Category: Moms

Metformin and cholesterol

Metformin and cholesterol

Metformin and cholesterol Metformib CAS Google Body composition tracking de la Cuesta-Zuluaga Metforminn, Mueller NT, Metformin and cholesterol V, Velásquez-Mejía EP, Carmona JA, Abad JM, Escobar JS. Measures of biological interaction. Preiss D, Dawed A, Welsh P, Heggie A, Jones AG, Dekker J, Koivula R, Hansen TH, Consortium D, Stewart MC, et al.

Metformin and cholesterol -

If the address matches an existing account you will receive an email with instructions to reset your password. If the address matches an existing account you will receive an email with instructions to retrieve your username.

Antipsychotics can cause numerous side effects, including weight gain and metabolic abnormalities. The off-label use of the type 2 diabetes medication metformin may be able to help patients who are unable to switch to a lower-risk antipsychotic. Despite the great benefit that second-generation antipsychotics can offer patients with schizophrenia and other psychiatric disorders, taking them can lead to serious side effects for some patients, including a high risk of weight gain and metabolic abnormalities.

To help minimize this risk, experts advise that clinicians discuss the importance of healthy eating and regular exercise with patients before prescribing antipsychotics; monitor any changes in weight, blood pressure, or fasting plasma glucose and lipid levels after initiating therapy; and, if significant changes are detected, consider switching patients to a lower-risk antipsychotic.

If switching antipsychotics is not an option, a growing body of literature suggests that the off-label use of metformin—approved for the treatment of type 2 diabetes—may be able to reduce antipsychotic-induced weight gain and insulin resistance.

A study published last month in Molecular Psychiatry also suggests that the medication may reduce low-density lipoprotein cholesterol LDL-C , which is a primary target of cardiovascular disease reduction.

Christoph Correll, M. As described in Molecular Psychiatry , a team of researchers from the United States and China pooled data from two randomized, placebo-controlled trials, including patients from China aged 18 to 40 who developed dyslipidemia within a year of taking clozapine, olanzapine, risperidone, or sulpiride for the treatment of a first episode of schizophrenia.

Sulpiride has not been approved for use in the United States. After 24 weeks of treatment, Of those who had dyslipidemia defined by LDL-C at baseline, 64 percent of patients who took metformin restored their LDL-C to below the normal level compared with Despite these findings, Correll advised that clinicians hold off on prescribing metformin to patients initiating antipsychotic treatment until they know whether significant weight gain will be an issue.

It is advised that patients take metformin with food to reduce the gastrointestinal side effects of the medication, which include nausea, vomiting, diarrhea, and abdominal pain. Inhibition of this pathway leads to inhibition of promoter activity of the insulin gene and to a decrease of insulin secretion [ ].

Insulin secretion may also be impaired via direct statin induced inhibition of mitochondrial oxidative phosphorylation at complex III [ ]. The resulting decrease in ATP synthesis may induce inhibition of insulin secretion via the cascade described above.

One of the mechanisms of an increased insulin resistance could be the effect on the glucose transporter GLUT-4 located in adipose tissue and muscle. Atorvastatin treatment was shown to reduce the surface expression of GLUT-4 in mice adipocytes by inhibiting isoprenylation via inhibition of the mevalonate pathway [ ].

Mevalonate is an important intermediate in cholesterol synthesis and hence also for the synthesis of isoprenoid intermediates Ras and Rho proteins important in cell proliferation. These are involved in intracellular mobilization and localization of proteins.

Statin treatment may cause inactivation of Ras and Rho molecules so that activation and membrane translocalization of GLUT-4 is inhibited. Experiments in mouse adipocytes confirmed that GLUT-4 located on the plasma membrane moved to the cytosol during atorvastatin treatment [ ].

This may result in an increased insulin resistance. In conclusion, statin treatment may lead to a decreased insulin secretion in the β cell via several mechanisms. However, these effects are up to now mainly seen in animal in vitro studies and so it remains elusive whether these results can be translated to humans.

In addition, it should also be kept in mind that 10 years of statin treatment in patients caused an increased BMI 1. It is not clear whether the patients that developed T2DM on statin treatment increased their BMI excessively.

This is in contrast with metformin. Metformin treated mice showed a decreased weight gain which was related to the increased energy consuming conversion of glucose to lactate in the intestinal wall [ ]. In diabetic rats — g it was shown that after 2 weeks metformin—atorvastatin combination therapy mg metformin and 20 mg atorvastatin per 70 kg body weight , glucose-lowering effects, lipid-lowering effects, reduction of oxidative stress, and positive effects on cardiovascular hypertrophy occurred [ ].

The reduction of oxidative stress and protection of the liver observed by studying the liver histology and blood measurement, e.

CRP, TNF-α, IL-6, protein carbonyl levels was also seen in T2DM rats treated with metformin and atorvastatin [ , ].

These positive effects and the fact that a great number of patients are treated with Metformin — statin combination therapy led to the design of a metformin—atorvastatin combination tablet used as a single daily dose [ , ].

There is only a minor chance for toxic drug interactions when metformin and statin are administered together because metformin is not metabolised and most statins are metabolised via the cytochrome P system [ ]. Patients with T2DM are often taking metformin and statins together to control CVD risk as well as glucose metabolism [ 82 ].

Since metformin shows beneficial effects on both dyslipidemia and glycemic control and has been shown to reduce CVD risk while statins may have an added beneficial effect on CVD risk, combined treatment with both drugs seems a good option.

As far as we have been able to discern no randomised clinical trials have been carried out to establish whether combination therapy is superior to monotherapy when focusing on CVD risk.

Ethical considerations maybe prohibitive in this respect but perhaps subgroup analysis in ongoing studies such as the DDPOS may provide an answer. Studies aiming at optimal dosing of both drugs have not been performed. Clinical studies on the effects of metformin and statin combination therapy have been carried out but for different purposes [ 82 , , , , , , , ].

Each of these studies had different objectives and included different patients groups, i. either with T2DM, dyslipidemia, treated different doses , untreated, or newly diagnosed T2DM.

This precludes comparing these studies to arrive at overall results of metformin statin combination therapy. These studies are now discussed briefly to obtain knowledge about the overall effects on glucose and lipid metabolism in T2DM patients with dyslipidemia Table 5.

Atorvastatin 20 mg showed to attenuate the glucose- and HbA1c-lowering effect in combination with and mg metformin. This may complicate analysis of the obtained changes on the glucose and lipid metabolism. However, it could be used for hypothesis-generation rather than making rigid decisions, considering the lack of multiple dose dependent combination studies.

The effects of metformin on lipid homeostasis as discussed in this review article, indicate that lipid metabolism is positively affected in the intestine and liver leading to decreased plasma triglycerides, LDL-C, and total cholesterol.

Metformins effects on lipid metabolism seem to be localized to the intestine. Statins mainly act on plasma cholesterol levels via activation of the LDL-receptor suggesting that combination therapy should show an additional effect on plasma lipids.

However, the data in Table 5 give little indication for an added beneficial effect of both drugs on lipid parameters.

Dedicated studies are required to further investigate the effects of both drugs by combination therapy in humans. Metformin has been shown to exert a significant influence on the composition of the gut microbiota. Interestingly, statins showed such effects as well, particularly in studies with mice and rats [ , ].

Statins were able to decrease the production of butyrate which may relate to the development of new onset T2DM [ ].

Atorvastatin given to hypercholesterolemic patients restored anti-inflammatory bacteria [ ]. In T2DM patients with non-alcoholic fatty liver disease NAFLD beneficial use of combination therapy seems indicated since statin therapy associates negatively with non-alcoholic steatohepatitis and significant fibrosis while a safe use of metformin in patients with T2DM and NAFLD was demonstrated [ ].

Combination therapy consisting of metformin and statin treatment is frequently prescribed to women with an endocrine disorder called polycystic ovary syndrome PCOS. PCOS increases the risk of T2DM and cardiovascular morbidity as it is associated with abnormal increased lipid levels, insulin resistance, systemic inflammation and endothelial dysfunction [ ].

