increased in NAFLD were also examined. Almost all indicators confirmed the development of NAFLD. The groups receiving atorvastatin in the present study had improved lipid profiles, and TG and cholesterol levels were significantly reduced compared with the control group.

CONCLUSION
According to the results of the present study, atorvastatin can be used as a potentially suitable medication for the treatment of NAFLD. However, additional studies are needed to determine the exact mechanism of action and its effect on NAFLD complications.

ACKNOWLEDGMENTS
The authors would like to thank the staff of Shahid Mirghani Research Institute and the Kavosh Laboratory in Gorgan.

DECLARATIONS
Funding
This study was supported by the Golestan University of Medical Sciences, Iran. (IR.GOUMS.REC.1397.274)

Ethics approvals and consent to participate
The study was performed in accordance with the “Guide for the Care and Use of Laboratory Animals” published by the US National Institutes of Health (NIH publication No. 85–23, revised 1996). The study protocol was approved by the local ethics committee (IR.GOUMS.REC.1397.274). All efforts were made to minimize animal suffering and reduce the number of animals used.

Conflict of interest
The authors declare that there is no conflict of interest regarding publication of this article.
Research Article: Research Article | Subject: Biochemistry
Received: 2021/07/13 | Accepted: 2021/08/7 | Published: 2022/05/14 | ePublished: 2022/05/14

