Anti-Inflammatory Effects of Metformin Irrespective of Diabetes Status

Amy R Cameron, Vicky L Morrison, Daniel Levin, Mohapradeep Mohan, Calum Forteath, Craig Beall, Alison D McNeilly, David J K Balfour, Terhi Savinko, Aaron K F Wong, Benoit Viollet, Kei Sakamoto, Susanna C Fagerholm, Marc Foretz, Chim C Lang, Graham Rena, Amy R Cameron, Vicky L Morrison, Daniel Levin, Mohapradeep Mohan, Calum Forteath, Craig Beall, Alison D McNeilly, David J K Balfour, Terhi Savinko, Aaron K F Wong, Benoit Viollet, Kei Sakamoto, Susanna C Fagerholm, Marc Foretz, Chim C Lang, Graham Rena

Abstract

Rationale: The diabetes mellitus drug metformin is under investigation in cardiovascular disease, but the molecular mechanisms underlying possible benefits are poorly understood.

Objective: Here, we have studied anti-inflammatory effects of the drug and their relationship to antihyperglycemic properties.

Methods and results: In primary hepatocytes from healthy animals, metformin and the IKKβ (inhibitor of kappa B kinase) inhibitor BI605906 both inhibited tumor necrosis factor-α-dependent IκB degradation and expression of proinflammatory mediators interleukin-6, interleukin-1β, and CXCL1/2 (C-X-C motif ligand 1/2). Metformin suppressed IKKα/β activation, an effect that could be separated from some metabolic actions, in that BI605906 did not mimic effects of metformin on lipogenic gene expression, glucose production, and AMP-activated protein kinase activation. Equally AMP-activated protein kinase was not required either for mitochondrial suppression of IκB degradation. Consistent with discrete anti-inflammatory actions, in macrophages, metformin specifically blunted secretion of proinflammatory cytokines, without inhibiting M1/M2 differentiation or activation. In a large treatment naive diabetes mellitus population cohort, we observed differences in the systemic inflammation marker, neutrophil to lymphocyte ratio, after incident treatment with either metformin or sulfonylurea monotherapy. Compared with sulfonylurea exposure, metformin reduced the mean log-transformed neutrophil to lymphocyte ratio after 8 to 16 months by 0.09 U (95% confidence interval, 0.02-0.17; P=0.013) and increased the likelihood that neutrophil to lymphocyte ratio would be lower than baseline after 8 to 16 months (odds ratio, 1.83; 95% confidence interval, 1.22-2.75; P=0.00364). Following up these findings in a double-blind placebo controlled trial in nondiabetic heart failure (trial registration: NCT00473876), metformin suppressed plasma cytokines including the aging-associated cytokine CCL11 (C-C motif chemokine ligand 11).

Conclusion: We conclude that anti-inflammatory properties of metformin are exerted irrespective of diabetes mellitus status. This may accelerate investigation of drug utility in nondiabetic cardiovascular disease groups.

Clinical trial registration: Name of the trial registry: TAYSIDE trial (Metformin in Insulin Resistant Left Ventricular [LV] Dysfunction). URL: https://www.clinicaltrials.gov. Unique identifier: NCT00473876.

Keywords: NF-kappa B; cardiovascular diseases; diabetes mellitus; heart failure; inflammation; metabolism; metformin.

© 2016 The Authors.