Meta-analysis showed that combined statin-metformin therapy in women with PCOS resulted in improved lipid and inflammation markers but it did not improve insulin sensitivity [ ].

Additional studies are recommended to confirm these results. Combination therapy could also be considered for T2DM patients with diabetic retinopathy.

Diabetic retinopathy DR is a microvascular complication of diabetes caused by hyperglycemia and hyperosmolarity. Leakage and accumulation of fluid in the macula is known as macular edema and results in severe vision loss in DR patients. The use of statins in T2DM patients and pre-existing DR showed a protective effect against development of diabetic macular edema [ ].

Remarkable is that T2DM patients receiving statin therapy in combination with increased levels of cholesterol remnants and triglycerides were associated with slight decreased in left ventricular systolic function. Targeting cholesterol remnants in addition to T2DM patients receiving statins might be beneficial on cardiac function [ ].

From a clinical perspective, it was shown that many patients with T2DM and CVD did not receive lipid lowering therapy while their lipid levels were not in the optimal range [ ].

Increased implementation of guideline recommendations for dyslipidemic T2DM patients is therefore recommended [ ]. Metformin is generally thought to exert its beneficial effects on glucose metabolism mainly in the liver. In line with recent literature on the topic we conclude that the drug acts primarily in the intestine.

This is due to the at least one order of magnitude higher concentrations of metformin in the intestine than in the liver. The drug is certainly not absent in the liver hence parts of its effects may be localized to this organ most probably via its effects on gluconeogenesis.

To treat T2DM and its cardiovascular comorbidity combination therapy of metformins with statins seems well placed and may act as a double-sided sword particularly in the case of statins. This drug increases the risk on T2DM particularly in prediabetic subjects, and cotreatment with metformin might reduce this risk.

However, this hypothesis has not yet been systematically verified. In this review, we have investigated possible sites of interaction of metformin and statins and conclude that they act on largely parallel pathways. Statins reduce plasma cholesterol via activation of LDL-C receptor in the liver and may influence glucose homeostasis primarily by inhibition of insulin secretion in pancreatic β cells.

We propose that combination therapy will ameliorate the risk of statin induced T2DM. Bosi E. Metformin—the gold standard in type 2 diabetes: what does the evidence tell us? Diabetes Obes Metab. Article PubMed CAS Google Scholar. Wu L, Parhofer KG.

Diabetic dyslipidemia. Grundy SM, Brewer HB Jr, Cleeman JI, Smith SC Jr, Lenfant C, American Heart A, National Heart L, Blood I. Article PubMed Google Scholar.

Kitao N, Miyoshi H, Furumoto T, Ono K, Nomoto H, Miya A, Yamamoto C, Inoue A, Tsuchida K, Manda N, et al. The effects of vildagliptin compared with metformin on vascular endothelial function and metabolic parameters: a randomized, controlled trial Sapporo Athero-Incretin Study 3. Cardiovasc Diabetol.

Article PubMed PubMed Central Google Scholar. Mortensen MB, Kulenovic I, Falk E. Statin use and cardiovascular risk factors in diabetic patients developing a first myocardial infarction. Article PubMed PubMed Central CAS Google Scholar. Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW, et al.

Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Corrao G, Ibrahim B, Nicotra F, Soranna D, Merlino L, Catapano AL, Tragni E, Casula M, Grassi G, Mancia G.

Statins and the risk of diabetes: evidence from a large population-based cohort study. Diabetes Care. Fischer J, Ganellin CR, Ganesan A, Proudfoot J. Standalone drugs. In: Ganellin JFACR, editor. Analogue-based drug discovery. Chapter Google Scholar. Timmins P, Donahue S, Meeker J, Marathe P.

Steady-state pharmacokinetics of a novel extended-release metformin formulation. Clin Pharmacokinet. Buse JB, DeFronzo RA, Rosenstock J, Kim T, Burns C, Skare S, Baron A, Fineman M.

The primary glucose-lowering effect of metformin resides in the gut, not the circulation: results from short-term pharmacokinetic and week dose-ranging studies. PubMed CAS Google Scholar. Hashimoto Y, Tanaka M, Okada H, Mistuhashi K, Kimura T, Kitagawa N, Fukuda T, Majima S, Fukuda Y, Tanaka Y, et al.

Postprandial hyperglycemia was ameliorated by taking metformin 30 min before a meal than taking metformin with a meal; a randomized, open-label, crossover pilot study. Hernandez B, Pfluger F, Kruglik SG, Cohen R, Ghomi M.

Protonation-deprotonation and structural dynamics of antidiabetic drug metformin. J Pharm Biomed Anal. Bretnall AE, Clarke GS. Metformin hydrochloride.

Anal Profiles Drug subst Excipients. Article CAS Google Scholar. Orgovan G, Noszal B. Electrodeless, accurate pH determination in highly basic media using a new set of 1 H NMR pH indicators.

Wilcock C, Wyre ND, Bailey CJ. Subcellular distribution of metformin in rat liver. J Pharm Pharmacol. Wiernsperger NF. Membrane physiology as a basis for the cellular effects of metformin in insulin resistance and diabetes.

Diabetes Metab. Kinaan M, Ding H, Triggle CR. Metformin: an old drug for the treatment of diabetes but a new drug for the protection of the endothelium. Med Princ Pract. Bridges HR, Sirvio VA, Agip AN, Hirst J. Molecular features of biguanides required for targeting of mitochondrial respiratory complex I and activation of AMP-kinase.

BMC Biol. Chien HC, Zur AA, Maurer TS, Yee SW, Tolsma J, Jasper P, Scott DO, Giacomini KM. Rapid method to determine intracellular drug concentrations in cellular uptake assays: application to metformin in organic cation transporter 1-transfected human embryonic kidney cells.

Drug Metab Dispos. Article PubMed CAS PubMed Central Google Scholar. He L, Wondisford FE. Metformin action: concentrations matter. Cell Metab. Wilcock C, Bailey CJ. Sites of metformin-stimulated glucose metabolism.

Biochem Pharmacol. Gormsen LC, Sundelin EI, Jensen JB, Vendelbo MH, Jakobsen S, Munk OL, Christensen MM, Brosen K, Frokiaer J, Jessen N. In vivo imaging of human 11C-metformin in peripheral organs: dosimetry, biodistribution and kinetic analyses.

J Nucl Med. Han TK, Proctor WR, Costales CL, Cai H, Everett RS, Thakker DR. Four cation-selective transporters contribute to apical uptake and accumulation of metformin in Caco-2 cell monolayers.

J Pharmacol Exp Ther. McCreight LJ, Bailey CJ, Pearson ER. Metformin and the gastrointestinal tract. Liang X, Chien HC, Yee SW, Giacomini MM, Chen EC, Piao M, Hao J, Twelves J, Lepist EI, Ray AS, et al.

Metformin is a substrate and inhibitor of the human thiamine transporter, THTR-2 SLC19A3. Mol Pharm. Zhou M, Xia L, Wang J. Metformin transport by a newly cloned proton-stimulated organic cation transporter plasma membrane monoamine transporter expressed in human intestine.

Dujic T, Zhou K, Donnelly LA, Tavendale R, Palmer CN, Pearson ER. Association of organic cation transporter 1 with intolerance to metformin in type 2 diabetes: a GoDARTS study.

Muller J, Lips KS, Metzner L, Neubert RH, Koepsell H, Brandsch M. Drug specificity and intestinal membrane localization of human organic cation transporters OCT. Proctor WR, Ming X, Bourdet D, Han TK, Everett RS, Thakker DR.

Why does the intestine lack basolateral efflux transporters for cationic compounds? A provocative hypothesis. J Pharm Sci. Bailey CJ, Wilcock C, Scarpello JH. Metformin and the intestine. Duez H, Lamarche B, Uffelman KD, Valero R, Cohn JS, Lewis GF.

Hyperinsulinemia is associated with increased production rate of intestinal apolipoprotein Bcontaining lipoproteins in humans. Arterioscler Thromb Vasc Biol. Xiao C, Dash S, Morgantini C, Adeli K, Lewis GF.