References
1. Johnson NA, Sachinwalla T, Walton DW, Smith K, Armstrong A, Thompson MW, et al. Aerobic exercise training reduces hepatic and visceral lipids in obese individuals without weight loss. Hepatology. 2009 ;50(4):1105-12. [View at Publisher] [DOI:10.1002/hep.23129] [PubMed] [Google Scholar]
2. Zelber-Sagi S, Ratziu V, Oren R. Nutrition and physical activity in NAFLD: an overview of the epidemiological evidence. World J Gastroenterol. 2011; 17(29): 3377-89. [DOI:10.3748/wjg.v17.i29.3377] [PubMed] [Google Scholar]
3. Hallsworth K, Fattakhova G, Hollingsworth KG, Thoma C, Moore S, Taylor R, et al. Resistance exercise reduces liver fat and its mediators in non-alcoholic fatty liver disease independent of weight loss. Gut. 2011; 60(9): 1278-83. [DOI:10.1136/gut.2011.242073] [PubMed] [Google Scholar]
4. Tazhibi m, Kelishadi r, KHalili Tahmasebi h, Adibi a, Beihaghi a, Salehi hr, et al. Association of lifestyle with metabolic syndrome and non-alcoholic fatty liver in children and adolescence. Hormozgan medical journal. 2010; 14(2): 115-123. [View at Publisher] [Google Scholar]
5. Seyed Khoei, Nazli; Hosseini, Saeed Meraat, Shahin, Golestan, et al. Evaluation of resting metabolism, body composition and some blood structures in men with non-alcoholic fatty liver and its comparison with healthy men. Iranian Journal of Diabetes and Lipids. 2011; 10 (1): 98-101. [View at Publisher] [Google Scholar]
6. Orangi E, Ostad Rahimi A, Mahdavi R, Somi M H, Tarzemani M K. Oxidative Stress-related Parameters and Antioxidant Status in Non-alcoholic Fatty Liver Disease Patients. Iranian Journal of Endocrinology and Metabolism. 2011; 12(5): 493-499. [View at Publisher] [PubMed] [Google Scholar]
7. Tarantino G, Saldalamacchia G, Conca P, Arena A. Non-alcoholic fatty liver disease: Further expression of the metabolic syndrome. JGH. 2007; 22(3): 293-303. [View at Publisher] [PubMed] [Google Scholar]
8. Qureshi K, Abrams GA. Metabolic liver disease of obesity and role of adipose tissue in the pathogenesis of nonalcoholic fatty liver disease. World J Gastroenterol. 2007 14;13(26):3540-53. [View at Publisher] [DOI:10.3748/wjg.v13.i26.3540] [PubMed] [Google Scholar]
9. Barzegarzadeh-Zarandi H, Dabidy-Roshan V.Changes in some liver enzymes and blood lipid level following interval and continuous regular aerobic training in old rats. J Shahrekord Univ Med Sci. 2012; 14(5): 13-23. [View at Publisher] [Google Scholar]
10. Mulhall BP, Ong JP, younossi zm. Non- alacoholic fatty liver disease: an over view. J GastroentrolHepatol. 2002; 17: 1136-43. [View at Publisher] [DOI:10.1046/j.1440-1746.2002.02881.x] [Google Scholar]
11. Mirdar S, Raisi M, Nobahar M. The effect of two-peak exercise training program on some of hepatic stress indexes in active girls. Metabolism and Exercise. 2011; 1(1):11-23. [View at Publisher] [PubMed] [Google Scholar]
12. Chan DF, Li AM, Chu WC, Chan MH, Wong EM, Liu EK, et al. Hepatic steatosis in obese Chinese children. Int J Obes Relat Metab Disord. 2004; 28(10): 1257-63. [View at Publisher] [DOI:10.1038/sj.ijo.0802734] [PubMed] [Google Scholar]
13. Kubota N, Kado S, Kano M, Masuoka N, Nagata Y, Kobayashi T, et al. A high-fat diet and multiple administration of carbon tetrachloride induces liver injury and pathological features associated with non-alcoholic steatohepatitis in mice. Clin Exp Pharmacol Physiol. 2013; 40(7): 422-30. [View at Publisher] [DOI:10.1111/1440-1681.12102] [PubMed] [Google Scholar]
14. Tsuchida T, Lee YA, Fujiwara N, Ybanez M, Allen B, Martins S, et al. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J Hepatol. 2018; 69(2): 385-95. [View at Publisher] [DOI:10.1016/j.jhep.2018.03.011] [PubMed] [Google Scholar]
15. Zhang G, Wang X, Chung TY, Ye W, Hodge L, Zhang L, et al. Carbon tetrachloride (CCl4) accelerated development of non-alcoholic fatty liver disease (NAFLD)/steatohepatitis (NASH) in MS-NASH mice fed western diet supplemented with fructose (WDF). BMC Gastroenterol. 2020; 15; 20(1): 339. [View at Publisher] [DOI:10.21203/rs.3.rs-30324/v1] [PubMed] [Google Scholar]
16. Ridker PM, Rifai N, Pfeffer MA, Sacks FM, Moye LA, Goldman S, et al. Inflammation, pravastatin, and the risk of coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events (CARE) Investigators. Circulation. 1998; 98(9): 839-44. [DOI:10.1161/01.CIR.98.9.839] [PubMed] [Google Scholar]
17. Steinberg D. Hypercholesterolemia and inflammation in atherogenesis: two sides of the same coin. Mol Nutr Food Res. 2005; 49(11): 995-8. [View at Publisher] [DOI:10.1002/mnfr.200500081] [PubMed] [Google Scholar]
18. Yan-Wang A, HeChang A, JunZou BC, XinJin B, Zhongquan QI. The effect of atorvastatin on m-RNA levels of inflammatorygenes expressionin human peripheral blood lymphocytes by DNA microarray. Biomed Pharmacother. 2011; 65(2): 118-22. [View at Publisher] [DOI:10.1016/j.biopha.2010.12.005] [Google Scholar]
