Review Article | Volume: 12, Issue: 4, April, 2022

Effect of metformin treatment on inflammatory markers in type 2 diabetes mellitus—A systematic review and meta-analysis

Renuka Suvarna Revathi P. Shenoy M. Mukhyaprana Prabhu Guruprasad Kalthur Basavaraj S. Hadapad Varashree Bolar Suryakanth   

Open Access   

Published:  Apr 05, 2022

DOI: 10.7324/JAPS.2022.120401
Abstract

Background: Inflammatory markers have a crucial role in the development and pathogenesis of type 2 diabetes mellitus (T2DM). Therefore, an ideal treatment for T2DM should exert multidimensional beneficial effects for the management of diabetes. Metformin, being a first-line therapy for T2DM, has proved to have an excellent hypoglycemic effect, but the conclusion of its effect on inflammation is inconsistent. This study aims to evaluate the pooled effect of metformin on inflammatory markers in T2DM.

Methods: PubMed, CINHAL, and Scopus were searched systematically, and the references were further explored for eligible articles. 28 articles were extracted from 2,514 studies after eligibility screening based on the selection criteria. The data of inflammatory markers were then analyzed for meta-analysis in RevMan software.

Results: The result of the subgroup meta-analysis shows that C-reactive protein (CRP) and high sensitivity C-reactive protein proved to be statistically significant for metformin in the placebo compared group. However, interleukin-6 and adiponectin proved to be beneficial for the comparator group.

Conclusion: It is important to understand the validated effect of metformin on inflammatory markers in T2DM, which is possible by following an appropriate and universal assessment method with a uniform time and dose of metformin.

Systematic Review Registration: The protocol for this systematic review is registered with PROSPERO (CRD42020180403).


Keyword:     Metformin inflammatory markers metformin treatment type 2 diabetes mellitus


Citation:

Suvarna R, Shenoy RP, Prabhu M, Kalthur G, Hadapad BS, Varashree BS. Effect of metformin treatment on inflammatory markers in type 2 diabetes mellitus—A systematic review and meta-analysis. J Appl Pharm Sci, 2022; 12(04):001–011.

Copyright: © The Author(s). This is an open-access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

HTML Full Text

INTRODUCTION

Type 2 diabetes mellitus (T2DM), a metabolic disorder, is characterized by high blood sugar (Zubair and Ahmad, 2019), which arises due to insulin insufficiency and pancreatic cell destruction (Engelmann et al., 2016; Padilha et al., 2016). T2DM is a complex disease that poses a critical health problem worldwide (Chaudhury et al., 2017; Xie and Du, 2011; Zheng et al., 2018). Impaired insulin secretion due to pancreatic beta-cell destruction and insulin resistance are the main factors in the development and pathogenesis of type 2 diabetes (Pradhan and Ridker, 2002).