Gut peptides are novel regulators of intestinal lipoprotein secretion: experimental and pharmacological manipulation of lipoprotein metabolism. Gutierrez-Repiso C, Rodriguez-Pacheco F, Garcia-Arnes J, Valdes S, Gonzalo M, Soriguer F, Moreno-Ruiz FJ, Rodriguez-Cañete A, Gallego-Perales JL, Alcain-Martinez G.

The expression of genes involved in jejunal lipogenesis and lipoprotein synthesis is altered in morbidly obese subjects with insulin resistance. Lab Invest. Jeppesen J, Zhou MY, Chen YD, Reaven GM. Effect of metformin on postprandial lipemia in patients with fairly to poorly controlled NIDDM.

Field FJ, Born E, Murthy S, Mathur SN. Gene expression of sterol regulatory element-binding proteins in hamster small intestine. J Lipid Res. Kohjima M, Higuchi N, Kato M, Kotoh K, Yoshimoto T, Fujino T, Yada M, Yada R, Harada N, Enjoji M, et al.

SREBP-1c, regulated by the insulin and AMPK signaling pathways, plays a role in nonalcoholic fatty liver disease. Int J Mol Med. Srivastava RA, Pinkosky SL, Filippov S, Hanselman JC, Cramer CT, Newton RS. AMP-activated protein kinase: an emerging drug target to regulate imbalances in lipid and carbohydrate metabolism to treat cardio-metabolic diseases.

Tso P, Sun W, Liu M. Gastrointestinal satiety signals IV. Apolipoprotein A-IV. Am J Physiol Gastrointest Liver Physiol. Lutz TA, Osto E. Glucagon-like peptide-1, glucagon-like peptide-2, and lipid metabolism. Curr Opin Lipidol. Dash S, Xiao C, Morgantini C, Lewis GF.

New insights into the regulation of chylomicron production. Annu Rev Nutr. Harmel E, Grenier E, Bendjoudi Ouadda A, El Chebly M, Ziv E, Beaulieu JF, Sane A, Spahis S, Laville M, Levy E.

AMPK in the small intestine in normal and pathophysiological conditions. Rajas F, Bruni N, Montano S, Zitoun C, Mithieux G. The glucose-6 phosphatase gene is expressed in human and rat small intestine: regulation of expression in fasted and diabetic rats. Mithieux G, Gautier-Stein A. Intestinal glucose metabolism revisited.

Diabetes Res Clin Pract. Soty M, Penhoat A, Amigo-Correig M, Vinera J, Sardella A, Vullin-Bouilloux F, Zitoun C, Houberdon I, Mithieux G.

A gut-brain neural circuit controlled by intestinal gluconeogenesis is crucial in metabolic health. Mol Metab. Mithieux G, Rajas F, Zitoun C. Glucose utilization is suppressed in the gut of insulin-resistant high fat-fed rats and is restored by metformin.

Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, Prifti E, Vieira-Silva S, Gudmundsdottir V, Krogh Pedersen H, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, Manneras-Holm L, Stahlman M, Olsson LM, Serino M, Planas-Felix M, et al.

Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med.

Duca FA, Cote CD, Rasmussen BA, Zadeh-Tahmasebi M, Rutter GA, Filippi BM, Lam TK. Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production in rats.

Hardie DG. AMP-activated protein kinase: a master switch in glucose and lipid metabolism. Rev Endocr Metab Disord. Coughlan KA, Valentine RJ, Ruderman NB, Saha AK. AMPK activation: a therapeutic target for type 2 diabetes? Diabetes Metab Syndr Obes.

PubMed PubMed Central Google Scholar. Ruderman NB, Carling D, Prentki M, Cacicedo JM. AMPK, insulin resistance, and the metabolic syndrome. J Clin Invest. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease.

Physiol Rev. Habener JF, Kieffer TJ. Glucagon and glucagon-like peptides, chap. In: Ronald Kahn C, et al. Google Scholar. Mannucci E, Tesi F, Bardini G, Ognibene A, Petracca MG, Ciani S, Pezzatini A, Brogi M, Dicembrini I, Cremasco F, et al. Effects of metformin on glucagon-like peptide-1 levels in obese patients with and without type 2 diabetes.

Diabetes Nutr Metab. Napolitano A, Miller S, Nicholls AW, Baker D, Van Horn S, Thomas E, Rajpal D, Spivak A, Brown JR, Nunez DJ. Novel gut-based pharmacology of metformin in patients with type 2 diabetes mellitus. PLoS ONE. Preiss D, Dawed A, Welsh P, Heggie A, Jones AG, Dekker J, Koivula R, Hansen TH, Consortium D, Stewart MC, et al.

The sustained influence of metformin therapy on circulating GLP-1 levels in individuals with and without type 2 diabetes. Rohde U, Sonne DP, Christensen M, Hansen M, Bronden A, Torang S, Rehfeld JF, Holst JJ, Vilsboll T, Knop FK.

Cholecystokinin-induced gallbladder emptying and metformin elicit additive glucagon-like peptide-1 responses. J Clin Endocrinol Metab. Kim MH, Jee JH, Park S, Lee MS, Kim KW, Lee MK.

Metformin enhances glucagon-like peptide 1 via cooperation between insulin and Wnt signaling. J Endocrinol. Liu YX, Si MM, Lu W, Zhang LX, Zhou CX, Deng SL, Wu HS.

Effects and molecular mechanisms of the antidiabetic fraction of Acorus calamus L. on GLP-1 expression and secretion in vivo and in vitro. J Ethnopharmacol. Trabelsi MS, Daoudi M, Prawitt J, Ducastel S, Touche V, Sayin SI, Perino A, Brighton CA, Sebti Y, Kluza J, et al.

Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat Commun. Jensen JB, Sundelin EI, Jakobsen S, Gormsen LC, Munk OL, Frokiaer J, Jessen N. Zamek-Gliszczynski MJ, Bao JQ, Day JS, Higgins JW. Metformin sinusoidal efflux from the liver is consistent with negligible biliary excretion and absence of enterohepatic cycling.

Scarpello JH, Hodgson E, Howlett HC. Effect of metformin on bile salt circulation and intestinal motility in type 2 diabetes mellitus. Diabet Med. Ryan P, Delzenne N. Gut microbiota and metabolism. In: Hyland N, Stanton C, editors. The Gut-Brain axis.

New York: Elsevier; Wang Z, Koonen D, Hofker M, Fu J. Gut microbiome and lipid metabolism: from associations to mechanisms. Wahlstrom A, Sayin SI, Marschall HU, Backhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism.

de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, Velásquez-Mejía EP, Carmona JA, Abad JM, Escobar JS. Metformin is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut.

Dao MC, Everard A, Aron-Wisnewsky J, Sokolovska N, Prifti E, Verger EO, Kayser BD, Levenez F, Chilloux J, Hoyles L, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Zhou K, Pedersen HK, Dawed AY, Pearson ER.

Pharmacogenomics in diabetes mellitus: insights into drug action and drug discovery. Nat Rev Endocrinol. Wang DS, Jonker JW, Kato Y, Kusuhara H, Schinkel AH, Sugiyama Y.

Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin. Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B. Metformin: from mechanisms of action to therapies.

Gruszka A. New insight into the mechanisms of the anti-hyperglycemic action of metformin. Br J Med Med Res. Article Google Scholar. Baur JA, Birnbaum MJ. Control of gluconeogenesis by metformin: does redox trump energy charge? Viollet B, Guigas B, Leclerc J, Hebrard S, Lantier L, Mounier R, Andreelli F, Foretz M.

AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives. Acta Physiol Oxf. Madsen A, Bozickovic O, Bjune JI, Mellgren G, Sagen JV. Metformin inhibits hepatocellular glucose, lipid and cholesterol biosynthetic pathways by transcriptionally suppressing steroid receptor coactivator 2 SRC Sci Rep.

Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, et al. Role of AMP-activated protein kinase in mechanism of metformin action. Maziere JC, Maziere C, Mora L, Gardette J, Salmon S, Auclair M, Polonovski J.

The antidiabetic drug metformin decreases cholesterol metabolism in cultured human fibroblasts. Koren-Gluzer M, Aviram M, Hayek T. Metformin inhibits macrophage cholesterol biosynthesis rate: possible role for metformin-induced oxidative stress.