Figures

Figure 1.
Figure 1.
Effect of metformin on nuclear factorB (NF-κB) signaling and gene expression. AC, Primary hepatocytes were incubated in serum-free medium overnight and then stimulated for 3 h with or without 0.5 to 5 mmol/L metformin. For the last 15 min, cells were treated with or without 10 ng/mL tumor necrosis factor (TNF)-α. Cells were lysed and prepared for immunoblotting using antibodies as described in the Methods section of this article. In this figure and elsewhere, each blot is representative of experiments carried out at least 3×. DF, Primary hepatocytes were incubated as in AC, before stimulation for 3 h with or without 2 mmol/L metformin and TNF-α. In addition, cells were incubated with/without 10 μmol/L BI605906 or 100 nmol/L rapamycin as shown, before lysis and immunoblotting as described in the Methods section of this article. GJ, Primary hepatocytes were treated with or without 10 ng/mL TNF-α, 2 mmol/L metformin, or 10 μmol/L BI605906 for 8 h followed by cell lysis, RNA extraction, and preparation of cDNA for real-time-polymerase chain reaction using primer sets for individual genes shown as described in the Methods section of this article. ACC indicates acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; p-ACC, phospho–acetyl-CoA carboxylase; and pAMPK, phospho–AMP-activated protein kinase.
Figure 2.
Figure 2.
Effect of 5-aminoimidazole-4-carboxamide riboside (AICAR) and A769662 on nuclear factorB (NF-κB) signaling. AD, Primary wild-type (WT) hepatocytes (A and B) and those taken from double-knockout (KO) AMPK animals or matched controls (WT) (C) were incubated in serum-free medium overnight, before stimulation for 3 h with or without doses of A769662 and AICAR as shown. For the last 15 min, cells were treated with or without 10 ng/mL tumor necrosis factor (TNF)-α. D, Hepatocytes from KO or WT animals treated with and without doses of metformin for 3 h. For the last 15 min, cells were treated with 10 ng/mL TNF-α. Cells were then lysed, and immunoblots were prepared as described in the Methods section of this article and in Figure 1. ACC indicates acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; IKK, inhibitor of kappa B kinase; p-ACC, phospho–acetyl-CoA carboxylase; and pAMPK, phospho–AMP-activated protein kinase.
Figure 3.
Figure 3.
Effects of cytokines on glucose production and lipogenic gene expression in primary hepatocytes. A and B, Primary hepatocytes were treated with/without metformin (2 mmol/L), interleukin (IL)-6 (5 ng/mL), IL-1β (10 ng/mL), chemokine (C-X-C motif) ligand (CXCL) 1 (100 ng/mL) and tumor necrosis factor (TNF)-α (10 ng/mL) for 12 h, and glucose production was measured by GAGO (glucose [glucose oxidase]) assay as described in the Methods section of this article. CE, Primary hepatocytes were treated with or without 10 ng/mL tumor necrosis factor (TNF)-α, 2 mmol/L metformin, and 10 μmol/L BI605906 for 8 h followed by cell lysis, RNA extraction, and preparation of cDNA for real-time polymerase chain reaction using primer sets for individual genes shown as described in the Methods section of this article. FASN indicates fatty acid synthase; PPAR, peroxisome proliferator–activated receptor; and SREBP, sterol regulatory element-binding protein.
Figure 4.
Figure 4.
Effect of metformin and its analogue biguanide on bone marrow–derived macrophages: phenotypic markers and cytokine secretion. A, Macrophages were treated with/without metformin (2 mmol/L) or biguanide (BIG 2 mmol/L) to determine the effect on the M1 and M2 phenotypes of macrophages, which was measured by flow cytometry for CD11c and CD206 expression. The colors denote the following: red, undifferentiated; blue, differentiated, untreated; orange, differentiated, metformin; green, differentiated, BIG. B, Macrophages were treated with/without metformin (2 mmol/L) or biguanide (BIG, 2 mmol/L) to determine the effect on activation in response to 100 ng/mL lipopolysaccharide (LPS), which was measured by studying CD69 and CD40 expression. Histograms are representative of n=4. The colors denote the following: red, unactivated; blue, activated, untreated; orange, activated, metformin; green, activated, BIG. CE, Macrophages were treated with/without metformin (Met) or BIG (2 mmol/L) to determine the effect of these drugs on IL-6 (C), IL-12p40 (D), and IL-10 (E) production (n=4).
Figure 5.
Figure 5.
Effect of long-term metformin treatment on nuclear factorB (NF-κB) signaling responses in hepatocytes. A and B, Primary hepatocytes were treated as in Figure 1 with metformin or propanediimidamide (PDI) at the doses indicated except that the treatment time was 24 h. For the last 15 min, cells were treated with 10 ng/mL tumor necrosis factor (TNF)-α. In addition to antibodies used elsewhere, phosphorylation of inhibitor of kappa B kinase (IKK)α/β was investigated using the phosphospecific antibodies indicated. After cell lysis, SDS-PAGE and immunoblotting were performed as in Figure 1. C and D, Hepatocytes from wild-type (WT) and AMPK double-knockout (KO) livers treated as in A or with doses of rotenone for 45 min before cell lysis, SDS-PAGE and immunoblotting. E, Primary hepatocytes were treated in the presence or absence of the agents shown. Cells were treated with/without metformin (2 mmol/L), C-C motif chemokine ligand (CCL)-11 (5 ng/mL), interleukin (IL)-2, IL-4, stromal cell–derived factor (SDF), and CCL22 (10 ng/mL) for 12 h, and glucose production was measured by GAGO (glucose [glucose oxidase]) assay as described in the Methods section of this article. ACC indicates acetyl-CoA carboxylase; AMPK, AMP-activated protein kinase; p-ACC, phospho–acetyl-CoA carboxylase; and pAMPK, phospho–AMP-activated protein kinase.