The research has been conducted to indicate that inflammation is a pathogenic element in the occurrence of hyperglycemia in T2DM (Sjöholm and Nyström, 2006). The association of inflammation in diabetes is characterized by the presence of an increased level of circulatory cytokines, chemokines, and acute phase proteins (Greevenbroek et al., 2013; Herder et al., 2009; Spranger et al., 2003). Even though the magnitude of these inflammatory markers in different peripheral tissue is unclear, it is known that an increased level of these markers will activate the innate immunity in T2DM due to the overproduction of free fatty acids (Eguchi and Nagai, 2017). Various factors have been associated with an inflammatory process in diabetes which may be common or tissue-specific. The action of these inflammatory mediators that induces insulin resistance in different tissue involves many metabolic signaling pathways like IkB kinase-b and c-jun N-terminal kinase. These pathways are involved in the pathogenesis of diabetes (Shoelson, 2006) and activate nuclear factor-kB, inducing the production of cytokines like tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-1β, and acute-phase proteins in tissues like liver and adipose and provoking insulin resistance in these tissues (Arkan et al., 2005). Cytokines such as TNF-α, IL-1β, and IL-6 play an important role in the development of T2DM. Inflammation is mainly induced by the accumulation of fatty acids in islets of the pancreas, leading to the activation of the IL system and the production of these cytokines and chemokines. Thus, it is critical to recognize the function of inflammation in the development and pathogenesis of T2DM to find a way for causative treatment. The advancement in the development of the new drug for many other diseases associated with inflammation provides a unique opportunity in the field of research that led to the expressive and fast performance of clinical trials (Coughlan et al., 2014; Donath, 2014; Dunmore and Brown, 2013; Esser et al., 2014; Tilg and Moschen, 2008). The primary purpose of diabetes treatment is to improve the quality of life and lifespan in comparison with healthy individuals, and the necessity for achieving this goal is to prevent the progression of diabetic complications (Knowler, 2005). Metformin is the first-line medication for T2DM, which controls hyperglycemia by decreasing glucose secretion from the liver and boosting insulin sensitivity (Foretz et al., 2019). Metformin activity is believed to intercede through the enactment of AMPK (Adenosine Monophosphate-activated protein kinase), a key controller of cell vitality homeostasis known to apply both antioxidant and anti-inflammatory impact (Pollack et al., 2016). The lone effect of metformin proved to have good management in control of inflammation, but it is essential to know the pooled effect of metformin on markers of inflammation in type 2 diabetes. Meta-analysis is an important technique for evaluating the comparative efficacy of different treatments. Therefore, an attempt is made to review the effect of metformin on inflammatory markers to examine the primary research and summarize the overall findings objectively. This review provides an updated view of the status of inflammatory markers in metformin-treated alone or with combination in T2DM patients. This study’s approach may validate the future implementation of drugs targeting multiple effects that immensely improve the quality of life in type 2 diabetes patients.


METHODS

We directed this meta-analysis utilizing the preferred reporting items of systematic review and meta-analysis (PRISMA) guidelines.

Search strategy

Eligible studies were searched in electronic databases such as Medline, Scopus, Web of Science, and CINAHL complete from 2000. The search terms “Metformin” AND “Inflammatory markers” OR “Inflammatory biomarkers” OR “Markers of inflammation” AND “Type 2 Diabetes Mellitus” were included to identify relevant studies. The search screening was also done using separate terms instead of inflammatory markers such as “IL-6,” “TNF-α,” and “CRP” to extract more studies. No limitations were made in the study language, and also the references of included studies were checked to prevent missing publications.

Inclusion and exclusion criteria

The inclusion criteria include all the studies that evaluated metformin’s effects on inflammatory markers in T2DM patients. The studies with Metformin and control or placebo or any other treatment or with combination treatment group were evaluated. Full-length publication studies reported with at least one biomarker outcome and published in the English language were included. The result should be reported as mean or median for inflammatory markers at pretreatment and posttreatment in both experimental and control groups. The intervention period of more than 4 weeks was only considered.

Studies were excluded if they had no comparison group, no intervention given, type 1 diabetes, and if experimental models were animals.

Data extraction and quality assessment

Data were extracted by three authors independently, and the findings were compiled. The fourth author reviewed the extracted data. Any disagreement regarding the extracted data was settled by the viewpoint of a fourth author if required. The method of data extraction was done using standard data extraction form, which included the title, name of author, publication year, study design, study duration, country, sample size, details of the intervention, any co-interventions, and outcome measure.

The quality assessment of the included studies was evaluated using the Cochrane collaboration modified tool. This tool is assessed based on randomization and allocation concealment, blinding of the participants and researchers, attrition bias, selective reporting, and other biases.

Outcome measures and data analysis

Metformin and placebo were the key groups studied to determine the precise effectiveness of metformin on inflammatory markers. The effectiveness of metformin was also compared to that of other comparator groups to see if there was a difference. Metformin’s efficacy was also tested in conjunction with other comparators to determine its efficacy both alone and in combination. The outcome measures include C-reactive protein (CRP), high sensitivity C-reactive protein (hs-CRP), TNF-α, IL-6, monocyte chemoattractant protein-1 (MCP-1), adiponectin, and intercellular adhesion molecule (ICAM-1). The pre- and post-treatment changes in the experimental and control/comparator groups were pooled to evaluate each outcome’s effects. The outcome measure was calculated as mean and standard deviation. The random-effect model was used to find the total effect which detects the variation between the studies. I² statistics was used to determine the study heterogeneity. Subgroup analyses were conducted to find the effect of different treatment modalities. The quantitative analysis of the data was done using the software Review Manager 5.2 (https://review-manager.software.informer.com/5.2/).