Biochem Biophys Res Commun. Liu ZQ, Song XM, Chen QT, Liu T, Teng JT, Zhou K, Luo DQ. Effect of metformin on global gene expression in liver of KKAy mice. Pharmacol Rep. Scott LM, Tomkin GH.

Changes in hepatic and intestinal cholesterol regulatory enzymes. The influence of metformin. Chakraborty A, Chowdhury S, Bhattacharyya M.

Effect of metformin on oxidative stress, nitrosative stress and inflammatory biomarkers in type 2 diabetes patients. Kashi Z, Mahrooz A, Kianmehr A, Alizadeh A. The role of metformin response in lipid metabolism in patients with recent-onset type 2 diabetes: HbA1c level as a criterion for designating patients as responders or nonresponders to metformin.

Ohira M, Miyashita Y, Ebisuno M, Saiki A, Endo K, Koide N, Oyama T, Murano T, Watanabe H, Shirai K. Effect of metformin on serum lipoprotein lipase mass levels and LDL particle size in type 2 diabetes mellitus patients.

Zhang C, Gao F, Luo H, Zhang CT, Zhang R. Sonne DP, Knop FK. Comment on Xu et al. Effects of metformin on metabolite profiles and LDL cholesterol in patients with type 2 diabetes.

Diabetes Care ;— Hofmann AF, Hagey LR. Key discoveries in bile acid chemistry and biology and their clinical applications: history of the last eight decades. The lipid research clinics coronary primary prevention trial results. Reduction in incidence of coronary heart disease.

Luo F, Guo Y, Ruan G, Li X. Metformin promotes cholesterol efflux in macrophages by up-regulating FGF21 expression: a novel anti-atherosclerotic mechanism.

Lipids Health Dis. Nygaard EB, Vienberg SG, Orskov C, Hansen HS, Andersen B. Metformin stimulates FGF21 expression in primary hepatocytes. Exp Diabetes Res. Fan H, Sun X, Zhang H, Liu J, Zhang P, Xu Y, Pan Q, Wang G.

Effect of metformin on fibroblast growth factor levels in patients with newly diagnosed type 2 diabetes. Diabetes Technol Ther. Xu T, Brandmaier S, Messias AC, Herder C, Draisma HH, Demirkan A, Yu Z, Ried JS, Haller T, Heier M, et al. Sone Y, Kido T, Ainuki T, Sonoda M, Ichi I, Kodama S, Sone H, Kondo K, Morita Y, Egawa S, et al.

Genetic variants of the fatty acid desaturase gene cluster are associated with plasma LDL cholesterol levels in Japanese males. J Nutr Sci Vitaminol Tokyo. Zang M, Zuccollo A, Hou X, Nagata D, Walsh K, Herscovitz H, Brecher P, Ruderman NB, Cohen RA.

AMP-activated protein kinase is required for the lipid-lowering effect of metformin in insulin-resistant human HepG2 cells.

J Biol Chem. Wang J, Yang X, Zhang J. Bridges between mitochondrial oxidative stress, ER stress and mTOR signaling in pancreatic β cells.

Cell Signal. Tanaka K, Saisho Y, Manesso E, Tanaka M, Meguro S, Irie J, Sugiura H, Kawai T, Jinzaki M, Cobelli C, et al. Clin Drug Investig.

Cobelli C, Dalla Man C, Toffolo G, Basu R, Vella A, Rizza R. The oral minimal model method. Konopka AR, Esponda RR, Robinson MM, Johnson ML, Carter RE, Schiavon M, Cobelli C, Wondisford FE, Lanza IR, Nair KS.

Hyperglucagonemia mitigates the effect of metformin on glucose production in prediabetes. Cell Rep. Lupi R, Del Guerra S, Fierabracci V, Marselli L, Novelli M, Patane G, Boggi U, Mosca F, Piro S, Del Prato S, et al.

Analysis of data from the UKPDS 34 study [2] showed that intensive therapy with metformin significantly reduces diabetes-related end points and deaths, and a meta-analysis of clinical trials found that the drug could also significantly lower total and LDL cholesterol levels [3].

However, its mechanism of action on blood lipids is still not understood. To try to find out more about how a single drug can have an impact on two completely different aspects of the disease, the researchers pulled data on metformin-treated patients with type 2 diabetes who were not using insulin from the Cooperative Health Research in the Region of Augsburg KORA cohort.

This was a large-scale population-based cohort followed between and , and the researchers analysed the genetic and metabolomic profiles of the patients. The team found that patients with type 2 diabetes treated with metformin had lower levels of three cholesterol metabolites along with lower LDL cholesterol levels.

The findings pointed to metformin working via the AMP-activated protein kinase pathway, and will help to learn more about the drug. Effects of metformin on metabolite profiles and LDL cholesterol in patients with type 2 diabetes.

Metformin and cholesterol Diabetology volume 17 cholwsterol, Article Metformin and cholesterol znd Cite Metfirmin article. Metrics details. Patients with diabetes type 2 have chokesterol increased Antioxidant-Infused Drinks for snd disease and commonly use combination therapy Metformin and cholesterol of Metformln anti-diabetic drug metformin and a cholesterol-lowering statin. However, both drugs act on glucose and lipid metabolism which could lead to adverse effects when used in combination as compared to monotherapy. In this review, the proposed molecular mechanisms of action of statin and metformin therapy in patients with diabetes and dyslipidemia are critically assessed, and a hypothesis for mechanisms underlying interactions between these drugs in combination therapy is developed. Type 2 diabetes mellitus T2DM and cardiovascular diseases are common medical conditions that are often found comorbid with each other. Metformin and cholesterol

Metformin and cholesterol -

Kong , Heung Man Lee , Linda W. Yu , Chun-Chung Chow , Risa Ozaki , Gary T. Ko , Juliana C. Chan; Low HDL Cholesterol, Metformin Use, and Cancer Risk in Type 2 Diabetes : The Hong Kong Diabetes Registry.

Diabetes Care 1 February ; 34 2 : — The AMP-activated protein kinase AMPK pathway is a master regulator in energy metabolism and may be related to cancer. In type 2 diabetes, low HDL cholesterol predicts cancer, whereas metformin usage is associated with reduced cancer risk. Both metformin and apolipoprotein A1 activate the AMPK signaling pathway.

We hypothesize that the anticancer effects of metformin may be particularly evident in type 2 diabetic patients with low HDL cholesterol. In a consecutive cohort of 2, Chinese type 2 diabetic patients enrolled in the study between and , who were free of cancer and not using metformin at enrollment or during 2.

During 13, person-years of follow-up median 5. The anticancer effect of metformin was most evident in type 2 diabetic patients with low HDL cholesterol.

Patients with type 2 diabetes have increased cancer risk, although the risk association of cancer with antidiabetes drugs remains controversial 1. This is in part attributed to heterogeneity in causalities, phenotypes, and treatment responses.

In addition to age and smoking status, abnormal lipids are strong predictors of cancer in type 2 diabetes 2. Our group has reported a V-shaped risk association of HDL cholesterol with cancer, with an optimal level of 1.

Low HDL cholesterol is a common feature of type 2 diabetes 3 and obesity, the latter often considered a linking factor for cancer risk because of insulin resistance 4.

However, apolipoprotein Apo A-I, the main lipoprotein of HDL cholesterol, also can stimulate the phosphorylation of AMP-activated protein kinase AMPK and acetyl-CoA carboxylase ACC to increase glucose uptake in muscle 5 and insulin sensitivity. In this light, the AMPK pathway is considered to be a master switch in sensing and regulating energy metabolism by balancing catabolism lipolysis and anabolism protein and glycogen storage 6.

A large number of factors can activate or inhibit the AMPK signaling pathway, and one of these is the upstream signal, LKB1, a tumor suppressor. In experimental studies, inhibition of the LKB1-AMPK pathway results in tumor formation 7 , whereas metformin activates the LKB1-AMPK pathway and inhibits cancer cell growth 8.

In support of these findings, epidemiological studies have reported reduced cancer risk 9 and associated mortality 10 in type 2 diabetic patients treated with metformin compared with other antidiabetes drugs.