References

    1. Evans JM, Ogston SA, Emslie-Smith A, Morris AD. Risk of mortality and adverse cardiovascular outcomes in type 2 diabetes: a comparison of patients treated with sulfonylureas and metformin. Diabetologia. 2006;49:930–936. doi: 10.1007/s00125-006-0176-9.
    1. UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet. 1998;352:854–865.
    1. Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005;330:1304–1305. doi: 10.1136/bmj.38415.708634.F7.
    1. Rena G, Pearson ER, Sakamoto K. Molecular mechanism of action of metformin: old or new insights? Diabetologia. 2013;56:1898–1906. doi: 10.1007/s00125-013-2991-0.
    1. Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F. Cellular and molecular mechanisms of metformin: an overview. Clin Sci (Lond) 2012;122:253–270. doi: 10.1042/CS20110386.
    1. Logie L, Harthill J, Patel K, Bacon S, Hamilton DL, Macrae K, McDougall G, Wang HH, Xue L, Jiang H, Sakamoto K, Prescott AR, Rena G. Cellular responses to the metal-binding properties of metformin. Diabetes. 2012;61:1423–1433. doi: 10.2337/db11-0961.
    1. Repiščák P, Erhardt S, Rena G, Paterson MJ. Biomolecular mode of action of metformin in relation to its copper binding properties. Biochemistry. 2014;53:787–795. doi: 10.1021/bi401444n.
    1. Quan X, Uddin R, Heiskanen A, Parmvi M, Nilson K, Donolato M, Hansen MF, Rena G, Boisen A. The copper binding properties of metformin–QCM-D, XPS and nanobead agglomeration. Chem Commun (Camb) 2015;51:17313–17316. doi: 10.1039/c5cc04321b.
    1. Rena G, Pearson ER, Sakamoto K. Molecular action and pharmacogenetics of metformin: current understanding of an old drug. Diabetes Management. 2012;2:439–452.
    1. Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J. 2000;348 Pt 3:607–614.
    1. El-Mir MY, Nogueira V, Fontaine E, Avéret N, Rigoulet M, Leverve X. Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J Biol Chem. 2000;275:223–228.
    1. Madiraju AK, Erion DM, Rahimi Y, et al. Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature. 2014;510:542–546. doi: 10.1038/nature13270.
    1. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller DE. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167–1174. doi: 10.1172/JCI13505.
    1. Duca FA, Côté 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. Nat Med. 2015;21:506–511. doi: 10.1038/nm.3787.
    1. Foretz M, Hébrard S, Leclerc J, Zarrinpashneh E, Soty M, Mithieux G, Sakamoto K, Andreelli F, Viollet B. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest. 2010;120:2355–2369. doi: 10.1172/JCI40671.
    1. Miller RA, Chu Q, Xie J, Foretz M, Viollet B, Birnbaum MJ. Biguanides suppress hepatic glucagon signalling by decreasing production of cyclic AMP. Nature. 2013;494:256–260. doi: 10.1038/nature11808.
    1. Calvert JW, Gundewar S, Jha S, Greer JJ, Bestermann WH, Tian R, Lefer DJ. Acute metformin therapy confers cardioprotection against myocardial infarction via AMPK-eNOS-mediated signaling. Diabetes. 2008;57:696–705. doi: 10.2337/db07-1098.