FINDINGS AND DISCUSSION

A total of 237 articles were obtained after the title and abstract screening from 1,510 articles filtered after duplicate removal. Subsequently, after the full-text screening of articles, 28 studies were found to be eligible for meta-analysis. The detailed procedure of study choice and screening is displayed in Figure 1. The baseline characteristic of included studies is depicted in Table 1.

Study characteristics

The research studies included in the review were published between 2000 and 2020. The study duration ranged from 4 to 52 weeks. Totally 2,975 subjects were involved in this study. The mean age in three studies (Carter et al., 2005; Chakraborty et al., 2011; Zhang et al., 2018) was not reported, in few studies, the average mean age was mentioned (Eriksson et al., 2007; Lund et al., 2008; Ragonesi et al., 2012; Schiapaccassa et al., 2019), and rest all the studies reported mean age in both groups. The eligible criteria for glycated hemoglobin are >6.5 (Eriksson et al., 2007; Hanefeld et al., 2011; Lund et al., 2008; Natali et al., 2004; Ragonesi et al., 2012; Zhang et al., 2018), >7.0 (Abdulkadir, 2012; Chakraborty et al., 2011; Derosa et al., 2008; Erem et al., 2014; Esteghamati et al., 2013; Jager et al., 2005, 2014; Mo et al., 2019; Pradhan and Ridker, 2002; Schiapaccassa et al., 2019; Tousoulis et al., 2011), >7.5 (Derosa et al., 2013) and >8.0 (Chu et al., 2002; Derosa et al., 2012; Li and Shen, 2019). The study population comprised T2DM patients with participants having several other criteria’s including overweight or obesity (Carter et al., 2005; Derosa et al., 2008, 2013; Schiapaccassa et al., 2019), with coronary artery disease (Derosa et al., 2012; Jager et al., 2005; Lund et al., 2008), newly diagnosed type 2 diabetes (Abdulkadir, 2012; Erem et al., 2014; Esteghamati et al., 2013; Li and Shen, 2019; Mo et al., 2019; Tousoulis et al., 2011; Zhang et al., 2018), hypertension (Chu et al., 2002; Jager et al., 2014; Natali et al., 2004; Ragonesi et al., 2012), female participants only(Mo et al., 2019; Schiapaccassa et al., 2019), and elderly patients above the age of 50 (Derosa et al., 2008; Kadoglou et al., 2010).

Study interventions

The different groups of interventions were used in the comparator group in different studies. The intervention groups in one study (Derosa et al., 2013) were assigned to receive metformin (dosage of 2,500 ± 500 mg) for 8 ± 2 months and then were randomly assigned, in addition to the previously determined metformin dose, 100 mg of sitagliptin in the comparator group. In contrast, the metformin group continued the predetermined dosage. The subjects in another study (Jager et al., 2005) were randomly assigned to receive either metformin or placebo in addition to the existing insulin therapy. In some of the studies (Derosa et al., 2008, 2012, 2013; Forst et al., 2012; Kadoglou et al., 2010; Tousoulis, 2019), the comparator group had a combination therapy of another drug along with metformin. In one study (Abdulkadir, 2012), the process and the dosage of interventions were not mentioned.

Risk of bias assessment

Figure 2 shows the Cochrane risk of bias in these randomized clinical trial studies. We assessed the overall quality of 28 studies. Two of the included studies did not state the random sequence generation, and four studies did not mention group allocation concealment. The performance and detection criteria had a high risk of bias for most of the studies, as clarity for blinding of participants in seventeen studies and blinding of outcome assessment in 20 studies is not clear. Twenty-six studies described the clear outcome and reporting data. Four articles were evaluated as high risk due to the poor quality of study design.

Figure 1. PRISMA stream chart for study choice.

[Click here to view]

Table 1. Baseline qualities of eligible studies.

[Click here to view]

Meta-analysis

The meta-analysis of sub-group based on intervention was conducted to determine the disparities in the studies.