This study argued that the anticancer effects of metformin, if any, would be most evident in patients with low HDL cholesterol. We selected a prospective cohort from the Hong Kong Diabetes Registry enrolled between 1 December and 8 January because drug dispensary data became fully computerized and available for analysis purposes in A detailed description of the Hong Kong Diabetes Registry is available elsewhere 11 — The referral sources of the cohort included general practitioners, community clinics, other specialty clinics, the Prince of Wales Hospital, and other hospitals.

A 4-h assessment of complications and risk factors was performed on an outpatient basis, modified from the European DiabCare protocol Ethical approval was obtained from the Chinese University of Hong Kong Clinical Research Ethics Committee.

This study adhered to the Declaration of Helsinki, and written informed consent was obtained from all patients at the time of assessment, for research purposes. By , 7, diabetic patients were enrolled in the registry since December We sequentially excluded 1 patients with type 1 diabetes or missing data on types of diabetes; 2 45 patients with non-Chinese or unknown nationality; 3 patients with a known history of cancer or receiving cancer treatment at enrollment; 4 patients with missing values on any variables used in the analysis see Table 1 for a list of variables ; and 5 3, patients who used metformin during 2.

The remaining 2, patients were included in the analysis. The cutoff point of 2. Clinical and biochemical characteristics of the study cohort stratified according to occurrence of cancer during follow-up period. Patients attended the center after an 8-h fast and underwent a 4-h structured clinical assessment that included laboratory investigations.

A sterile, random-spot urinary sample was collected to measure albumin-to-creatinine ratio ACR. Total cholesterol, triglycerides, and HDL cholesterol were measured by enzymatic methods on a Hitachi automated analyzer Boehringer Mannheim, Mannheim, Germany using reagent kits supplied by the manufacturer of the analyzer, whereas LDL cholesterol was calculated using the Friedewald equation Drug usage data were extracted from the Hong Kong Hospital Authority Central Computer System, which recorded all drug dispensary data in public hospitals, including the start dates and end dates for each of the drugs of interest.

In Hong Kong, all medications are dispensed on site in both inpatient and outpatient settings. These databases were matched by a unique identification number, the Hong Kong identity card number, which is compulsory for all residents in Hong Kong. A trained team at the Hong Kong Hospital Authority coded all hospital admissions.

All medical admissions of the cohort from enrollment to 30 July were retrieved from the Hong Kong Hospital Authority Central Computer System, which recorded admissions to all public hospitals in Hong Kong.

Additionally, mortality data from the Hong Kong Death Registry during the period were retrieved and cross-checked with hospital discharge status. Hospital discharge principle diagnoses, coded by the International Statistical Classification of Diseases and Related Health Problems 9th Revision ICD-9 , were used to identify cancer events.

The outcome measure of this study was incident cancer fatal or nonfatal: codes — during the follow-up period. We used biological interactions to test whether metformin use was associated with a greater cancer risk reduction in patients with low HDL cholesterol than in those with normal or high HDL cholesterol.

The Statistical Analysis System release 9. Follow-up time was calculated as the period in years from the first enrollment since 1 December to the date of the first cancer event, death, or censoring, whichever came first. We first plotted the full-range association of HDL cholesterol and cancer and further refined cutoff points of HDL cholesterol for cancer risk in the cohort without prevalent metformin users, using restricted cubic spline Cox models Then, we examined the biological interaction for cancer risk between low HDL cholesterol and nonuse of metformin using three measures: 1 relative excess risk caused by interaction RERI ; 2 attributable proportion AP caused by interaction; and 3 the synergy index S A detailed calculation method of additive interaction, including the definition of three indicator variables, an SAS program, and a calculator in Microsoft Excel www.

se , was described by the authors. The RERI is the excess risk attributed to interaction relative to the risk without exposure. AP refers to the attributable proportion of disease, which is caused by the interaction in subjects with both exposures. S is the excess risk from both exposures when there is biological interaction relative to the risk from both exposures without interaction.

A simulation study showed that RERI performed best and AP performed fairly well, but S was problematic in the measure of additivity in the proportional hazard model Use of drugs during follow-up was defined as use of the drugs from enrollment to cancer, death, or censoring date, whichever came first.

By definition, the use of any drugs after the first cancer event was coded as nonuse of these drugs, and any drug users had been given at least one prescription of the drug during follow-up. The total metformin dosage divided by the total number of days during which metformin was prescribed was used as daily metformin dosage.

We also used propensity score to adjust for the likelihood of initiation of metformin during the follow-up period If switching antipsychotics is not an option, a growing body of literature suggests that the off-label use of metformin—approved for the treatment of type 2 diabetes—may be able to reduce antipsychotic-induced weight gain and insulin resistance.

A study published last month in Molecular Psychiatry also suggests that the medication may reduce low-density lipoprotein cholesterol LDL-C , which is a primary target of cardiovascular disease reduction.

Christoph Correll, M. As described in Molecular Psychiatry , a team of researchers from the United States and China pooled data from two randomized, placebo-controlled trials, including patients from China aged 18 to 40 who developed dyslipidemia within a year of taking clozapine, olanzapine, risperidone, or sulpiride for the treatment of a first episode of schizophrenia.

Sulpiride has not been approved for use in the United States. After 24 weeks of treatment, Of those who had dyslipidemia defined by LDL-C at baseline, 64 percent of patients who took metformin restored their LDL-C to below the normal level compared with Despite these findings, Correll advised that clinicians hold off on prescribing metformin to patients initiating antipsychotic treatment until they know whether significant weight gain will be an issue.

It is advised that patients take metformin with food to reduce the gastrointestinal side effects of the medication, which include nausea, vomiting, diarrhea, and abdominal pain. Patients taking metformin are also at a low risk of lactic acidosis.

Another medication that may be able to decrease weight gain in patients taking antipsychotics is topiramate, approved for the treatment of seizure disorders and the prevention of migraine headaches.

While there is some evidence that topiramate can reduce weight gain in patients taking antipsychotic medications, cognitive side effects of the medication have been reported. The research published in Molecular Psychiatry was supported in part by the National Natural Science Foundation of China.

Back to Psychopharmacology Newsletter Table of Contents. Forgot Username? Srivastava RA, Pinkosky SL, Filippov S, Hanselman JC, Cramer CT, Newton RS. AMP-activated protein kinase: an emerging drug target to regulate imbalances in lipid and carbohydrate metabolism to treat cardio-metabolic diseases.

Tso P, Sun W, Liu M. Gastrointestinal satiety signals IV. Apolipoprotein A-IV. Am J Physiol Gastrointest Liver Physiol. Lutz TA, Osto E. Glucagon-like peptide-1, glucagon-like peptide-2, and lipid metabolism.

Curr Opin Lipidol. Dash S, Xiao C, Morgantini C, Lewis GF. New insights into the regulation of chylomicron production. Annu Rev Nutr. Harmel E, Grenier E, Bendjoudi Ouadda A, El Chebly M, Ziv E, Beaulieu JF, Sane A, Spahis S, Laville M, Levy E.

AMPK in the small intestine in normal and pathophysiological conditions. Rajas F, Bruni N, Montano S, Zitoun C, Mithieux G. The glucose-6 phosphatase gene is expressed in human and rat small intestine: regulation of expression in fasted and diabetic rats. Mithieux G, Gautier-Stein A. Intestinal glucose metabolism revisited.

Diabetes Res Clin Pract. Soty M, Penhoat A, Amigo-Correig M, Vinera J, Sardella A, Vullin-Bouilloux F, Zitoun C, Houberdon I, Mithieux G. A gut-brain neural circuit controlled by intestinal gluconeogenesis is crucial in metabolic health.

Mol Metab. Mithieux G, Rajas F, Zitoun C. Glucose utilization is suppressed in the gut of insulin-resistant high fat-fed rats and is restored by metformin. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, Prifti E, Vieira-Silva S, Gudmundsdottir V, Krogh Pedersen H, et al.

Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, Manneras-Holm L, Stahlman M, Olsson LM, Serino M, Planas-Felix M, et al.

Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug.