    1. Gundewar S, Calvert JW, Jha S, Toedt-Pingel I, Ji SY, Nunez D, Ramachandran A, Anaya-Cisneros M, Tian R, Lefer DJ. Activation of AMP-activated protein kinase by metformin improves left ventricular function and survival in heart failure. Circ Res. 2009;104:403–411. doi: 10.1161/CIRCRESAHA.108.190918.
    1. Yin M, van der Horst IC, van Melle JP, Qian C, van Gilst WH, Silljé HH, de Boer RA. Metformin improves cardiac function in a nondiabetic rat model of post-MI heart failure. Am J Physiol Heart Circ Physiol. 2011;301:H459–H468. doi: 10.1152/ajpheart.00054.2011.
    1. Xu X, Lu Z, Fassett J, et al. Metformin protects against systolic overload-induced heart failure independent of AMP-activated protein kinase α2. Hypertension. 2014;63:723–728. doi: 10.1161/HYPERTENSIONAHA.113.02619.
    1. Cittadini A, Napoli R, Monti MG, Rea D, Longobardi S, Netti PA, Walser M, Samà M, Aimaretti G, Isgaard J, Saccà L. Metformin prevents the development of chronic heart failure in the SHHF rat model. Diabetes. 2012;61:944–953. doi: 10.2337/db11-1132.
    1. Sasaki H, Asanuma H, Fujita M, et al. Metformin prevents progression of heart failure in dogs: role of AMP-activated protein kinase. Circulation. 2009;119:2568–2577. doi: 10.1161/CIRCULATIONAHA.108.798561.
    1. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444:860–867. doi: 10.1038/nature05485.
    1. Pearson TA, Mensah GA, Alexander RW, Anderson JL, Cannon RO, 3rd, Criqui M, Fadl YY, Fortmann SP, Hong Y, Myers GL, Rifai N, Smith SC, Jr, Taubert K, Tracy RP, Vinicor F Centers for Disease Control and Prevention; American Heart Association. Markers of inflammation and cardiovascular disease: application to clinical and public health practice: A statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation. 2003;107:499–511.
    1. Isoda K, Young JL, Zirlik A, MacFarlane LA, Tsuboi N, Gerdes N, Schönbeck U, Libby P. Metformin inhibits proinflammatory responses and nuclear factor-kappaB in human vascular wall cells. Arterioscler Thromb Vasc Biol. 2006;26:611–617. doi: 10.1161/01.ATV.0000201938.78044.75.
    1. Woo SL, Xu H, Li H, et al. Metformin ameliorates hepatic steatosis and inflammation without altering adipose phenotype in diet-induced obesity. PLoS One. 2014;9:e91111. doi: 10.1371/journal.pone.0091111.
    1. Patel K, Foretz M, Marion A, et al. The LKB1-salt-inducible kinase pathway functions as a key gluconeogenic suppressor in the liver. Nat Commun. 2014;5:4535. doi: 10.1038/ncomms5535.
    1. Morris AD, Boyle DI, MacAlpine R, Emslie-Smith A, Jung RT, Newton RW, MacDonald TM. The diabetes audit and research in Tayside Scotland (DARTS) study: electronic record linkage to create a diabetes register. DARTS/MEMO Collaboration. BMJ. 1997;315:524–528.
    1. Zahorec R. Ratio of neutrophil to lymphocyte counts–rapid and simple parameter of systemic inflammation and stress in critically ill. Bratisl Lek Listy. 2001;102:5–14.
    1. Pinato DJ, Stavraka C, Flynn MJ, Forster MD, O’Cathail SM, Seckl MJ, Kristeleit RS, Olmos D, Turnbull SJ, Blagden SP. An inflammation based score can optimize the selection of patients with advanced cancer considered for early phase clinical trials. PLoS One. 2014;9:e83279. doi: 10.1371/journal.pone.0083279.