C-reactive protein

A total of eight studies were analyzed for CRP as shown in Figure 3. There was no significant effect overall (SMD = −0.20, 95% CI = −0.72 to 0.31, p = 0.44, n = 1,167) but the four studies for the effect of metformin on CRP in metformin and placebo group proved to be significant (SMD = −0.73, 95% CI = −1.43 to −0.04, p = 0.04, n = 953), whereas the other four studies in metformin in comparison to the other comparator group did not show any significance (SMD = 0.40, 95% CI = −0.05 to 0.86, p = 0.08, n = 214).

High sensitivity C-reactive protein

Figure 4 depicts meta-analysis of hs-CRP. Four studies in comparison of metformin with placebo reported significance effect (SMD = −0.31, 95% CI = −0.60 to −0.01, p = 0.04, n = 560), and also hs-CRP showed significant effect (SMD = 0.27, 95% CI = 0.00 to 0.54, p = 0.05, n = 467) in metformin and comparator group in six studies. The overall effect showed to be non-significant (SMD = −0.03, 95% CI = −0.24 to 0.30, p = 0.84, n = 1,027).

Tumor necrosis factor-α

A total of 11 studies were analyzed to study the effect of metformin intervention on TNF-α in Figure 5, and the pooled effect proved to be non-significant (SMD = −0.01, 95% CI = −0.41 to 0.39, p = 0.96, n = 1,045). No significant effect was achieved in metformin and comparator group (SMD = −0.16, 95% CI = −0.55 to 0.22, p = 0.40, n = 590) for eight studies as well as in metformin and combination therapy group (SMD = 0.42, 95% CI = −0.61 to 1.45, p = 0.42, n = 455) for three studies.

Interleukin-6

A significant difference was observed among six studies for IL-6 in metformin and comparator group (SMD = 0.37, 95% CI = 0.17 to 0.57, p = 0.0004, n = 393) as shown in Figure 6 favoring comparator group.

Figure 2. Quality evaluation of the studies contained in the meta-analysis. Risk of bias graph for the studies (above) and lanes (below).

[Click here to view]

Figure 3. Forest plots for CRP level in comparison with metformin and control group in T2DM.

[Click here to view]

Figure 4. Forest plots for hs-CRP level in comparison with metformin and control group in T2DM.

[Click here to view]

Figure 5. Forest plots for TNF-α level in metformin and control group.

[Click here to view]

Monocyte chemoattractant protein-1

Only two studies were eligible for meta-analysis of MCP-1 which showed to be non-significant (SMD = −0.44, 95% CI = −0.90 to 0.02, p = 0.06, n = 75) in metformin and combination treatment group as shown in Figure 7.

Intercellular adhesion molecule-1

The forest plot in Figure 8 for three studies in metformin and placebo group did not provide any significant effect (SMD = −0.12, 95% CI = −0.29 to 0.04, p = 0.14, n = 758).

Adiponectin

Six studies were analyzed to assess the effect of metformin treatment on adiponectin as shown in Figure 9 and the overall effect mentioned as statistically significant (SMD = −0.40, 95% CI = −0.56 to −0.24, p < 0.001, n = 622). The beneficial effect was found towards comparator group in three studies (SMD = −0.41, 95% CI = −0.72 to −0.11, p = 0.008, n = 174) that analyzed for metformin and comparator group, the same beneficial effect was found for combination therapy (SMD = −0.40, 95% CI = −0.59 to −0.21, p < 0.001, n = 448) when compared to metformin in another group of three studies.

Figure 6. Forest plots for IL-6 level in metformin and comparator group.

[Click here to view]

Figure 7. Forest plots for MCP-1 level in metformin versus combination treatment group.

[Click here to view]

Figure 8. Forest plots for ICAM-1 level in metformin and control group.

[Click here to view]

Figure 9. Forest plots for Adiponectin level in comparison with comparator and metformin group.

[Click here to view]

Publication bias

The publication bias for this meta-analysis was assessed by performing a funnel plot for hs-CRP as a representative index for inflammatory markers. Based on the plots, there was an indication that there had been minimum publication bias in metformin effect on type 2 diabetes for hs-CRP. This was authenticated by performing Egger’s linear regression in EZR (hs-CRP: intercept: 0.18; standard error: 1.41; 95% CI: −1.30, 1.68; t = 2.75, z = −0.26, p = 0.78) (Fig. 10) (https://www.R-project.org/).