Nat Med. Duca FA, Cote CD, Rasmussen BA, Zadeh-Tahmasebi M, Rutter GA, Filippi BM, Lam TK. Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production in rats.

Hardie DG. AMP-activated protein kinase: a master switch in glucose and lipid metabolism. Rev Endocr Metab Disord. Coughlan KA, Valentine RJ, Ruderman NB, Saha AK. AMPK activation: a therapeutic target for type 2 diabetes? Diabetes Metab Syndr Obes. PubMed PubMed Central Google Scholar.

Ruderman NB, Carling D, Prentki M, Cacicedo JM. AMPK, insulin resistance, and the metabolic syndrome. J Clin Invest. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease. Physiol Rev. Habener JF, Kieffer TJ. Glucagon and glucagon-like peptides, chap.

In: Ronald Kahn C, et al. Google Scholar. Mannucci E, Tesi F, Bardini G, Ognibene A, Petracca MG, Ciani S, Pezzatini A, Brogi M, Dicembrini I, Cremasco F, et al. Effects of metformin on glucagon-like peptide-1 levels in obese patients with and without type 2 diabetes. Diabetes Nutr Metab.

Napolitano A, Miller S, Nicholls AW, Baker D, Van Horn S, Thomas E, Rajpal D, Spivak A, Brown JR, Nunez DJ. Novel gut-based pharmacology of metformin in patients with type 2 diabetes mellitus.

PLoS ONE. Preiss D, Dawed A, Welsh P, Heggie A, Jones AG, Dekker J, Koivula R, Hansen TH, Consortium D, Stewart MC, et al. The sustained influence of metformin therapy on circulating GLP-1 levels in individuals with and without type 2 diabetes.

Rohde U, Sonne DP, Christensen M, Hansen M, Bronden A, Torang S, Rehfeld JF, Holst JJ, Vilsboll T, Knop FK. Cholecystokinin-induced gallbladder emptying and metformin elicit additive glucagon-like peptide-1 responses.

J Clin Endocrinol Metab. Kim MH, Jee JH, Park S, Lee MS, Kim KW, Lee MK. Metformin enhances glucagon-like peptide 1 via cooperation between insulin and Wnt signaling. J Endocrinol. Liu YX, Si MM, Lu W, Zhang LX, Zhou CX, Deng SL, Wu HS.

Effects and molecular mechanisms of the antidiabetic fraction of Acorus calamus L. on GLP-1 expression and secretion in vivo and in vitro. J Ethnopharmacol. Trabelsi MS, Daoudi M, Prawitt J, Ducastel S, Touche V, Sayin SI, Perino A, Brighton CA, Sebti Y, Kluza J, et al.

Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells. Nat Commun. Jensen JB, Sundelin EI, Jakobsen S, Gormsen LC, Munk OL, Frokiaer J, Jessen N.

Zamek-Gliszczynski MJ, Bao JQ, Day JS, Higgins JW. Metformin sinusoidal efflux from the liver is consistent with negligible biliary excretion and absence of enterohepatic cycling. Scarpello JH, Hodgson E, Howlett HC. Effect of metformin on bile salt circulation and intestinal motility in type 2 diabetes mellitus.

Diabet Med. Ryan P, Delzenne N. Gut microbiota and metabolism. In: Hyland N, Stanton C, editors. The Gut-Brain axis. New York: Elsevier; Wang Z, Koonen D, Hofker M, Fu J. Gut microbiome and lipid metabolism: from associations to mechanisms.

Wahlstrom A, Sayin SI, Marschall HU, Backhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, Velásquez-Mejía EP, Carmona JA, Abad JM, Escobar JS. Metformin is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut.

Dao MC, Everard A, Aron-Wisnewsky J, Sokolovska N, Prifti E, Verger EO, Kayser BD, Levenez F, Chilloux J, Hoyles L, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Zhou K, Pedersen HK, Dawed AY, Pearson ER.

Pharmacogenomics in diabetes mellitus: insights into drug action and drug discovery. Nat Rev Endocrinol. Wang DS, Jonker JW, Kato Y, Kusuhara H, Schinkel AH, Sugiyama Y. Involvement of organic cation transporter 1 in hepatic and intestinal distribution of metformin.

Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B. Metformin: from mechanisms of action to therapies. Gruszka A. New insight into the mechanisms of the anti-hyperglycemic action of metformin.

Br J Med Med Res. Article Google Scholar. Baur JA, Birnbaum MJ. Control of gluconeogenesis by metformin: does redox trump energy charge? Viollet B, Guigas B, Leclerc J, Hebrard S, Lantier L, Mounier R, Andreelli F, Foretz M.

AMP-activated protein kinase in the regulation of hepatic energy metabolism: from physiology to therapeutic perspectives.

Acta Physiol Oxf. Madsen A, Bozickovic O, Bjune JI, Mellgren G, Sagen JV. Metformin inhibits hepatocellular glucose, lipid and cholesterol biosynthetic pathways by transcriptionally suppressing steroid receptor coactivator 2 SRC Sci Rep.

Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, et al. Role of AMP-activated protein kinase in mechanism of metformin action.

Maziere JC, Maziere C, Mora L, Gardette J, Salmon S, Auclair M, Polonovski J. The antidiabetic drug metformin decreases cholesterol metabolism in cultured human fibroblasts.

Koren-Gluzer M, Aviram M, Hayek T. Metformin inhibits macrophage cholesterol biosynthesis rate: possible role for metformin-induced oxidative stress. Biochem Biophys Res Commun. Liu ZQ, Song XM, Chen QT, Liu T, Teng JT, Zhou K, Luo DQ. Effect of metformin on global gene expression in liver of KKAy mice.

Pharmacol Rep. Scott LM, Tomkin GH. Changes in hepatic and intestinal cholesterol regulatory enzymes. The influence of metformin. Chakraborty A, Chowdhury S, Bhattacharyya M. Effect of metformin on oxidative stress, nitrosative stress and inflammatory biomarkers in type 2 diabetes patients.

Kashi Z, Mahrooz A, Kianmehr A, Alizadeh A. The role of metformin response in lipid metabolism in patients with recent-onset type 2 diabetes: HbA1c level as a criterion for designating patients as responders or nonresponders to metformin. Ohira M, Miyashita Y, Ebisuno M, Saiki A, Endo K, Koide N, Oyama T, Murano T, Watanabe H, Shirai K.

Effect of metformin on serum lipoprotein lipase mass levels and LDL particle size in type 2 diabetes mellitus patients. Zhang C, Gao F, Luo H, Zhang CT, Zhang R. Sonne DP, Knop FK. Comment on Xu et al. Effects of metformin on metabolite profiles and LDL cholesterol in patients with type 2 diabetes.

Diabetes Care ;— Hofmann AF, Hagey LR. Key discoveries in bile acid chemistry and biology and their clinical applications: history of the last eight decades.

The lipid research clinics coronary primary prevention trial results. Reduction in incidence of coronary heart disease. Luo F, Guo Y, Ruan G, Li X. Metformin promotes cholesterol efflux in macrophages by up-regulating FGF21 expression: a novel anti-atherosclerotic mechanism. Lipids Health Dis. Nygaard EB, Vienberg SG, Orskov C, Hansen HS, Andersen B.

Metformin stimulates FGF21 expression in primary hepatocytes. Exp Diabetes Res. Fan H, Sun X, Zhang H, Liu J, Zhang P, Xu Y, Pan Q, Wang G. Effect of metformin on fibroblast growth factor levels in patients with newly diagnosed type 2 diabetes.

Diabetes Technol Ther. Xu T, Brandmaier S, Messias AC, Herder C, Draisma HH, Demirkan A, Yu Z, Ried JS, Haller T, Heier M, et al. Sone Y, Kido T, Ainuki T, Sonoda M, Ichi I, Kodama S, Sone H, Kondo K, Morita Y, Egawa S, et al.

Genetic variants of the fatty acid desaturase gene cluster are associated with plasma LDL cholesterol levels in Japanese males. J Nutr Sci Vitaminol Tokyo. Zang M, Zuccollo A, Hou X, Nagata D, Walsh K, Herscovitz H, Brecher P, Ruderman NB, Cohen RA.