    1. Wang X, Zhang G, Jiang X, Zhu H, Lu Z, Xu L. Neutrophil to lymphocyte ratio in relation to risk of all-cause mortality and cardiovascular events among patients undergoing angiography or cardiac revascularization: a meta-analysis of observational studies. Atherosclerosis. 2014;234:206–213. doi: 10.1016/j.atherosclerosis.2014.03.003.
    1. Bhalla K, Hwang BJ, Dewi RE, Twaddel W, Goloubeva OG, Wong KK, Saxena NK, Biswal S, Girnun GD. Metformin prevents liver tumorigenesis by inhibiting pathways driving hepatic lipogenesis. Cancer Prev Res (Phila) 2012;5:544–552. doi: 10.1158/1940-6207.CAPR-11-0228.
    1. Clark K, Peggie M, Plater L, Sorcek RJ, Young ER, Madwed JB, Hough J, McIver EG, Cohen P. Novel cross-talk within the IKK family controls innate immunity. Biochem J. 2011;434:93–104. doi: 10.1042/BJ20101701.
    1. Grunfeld C, Dinarello CA, Feingold KR. Tumor necrosis factor-alpha, interleukin-1, and interferon alpha stimulate triglyceride synthesis in HepG2 cells. Metabolism. 1991;40:894–898.
    1. Sozio MS, Lu C, Zeng Y, Liangpunsakul S, Crabb DW. Activated AMPK inhibits PPAR-{alpha} and PPAR-{gamma} transcriptional activity in hepatoma cells. Am J Physiol Gastrointest Liver Physiol. 2011;301:G739–G747. doi: 10.1152/ajpgi.00432.2010.
    1. Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci. 2008;13:453–461.
    1. Bridges HR, Jones AJ, Pollak MN, Hirst J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem J. 2014;462:475–487. doi: 10.1042/BJ20140620.
    1. Zhang J, Clark K, Lawrence T, Peggie MW, Cohen P. An unexpected twist to the activation of IKKβ: TAK1 primes IKKβ for activation by autophosphorylation. Biochem J. 2014;461:531–537. doi: 10.1042/BJ20140444.
    1. Wong AK, Symon R, AlZadjali MA, Ang DS, Ogston S, Choy A, Petrie JR, Struthers AD, Lang CC. The effect of metformin on insulin resistance and exercise parameters in patients with heart failure. Eur J Heart Fail. 2012;14:1303–1310. doi: 10.1093/eurjhf/hfs106.
    1. Poggi M, Bastelica D, Gual P, Iglesias MA, Gremeaux T, Knauf C, Peiretti F, Verdier M, Juhan-Vague I, Tanti JF, Burcelin R, Alessi MC. C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet. Diabetologia. 2007;50:1267–1276. doi: 10.1007/s00125-007-0654-8.
    1. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117:175–184. doi: 10.1172/JCI29881.
    1. Goldberg RB, Temprosa MG, Mather KJ, Orchard TJ, Kitabchi AE, Watson KE Diabetes Prevention Program Research Group. Lifestyle and metformin interventions have a durable effect to lower CRP and tPA levels in the diabetes prevention program except in those who develop diabetes. Diabetes Care. 2014;37:2253–2260. doi: 10.2337/dc13-2471.
    1. Evia-Viscarra ML, Rodea-Montero ER, Apolinar-Jiménez E, Muñoz-Noriega N, García-Morales LM, Leaños-Pérez C, Figueroa-Barrón M, Sánchez-Fierros D, Reyes-García JG. The effects of metformin on inflammatory mediators in obese adolescents with insulin resistance: controlled randomized clinical trial. J Pediatr Endocrinol Metab. 2012;25:41–49. doi: 10.1515/jpem-2011-0469.
    1. Evans JM, Doney AS, AlZadjali MA, Ogston SA, Petrie JR, Morris AD, Struthers AD, Wong AK, Lang CC. Effect of Metformin on mortality in patients with heart failure and type 2 diabetes mellitus. Am J Cardiol. 2010;106:1006–1010. doi: 10.1016/j.amjcard.2010.05.031.