Figure 10. Funnel plot of included articles in the meta-analysis for hs-CRP. SE: standard error, MD: mean difference.

[Click here to view]


CONCLUSION

The sub-group analysis of CRP and hs-CRP in the metformin and placebo groups showed a favorable effect for metformin. CRP in the metformin and comparator group did not show any significance whereas hs-CRP did. IL-6 and adiponectin in metformin and comparator group mentioned to be favoring comparator group and also the adiponectin levels seem to be improved in combination therapy when compared to metformin alone proving better efficacy in inflammation control. TNF-α, MCP-1, and ICAM-1 showed no improvement in the metformin group. The combination of metformin with other drugs is effective in controlling inflammation in type 2 diabetes. In this way, metformin has an advantage and can be used in tandem with other drugs in treating and management of diabetes. In the compromised studies, the span of the treatment period ranged from 4 weeks to 12 months and the metformin dose from 0.5 to 3.0 g/day. Because of the low number of the included studies, the ideal dose and the length of treatment were hard to determine.

The relation between inflammation, hyperglycemia, and complications of diabetes is well manifested now. Since low-grade inflammation is a predictor in the progression of diabetes and its complications, an increased level of CRP may be the significant risk marker in the pathogenesis of T2DM (Duncan et al., 2003). The evidence says that some metabolic factors such as hyperglycemia and free fatty acid may provoke CRP production by macrophages and endothelial cells (Mugabo et al., 2010), and increased production of CRP may be also due to increased IL-6 and TNF-α production that trigger inflammation in different tissues (Dehghan et al., 2007). Additionally, CRP has a role in impairing nitric oxide production, which leads to endothelial dysfunction (Stehouwer, 1987). IL-6 is associated with T2DM as well as impaired glucose tolerance specifying its role in the development of T2DM (Saxena et al., 2013). IL-6 has its contribution to the pathology and physiology of diabetes by its association with β-cell function and insulin-signaling pathways (Fève and Bastard, 2009), and also IL-6 triggers the CRP production (Pickup, 2004). TNF-α is a cytokine that is mainly delivered by monocytes and macrophages and has an action on insulin resistance in peripheral tissue (Ruan et al., 2002) as well as insulin secretion (Hotamisligil, 1999). Adiponectin inhibits the production and action of TNF- α, which may influence IL-6 and CRP production. Therefore, adiponectin may affect CRP levels in plasma and adipose tissue by modulating the inflammatory cascades. TNF-α development and action are inhibited by adiponectin, which can affect the production of IL-6 and CRP. As a result of inflammatory pathway modulation, adiponectin can also affect CRP levels and other inflammatory markers in plasma and adipose tissue (Schulze et al., 2004).

These inflammatory markers play a direct role in impairing insulin signaling pathways which contribute to insulin resistance in diabetes. However, the anti-inflammatory action of various medications is partial and conflicting, most likely because of deficient normalization of metabolic dysregulation or because diabetes-related aggravation is multifactorial but is not restricted to hyperglycemia. Improved diabetes prevention and treatment modalities could benefit from a greater understanding of the inflammatory basis for diabetes, which could involve innovative targeted therapies in addition to existing pharmacologic and lifestyle strategies. In this meta-analysis, metformin therapy did not show a true effect on inflammatory markers in T2DM patients. It was difficult to assess the positive effect of metformin which may be due to short treatment duration and different comparator groups. Therefore, further randomized, placebo-controlled clinical trials are required to confirm the metformin effect on inflammatory markers in T2DM patients.

Even though this systematic review gains the knowledge of metformin on inflammatory markers; it has few limitations. First, few of the trials had smaller sample sizes and shorter study duration. Second, wellbeing status, lifestyle, and essential oral anti-diabetic treatment were distinctive among the subjects and might be a significant source of heterogeneity. Third, the control group used in the review were not similar drugs which again accounts for heterogeneity. Fourth, though comprehensive information was separated for statistics investigation, studies included in this meta-analysis contained different ethnic populations and countries.