AMP-activated protein kinase is required for the lipid-lowering effect of metformin in insulin-resistant human HepG2 cells. J Biol Chem.

Wang J, Yang X, Zhang J. Bridges between mitochondrial oxidative stress, ER stress and mTOR signaling in pancreatic β cells. Cell Signal. Tanaka K, Saisho Y, Manesso E, Tanaka M, Meguro S, Irie J, Sugiura H, Kawai T, Jinzaki M, Cobelli C, et al.

Clin Drug Investig. Cobelli C, Dalla Man C, Toffolo G, Basu R, Vella A, Rizza R. The oral minimal model method. Konopka AR, Esponda RR, Robinson MM, Johnson ML, Carter RE, Schiavon M, Cobelli C, Wondisford FE, Lanza IR, Nair KS.

Hyperglucagonemia mitigates the effect of metformin on glucose production in prediabetes. Cell Rep. Lupi R, Del Guerra S, Fierabracci V, Marselli L, Novelli M, Patane G, Boggi U, Mosca F, Piro S, Del Prato S, et al.

Lipotoxicity in human pancreatic islets and the protective effect of metformin. Long-Term exposure of pancreatic beta-cells to palmitate results in SREBP-1C-dependent decreases in GLP-1 receptor signaling via CREB and AKT and insulin secretory response. Dai YL, Huang SL, Leng Y.

AICAR and metformin exert AMPK-dependent effects on INS-1E pancreatic beta-cell apoptosis via differential downstream mechanisms. Int J Biol Sci. Chang TJ, Tseng HC, Liu MW, Chang YC, Hsieh ML, Chuang LM. Glucagon-like peptide-1 prevents methylglyoxal-induced apoptosis of beta cells through improving mitochondrial function and suppressing prolonged AMPK activation.

Muhammed SJ, Lundquist I, Salehi A. Pancreatic beta-cell dysfunction, expression of iNOS and the effect of phosphodiesterase inhibitors in human pancreatic islets of type 2 diabetes.

Mezghenna K, Pomies P, Chalancon A, Castex F, Leroy J, Niclauss N, Nadal B, Cambier L, Cazevieille C, Petit P, et al.

Increased neuronal nitric oxide synthase dimerisation is involved in rat and human pancreatic beta cell hyperactivity in obesity. Lundquist I, Mohammed Al-Amily I, Meidute Abaraviciene S, Salehi A. Metformin ameliorates dysfunctional traits of glibenclamide- and glucose-induced insulin secretion by suppression of imposed overactivity of the islet nitric oxide synthase-NO system.

Brereton MF, Rohm M, Shimomura K, Holland C, Tornovsky-Babeay S, Dadon D, Iberl M, Chibalina MV, Lee S, Glaser B, et al. Hyperglycaemia induces metabolic dysfunction and glycogen accumulation in pancreatic beta-cells.

Consortium R. Restoring insulin secretion RISE : design of studies of beta-cell preservation in prediabetes and early type 2 diabetes across the life span.

Force USPST, Bibbins-Domingo K, Grossman DC, Curry SJ, Davidson KW, Epling JW Jr, Garcia FA, Gillman MW, Kemper AR, Krist AH, et al. Statin use for the primary prevention of cardiovascular disease in adults: US preventive services task force recommendation statement.

Gruzdeva O, Uchasova E, Dyleva Y, Akbasheva O, Karetnikova V, Shilov A, Barbarash O. Effect of different doses of statins on the development of type 2 diabetes mellitus in patients with myocardial infarction.

Diabetes Metab Syndr Obes Targets Ther. Gruzdeva O, Uchasova E, Dyleva Y, Akbasheva O, Karetnikova V, Barbarash O. Early effects of treatment low-dose atorvastatin on markers of insulin resistance and inflammation in patients with myocardial infarction.

Front Pharmacol. Anyanwagu U, Idris I, Donnelly R. Drug-induced diabetes mellitus: evidence for statins and other drugs affecting glucose metabolism. Clin Pharmacol Ther. Wang S, Cai R, Yuan Y, Varghese Z, Moorhead J, Ruan XZ.

Association between reductions in low-density lipoprotein cholesterol with statin therapy and the risk of new-onset diabetes: a meta-analysis. Gotoh S, Negishi M. Statin-activated nuclear receptor PXR promotes SGK2 dephosphorylation by scaffolding PP2C to induce hepatic gluconeogenesis.

Hakkola J, Rysa J, Hukkanen J. Regulation of hepatic energy metabolism by the nuclear receptor PXR. Biochim Biophys Acta.

He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. Ling Z, Shu N, Xu P, Wang F, Zhong Z, Sun B, Li F, Zhang M, Zhao K, Tang X, et al. Involvement of pregnane X receptor in the impaired glucose utilization induced by atorvastatin in hepatocytes.

Black RN, Ennis CN, Young IS, Hunter SJ, Atkinson AB, Bell PM. The peroxisome proliferator-activated receptor alpha agonist fenofibrate has no effect on insulin sensitivity compared to atorvastatin in type 2 diabetes mellitus; a randomised, double-blind controlled trial.

J Diabetes Complications. Szendroedi J, Anderwald C, Krssak M, Bayerle-Eder M, Esterbauer H, Pfeiler G, Brehm A, Nowotny P, Hofer A, Waldhausl W, et al.

Effects of high-dose simvastatin therapy on glucose metabolism and ectopic lipid deposition in nonobese type 2 diabetic patients. Ruscica M, Macchi C, Morlotti B, Sirtori CR, Magni P.

Statin therapy and related risk of new-onset type 2 diabetes mellitus. Eur J Intern Med. Hao M, Head WS, Gunawardana SC, Hasty AH, Piston DW. Direct effect of cholesterol on insulin secretion: a novel mechanism for pancreatic beta-cell dysfunction.

Zhou J, Li W, Xie Q, Hou Y, Zhan S, Yang X, Xu X, Cai J, Huang Z. Effects of simvastatin on glucose metabolism in mouse MIN6 cells. J Diabetes Res. Vergeer M, Brunham LR, Koetsveld J, Kruit JK, Verchere CB, Kastelein JJ, Hayden MR, Stroes ES.

Carriers of loss-of-function mutations in ABCA1 display pancreatic beta-cell dysfunction. Yaluri N, Modi S, Lopez Rodriguez M, Stancakova A, Kuusisto J, Kokkola T, Laakso M.

Simvastatin impairs insulin secretion by multiple mechanisms in MIN6 cells. Scattolini V, Luni C, Zambon A, Galvanin S, Gagliano O, Ciubotaru CD, Avogaro A, Mammano F, Elvassore N, Fadini GP.

Simvastatin rapidly and reversibly inhibits insulin secretion in intact single-islet cultures. Diabetes Ther. Sun H, Li Y, Sun B, Hou N, Yang J, Zheng M, Xu J, Wang J, Zhang Y, Zeng X, et al.

Medicine Baltimore. Schirris TJ, Renkema GH, Ritschel T, Voermans NC, Bilos A, van Engelen BG, Brandt U, Koopman WJ, Beyrath JD, Rodenburg RJ, et al.

Statin-induced myopathy is associated with mitochondrial complex iii inhibition. Elbadawi-Sidhu M, Baillie RA, Zhu H, Chen YDI, Goodarzi MO, Rotter JI, Krauss RM, Fiehn O, Kaddurah-Daouk R.

Pharmacometabolomic signature links simvastatin therapy and insulin resistance. Takaguri A, Satoh K, Itagaki M, Tokumitsu Y, Ichihara K. Effects of atorvastatin and pravastatin on signal transduction related to glucose uptake in 3T3L1 adipocytes.

J Pharmacol Sci. Nakata M, Nagasaka S, Kusaka I, Matsuoka H, Ishibashi S, Yada T. Effects of statins on the adipocyte maturation and expression of glucose transporter 4 SLC2A4 : implications in glycaemic control.

Sugiyama T, Tsugawa Y, Tseng CH, Kobayashi Y, Shapiro MF. Different time trends of caloric and fat intake between statin users and nonusers among US adults: gluttony in the time of statins?