    1. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359:1577–1589. doi: 10.1056/NEJMoa0806470.
    1. Zee RY, Cook NR, Cheng S, Erlich HA, Lindpaintner K, Lee RT, Ridker PM. Threonine for alanine substitution in the eotaxin (CCL11) gene and the risk of incident myocardial infarction. Atherosclerosis. 2004;175:91–94. doi: 10.1016/j.atherosclerosis.2004.01.042.
    1. Villeda SA, Luo J, Mosher KI, et al. The ageing systemic milieu negatively regulates neurogenesis and cognitive function. Nature. 2011;477:90–94. doi: 10.1038/nature10357.
    1. Bannister CA, Holden SE, Jenkins-Jones S, Morgan CL, Halcox JP, Schernthaner G, Mukherjee J, Currie CJ. Can people with type 2 diabetes live longer than those without? A comparison of mortality in people initiated with metformin or sulphonylurea monotherapy and matched, non-diabetic controls. Diabetes Obes Metab. 2014;16:1165–1173. doi: 10.1111/dom.12354.
    1. Martin-Montalvo A, Mercken EM, Mitchell SJ, et al. Metformin improves healthspan and lifespan in mice. Nat Commun. 2013;4:2192. doi: 10.1038/ncomms3192.
    1. Hulme MA, Wasserfall CH, Atkinson MA, Brusko TM. Central role for interleukin-2 in type 1 diabetes. Diabetes. 2012;61:14–22. doi: 10.2337/db11-1213.
    1. Bischoff L, Alvarez S, Dai DL, Soukhatcheva G, Orban PC, Verchere CB. Cellular mechanisms of CCL22-mediated attenuation of autoimmune diabetes. J Immunol. 2015;194:3054–3064. doi: 10.4049/jimmunol.1400567.
    1. Cameron MJ, Arreaza GA, Zucker P, Chensue SW, Strieter RM, Chakrabarti S, Delovitch TL. IL-4 prevents insulitis and insulin-dependent diabetes mellitus in nonobese diabetic mice by potentiation of regulatory T helper-2 cell function. J Immunol. 1997;159:4686–4692.
    1. Yano T, Liu Z, Donovan J, Thomas MK, Habener JF. Stromal cell derived factor-1 (SDF-1)/CXCL12 attenuates diabetes in mice and promotes pancreatic beta-cell survival by activation of the prosurvival kinase Akt. Diabetes. 2007;56:2946–2957. doi: 10.2337/db07-0291.
    1. Ip B, Cilfone N, Zhu M, Kuchibhatla R, Azer M, McDonnell M, Apovian C, Lauffenburger D, Nikolajczyk B. An inflammatory T cell signature predicts obesity-associated type 2 diabetes (HUM3P.262). J. Immunol. 2015;194:121–122.
    1. Derakhshan R, Arababadi MK, Ahmadi Z, Karimabad MN, Salehabadi VA, Abedinzadeh M, Khorramdelazad H, Balaei P, Kennedy D, Hassanshahi G. Increased circulating levels of SDF-1 (CXCL12) in type 2 diabetic patients are correlated to disease state but are unrelated to polymorphism of the SDF-1β gene in the Iranian population. Inflammation. 2012;35:900–904. doi: 10.1007/s10753-011-9391-8.
    1. D’Agostino RB., Jr Propensity score methods for bias reduction in the comparison of a treatment to a non-randomized control group. Stat Med. 1998;17:2265–2281.
    1. Trelle S, Reichenbach S, Wandel S, Hildebrand P, Tschannen B, Villiger PM, Egger M, Jüni P. Cardiovascular safety of non-steroidal anti-inflammatory drugs: network meta-analysis. BMJ. 2011;342:c7086.

Source: PubMed

3
Se inscrever