To find a more sustainable effect of metformin on inflammatory markers, future studies ought to build up a uniform assessment technique of inflammatory markers and explore the ideal dose and duration of metformin therapy along with better combination therapy in diabetes. In light of the prevalence of T2DM among Indians and the fact that metformin is the first-line therapy, it is important to control this disease effectively. This can be done by focusing on different mechanisms of metformin treatment, such as inflammatory markers in T2DM. Since inflammation is an additional risk factor in diabetes, the methodology of treatment towards this field will be gainful and accommodating in the management of T2DM.


LIST OF ABBREVIATIONS

hs-CRP, high sensitivity C-reactive protein; IL-6, interleukin-6; ICAM-1, intercellular adhesion molecule; MCP-1, monocyte chemoattractant protein-1; T2DM, type 2 diabetes mellitus; TNF-α, tumor necrosis factor-alpha.


AUTHORS CONTRIBUTION

All authors contributed to this review manuscript equally. Data were extracted and compiled with the aid of Renuka Suvarna, Revathi P. Shenoy, and M. Mukhyaprana Prabhu. Guruprasad Kalthur was able to review the extracted data and any discrepancies in the data extraction were further discussed with all the authors. Basavaraj S. Hadapad and Varashree B. S. collaborated to review the study’s methods, as well as the results and discussion of the manuscript. All the included authors of this study presented an equal contribution in data analysis and complete manuscript review.


CONFLICT OF INTEREST

The authors proclaim no conflict of interest.


FUNDING

This review does not have financial support from any source.


ETHICAL APPROVAL

Not applicable.


REFERENCES

Abdulkadir A. Comparative effects of glibenclamide and metformin on c-reactive protein and oxidant/antioxidant status in patients with type II diabetes mellitus. SQU Med J, 2012; 12:55–61. CrossRef

Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, Wynshaw-Boris A, Poli G, Olefsky J, Karin M. IKK-β links inflammation to obesity-induced insulin resistance. Nat Med, 2005; 11(2):191–8. CrossRef

Bulcao C, Ribeiro-Filho FF, Sanudo A, Ferreira SG. Effects of simvastatin and metformin on inflammation and insulin resistance in individuals with mild metabolic syndrome. Am J Cardiovasc Drugs, 2007; 7(3):219–24. CrossRef

Caballero AE, Delgado A, Aguilar-Salinas CA, Herrera AN, Castillo JL, Cabrera T, Gomez-Perez FJ, Rull JA. The differential effects of metformin on markers of endothelial activation and inflammation in subjects with impaired glucose tolerance: a placebo-controlled, randomized clinical trial. J Clin Endocrinol Metab, 2004; 89(8):3943–8. CrossRef

Carter AM, Bennett CE, Bostock JA, Grant PJ. Metformin reduces C–reactive protein but not complement factor C3 in overweight patients with type 2 diabetes mellitus. Diabetes Med, 2005; 22(9):1282–4. CrossRef

Chakraborty A, Chowdhury S, Bhattacharyya M. Effect of metformin on oxidative stress, nitrosative stress and inflammatory biomarkers in type 2 diabetes patients. Diabetes Res Clin Pract, 2011; 93(1):56–62. CrossRef

Chaudhury A, Duvoor C, Reddy Dendi VS, Kraleti S, Chada A, Ravilla R, Marco A, Shekhawat NS, Montales MT, Kuriakose K, Sasapu A. Clinical review of antidiabetic drugs: implications for type 2 diabetes mellitus management. Front Endocrinol, 2017; 8:6. CrossRef

Chu NV, Kong AP, Kim DD, Armstrong D, Baxi S, Deutsch R, Caulfield M, Mudaliar SR, Reitz R, Henry RR, Reaven PD. Differential effects of metformin and troglitazone on cardiovascular risk factors in patients with type 2 diabetes. Diabetes Care, 2002; 25(3):542–9. CrossRef

Coughlan KA, Valentine RJ, Ruderman NB, Saha AK. AMPK activation: a therapeutic target for type 2 diabetes? Diabetes Metab Syndr Obes, 2014; 7:241. CrossRef

Dehghan A, Van Hoek M, Sijbrands EJ, Stijnen T, Hofman A, Witteman JC. Risk of type 2 diabetes attributable to C-reactive protein and other risk factors. Diabetes Care, 2007; 30(10):2695–9. CrossRef