JAMA Intern Med. Schommers P, Thurau A, Bultmann-Mellin I, Guschlbauer M, Klatt AR, Rozman J, Klingenspor M, de Angelis MH, Alber J, Gründemann D. Metformin causes a futile intestinal—hepatic cycle which increases energy expenditure and slows down development of a type 2 diabetes-like state.

Molecular Metabolism. Islam M, Alam A, Rahman M, Ali Y, Mamun A, Rahman M, Hossain A, Rashid M. Effects of combination of antidiabetic agent and statin on alloxan-induced diabetes with cardiovascular diseases in rats. J Sci Res. CAS Google Scholar. Matafome P, Louro T, Rodrigues L, Crisostomo J, Nunes E, Amaral C, Monteiro P, Cipriano A, Seica R.

Metformin and atorvastatin combination further protect the liver in type 2 diabetes with hyperlipidaemia. Diabetes Metab Res Rev. Singh BK, Singh A, Kumar V. Ameliorative effect of adjunct therapy of metformin with atorvastatin on streptozotocin-induced diabetes mellitus in rats.

Drug Res Stuttg. Oh JH, Eun Lee J, Jeong Kim Y, Oh TO, Han S, Jeon EK, Shin K, Kim DH, Hye Park C, Lee YJ. Designing of the fixed-dose gastroretentive bilayer tablet for sustained release of metformin and immediate release of atorvastatin.

Drug Dev Ind Pharm. Kandhwal K, Dey S, Nazarudheen S, Arora R, Reyar S, Thudi NR, Monif T, Singh MK, Rao S. Pharmacokinetics of a fixed-dose combination of atorvastatin and metformin extended release versus concurrent administration of individual formulations: a randomized, open-label, two-treatment, two-period, two-sequence, single-dose, crossover, bioequivalence study.

Scheen AJ. Drug interactions of clinical importance with antihyperglycaemic agents: an update. Drug Saf.

Balasubramanian R, Varadharajan S, Kathale A, Nagraj LM, Periyandavar I, Nayak UP, Sharma A, Bolmall C, Baliga VP. J Indian Med Assoc. Krysiak R, Okopien B. Haemostatic effects of metformin in simvastatin-treated volunteers with impaired fasting glucose. Basic Clin Pharmacol Toxicol.

Lymphocyte-suppressing and systemic anti-inflammatory effects of high-dose metformin in simvastatin-treated patients with impaired fasting glucose.

The effect of metformin on monocyte secretory function in simvastatin-treated patients with impaired fasting glucose. Tousoulis D, Koniari K, Antoniades C, Papageorgiou N, Miliou A, Noutsou M, Nikolopoulou A, Marinou K, Stefanadi E, Siasos G, et al. Combined effects of atorvastatin and metformin on glucose-induced variations of inflammatory process in patients with diabetes mellitus.

Int J Cardiol. Tousoulis D, Koniari K, Antoniades C, Miliou A, Noutsou M, Nikolopoulou A, Papageorgiou N, Marinou K, Stefanadi E, Stefanadis C. Impact of 6 weeks of treatment with low-dose metformin and atorvastatin on glucose-induced changes of endothelial function in adults with newly diagnosed type 2 diabetes mellitus: a single-blind study.

Clin Ther. Hao Z, Liu Y, Liao H, Zheng D, Xiao C, Li G. Exp Clin Endocrinol Diabetes. Caparros-Martin JA, Lareu RR, Ramsay JP, Peplies J, Reen FJ, Headlam HA, Ward NC, Croft KD, Newsholme P, Hughes JD, et al. Statin therapy causes gut dysbiosis in mice through a PXR-dependent mechanism.

Khan TJ, Ahmed YM, Zamzami MA, Mohamed SA, Khan I, Baothman OAS, Mehanna MG, Yasir M. Effect of atorvastatin on the gut microbiota of high fat diet-induced hypercholesterolemic rats. Khan TJ, Ahmed YM, Zamzami MA, Siddiqui AM, Khan I, Baothman OAS, Mehanna MG, Kuerban A, Kaleemuddin M, Yasir M.

Atorvastatin treatment modulates the gut microbiota of the hypercholesterolemic patients. Nascimbeni F, Aron-Wisnewsky J, Pais R, Tordjman J, Poitou C, Charlotte F, Bedossa P, Poynard T, Clement K, Ratziu V, et al. Statins, antidiabetic medications and liver histology in patients with diabetes with non-alcoholic fatty liver disease.

BMJ Open Gastroenterol. Ehrmann DA. Polycystic ovary syndrome. N Engl J Med. Sun J, Yuan Y, Cai R, Sun H, Zhou Y, Wang P, Huang R, Xia W, Wang S. An investigation into the therapeutic effects of statins with metformin on polycystic ovary syndrome: a meta-analysis of randomised controlled trials.

BMJ Open. Chung Y-R, Park SW, Choi S-Y, Kim SW, Moon KY, Kim JH, Lee K. Association of statin use and hypertriglyceridemia with diabetic macular edema in patients with type 2 diabetes and diabetic retinopathy.

Jorgensen PG, Jensen MT, Biering-Sorensen T, Mogelvang R, Galatius S, Fritz-Hansen T, Rossing P, Vilsboll T, Jensen JS. Cholesterol remnants and triglycerides are associated with decreased myocardial function in patients with type 2 diabetes.

Hanefeld M, Traylor L, Gao L, Landgraf W. Besseling J, Hutten BA. Is there a link between diabetes and cholesterol metabolism? Expert Rev Cardiovasc Ther. Cederberg H, Stancakova A, Yaluri N, Modi S, Kuusisto J, Laakso M.

Increased risk of diabetes with statin treatment is associated with impaired insulin sensitivity and insulin secretion: a 6 year follow-up study of the METSIM cohort.

Chehade JM, Gladysz M, Mooradian AD. Dyslipidemia in type 2 diabetes: prevalence, pathophysiology, and management. Feingold K, Grunfeld C. Role of glucose and lipids in the cardiovascular disease of patients with diabetes. In: De Groot LJ, Chrousos G, Dungan K, et al. South Dartmouth MA : MDText.

com, Inc. Jaiswal M, Schinske A, Pop-Busui R. Lipids and lipid management in diabetes. Best Pract Res Clin Endocrinol Metab. Ng DS. Diabetic dyslipidemia: from evolving pathophysiological insight to emerging therapeutic targets. Can J Diabetes. Soran H, Schofield JD, Adam S, Durrington PN. Diabetic dyslipidaemia.

Schofield JD, Liu Y, Rao-Balakrishna P, Malik RA, Soran H. Diabetes dyslipidemia. Verges B. Pathophysiology of diabetic dyslipidaemia: where are we? Wang CCL, Hess CN, Hiatt WR, Goldfine AB. Clinical update: cardiovascular disease in diabetes mellitus atherosclerotic cardiovascular disease and heart failure in type 2 diabetes mellitus-mechanisms, management, and clinical considerations.

Arca M. Alterations of intestinal lipoprotein metabolism in diabetes mellitus and metabolic syndrome. Atheroscler Suppl. Tomkin GH, Owens D. Dyslipidaemia of diabetes and the intestine. World J Diabetes. Arca M, Pigna G, Favoccia C.

Mechanisms of diabetic dyslipidemia: relevance for atherogenesis. Curr Vasc Pharmacol. Perry RJ, Samuel VT, Petersen KF, Shulman GI. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes.

Taskinen MR, Boren J. New insights into the pathophysiology of dyslipidemia in type 2 diabetes.

Cardiovascular Diabetology volume 18Article cholesterlo 54 Cite this Nutrient absorption in the liver. Metrics details. Metformin and cholesterol an increasing global burden of coronary artery disease Metformin and cholesterolcholewterol detection and timely management of risk factors are crucial to reduce morbidity and mortality in such patients. Diabetes mellitus DM is considered an independent risk factor for the development of CAD. Metformin, an anti-diabetic drug, has been shown in pre-clinical and clinical studies, to lower the cardiovascular events in the DM patients.

Author: Tygogar

2 thoughts on “Metformin and cholesterol

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com