19. Eslami Z, Mirghani SJ, Moghanlou AE, Norouzi A, Naseh H, Joshaghani H, et al. An Efficient Model of Non-alcoholic Fatty Liver Disease (NAFLD) Versus Current Experimental Models: Effects of Fructose, Fat, and Carbon Tetrachloride on NAFLD. Hepat Mon. 2021;21(8): e117696. [View at Publisher] [DOI:10.5812/hepatmon.117696] [Google Scholar]
20. Ness GC, Chambers CM. Feedback and hormonal regulation of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase: the concept of cholesterol buffering capacity. Proc Soc Exp Biol Med. 2000; 224(1): 8-19. [View at Publisher] [DOI:10.1046/j.1525-1373.2000.22359.x] [PubMed] [Google Scholar]
21. Endo A. The discovery and development of HMG-CoA reductase inhibitors. J Lipid Res. 1992; 33: 1569-82. [View at Publisher] [DOI:10.1016/S0022-2275(20)41379-3] [PubMed] [Google Scholar]
22. Wilcox LJ, Barrett PHR, Huff MW. Differential regulation of apolipoprotein B secretion from HepG2 cells by two HMG-CoA reductase inhibitors, atorvastatin and simvastatin. J Lipid Res. 1999; 40: 1078-89. [View at Publisher] [DOI:10.1016/S0022-2275(20)33512-4] [PubMed] [Google Scholar]
23. Ness GC, Chambers CM, Lopez D. Atorvastatin action involves diminished recovery of hepatic HMG-CoA reductase activity. J. Lipid Res. 1998; 39: 75-84. [View at Publisher] [DOI:10.1016/S0022-2275(20)34205-X] [PubMed] [Google Scholar]
24. Burnett JR, Wilcox LJ, Telford DE, Kleinstiver SJ, Barrett PH, Newton RS, et al.The magnitude of decrease in hepatic very low density lipoprotein apolipoprotein B secretion is determined by the extent of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibition in miniature pigs. Endocrinology. 1999; 140(11): 5293-302. [View at Publisher] [DOI:10.1210/endo.140.11.7150] [PubMed] [Google Scholar]
25. Wibaut-Berlaimont V, Randi AM, Mandryko V, Lunnon MW, Haskard DO, Naoumova RP. Atorvastatin affects leukocyte gene expression in dyslipidemia patients: in vivo regulation of hemostasis, inflammation and apoptosis. J Thromb Haemost. 2005; 3(4): 677-85. [View at Publisher] [DOI:10.1111/j.1538-7836.2005.01211.x] [PubMed] [Google Scholar]
26. Gouni-Berthold I, Berthold HK, Gylling H, Hallikainen M, Giannakidou E, Stier S, et al. Effects of ezetimibe and/or simvastatin on LDL receptor protein expression and on LDL receptor and HMG-CoA reductase gene expression: a randomized trial in healthy men. Atherosclerosis. 2008; 198(1): 198-207. [View at Publisher] [DOI:10.1016/j.atherosclerosis.2007.09.034] [PubMed] [Google Scholar]
27. Cuthbert JA, Russell DW, Lipsky PE. Regulation of low densitiy lipoprotein receptor gene expression in human lymphocytes. J Biol Chem. 1989; 264: 1298-304. [DOI:10.1016/S0021-9258(19)85085-8] [Google Scholar]
28. Harwood HJ Jr, Bridge DM, Stacpoole PW. In vivo regulation of human mononuclear leukocyte 3-hydroxy-3-methylglutaryl coenzyme A reductase. Studies in normal subjects. J Clin Invest. 1987; 79(4): 1125-32. [View at Publisher] [DOI:10.1172/JCI112928] [PubMed] [Google Scholar]
29. Boucher P, de Lorgeril M, Salen P, Crozier P, Delaye J, Vallon JJ, et al. Effect of dietary cholesterol on low density lipoprotein-receptor, 3-hydroxy-3-methylglutaryl-CoA reductase, and low density lipoprotein receptor-related protein mRNA expression in healthy humans. Lipids. 1998; 33(12): 1177-86. [View at Publisher] [DOI:10.1007/s11745-998-0321-8] [PubMed] [Google Scholar]
30. Powell EE, Kroon PA. Low density lipoprotein receptor and 3-hydroxy-3-methylglutaryl coenzyme A reductase gene expression in human mononuclear leukocytes is regulated coordinately and parallels gene expression in human liver. J Clin Invest. 1994; 93(5): 2168-74. [View at Publisher] [DOI:10.1172/JCI117213] [PubMed] [Google Scholar]
31. BrownWV. Safety of statins. Curr Opin Lipidol. 2008; 19(6): 558-62. [DOI:10.1097/MOL.0b013e328319baba] [PubMed]
32. Ramkumar S, Raghunath A, Raghunath S. Statin Therapy: Review of Safety and Potential Side Effects. Acta Cardiol Sin. 2016 ;32(6):631-639. [PubMed] [Google Scholar]
33. Karanpour Gibchag Z, Heidari R, Abtshi FroushaniM. The effect of combined atorvastatin and zinc sulfate on serum levels of glucose and lipids in type i diabetic rats. Stud Med Sci. 2017; 28(9): 507-19. [View at Publisher] [Google Scholar]
34. Ebrahimi Daryani N, Hashtroudi AA. Drug induced liver diseases. J Med Couac IR Iran. 2004; 22 (2): 129-50. [View at Publisher] [Google Scholar]
35. Ji G, Zhao X, Leng L, Liu P, Jiang Z. Comparison of dietary control and atorvastatin on high fat diet induced hepatic steatosis and hyperlipidemia in rats. Lipids Health Dis. 2011; 10: 23. [View at Publisher] [DOI:10.1186/1476-511X-10-23] [PubMed] [Google Scholar]

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2007 All Rights Reserved | Medical Laboratory Journal

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.