Derosa G, Salvadeo SA, D’Angelo A, Fogari E, Ragonesi PD, Ciccarelli L, Piccinni MN, Ferrari I, Gravina A, Maffioli P, Cicero AF. Rosiglitazone therapy improves insulin resistance parameters in overweight and obese diabetic patients intolerant to metformin. Arch Med Res, 2008; 39(4):412–9. CrossRef

Derosa G, Franzetti IG, Querci F, Carbone A, Ciccarelli L, Piccinni MN, Fogari E, Maffioli P. Exenatide plus metformin compared with metformin alone on β-cell function in patients with type 2 diabetes. Diabet Med, 2012; 29(12):1515–23. CrossRef

Derosa G, Carbone A, D’Angelo A, Querci F, Fogari E, Cicero AF, Maffioli P. Variations in inflammatory biomarkers following the addition of sitagliptin in patients with type 2 diabetes not controlled with metformin. Intern Med, 2013; 52(19):2179–87. CrossRef

Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov, 2014; 13(6):465–76. CrossRef

Duncan BB, Schmidt MI, Pankow JS, Ballantyne CM, Couper D, Vigo A, Hoogeveen R, Folsom AR, Heiss G. Low-grade systemic inflammation and the development of type 2 diabetes: the atherosclerosis risk in communities study. Diabetes, 2003; 52(7):1799–805. CrossRef

Dunmore SJ, Brown JE. The role of adipokines in b-cell failure of type 2 diabetes. J Endocrinol, 2013; 216(1):37–45. CrossRef

Eguchi K, Nagai R. Islet inflammation in type 2 diabetes and physiology. J Clin Invest, 2017; 127(1):14–23. CrossRef

Engelmann J, Manuwald U, Rubach C, Kugler J, Birkenfeld AL, Hanefeld M, Rothe U. Determinants of mortality in patients with type 2 diabetes: a review. Rev Endocr Metab Disord, 2016; 17(1):129–37. CrossRef

Erem C?, Ozbas HM, Nuhoglu I, Deger OR, Civan NA, Ersoz HO. Comparison of effects of gliclazide, metformin and pioglitazone monotherapies on glycemic control and cardiovascular risk factors in patients with newly diagnosed uncontrolled type 2 diabetes mellitus. Exp Clin Endocrinol Diabetes, 2014; 122(05):295–302. CrossRef

Eriksson A, Attvall S, Bonnier M, Eriksson JW, Rosander B, Karlsson FA. Short-term effects of metformin in type 2 diabetes. Diabetes Obes Metab, 2007; 9(4):483–9. CrossRef

Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract, 2014; 105(2):141–50. CrossRef

Esteghamati A, Eskandari D, Mirmiranpour H, Noshad S, Mousavizadeh M, Hedayati M, Nakhjavani M. Effects of metformin on markers of oxidative stress and antioxidant reserve in patients with newly diagnosed type 2 diabetes: a randomized clinical trial. Clin Nutr, 2013; 32(2):179–85. CrossRef

Everett BM, Cook NR, Rifai N, Ridker PM. Effects of initiating insulin and metformin on glycemic control and inflammatory biomarkers among patients with type 2 diabetes: the LANCET randomized trial. JAMA, 2009; 302(11):1186–94. CrossRef

Fève B, Bastard JP. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat Rev Endocrinol, 2009; 5(6):305–11 CrossRef

Foretz M, Guigas B, Viollet B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat Rev Endocrinol, 2019; 15(10):569–89. CrossRef

Forst T, Michelson G, Ratter F, Weber MM, Anders S, Mitry M, Wilhelm B, Pfützner A. Addition of liraglutide in patients with type 2 diabetes well controlled on metformin monotherapy improves several markers of vascular function. Diabet Med, 2012; 29(9):1115–8. CrossRef

Greevenbroek MM, Schalkwijk CG, Stehouwer CD. Obesity-associated low-grade inflammation in type 2 diabetes mellitus: causes and consequences. Neth J Med, 2013; 71(4):174–87.

Hanefeld M, Pfützner A, Forst T, Kleine I, Fuchs W. Double-blind, randomized, multicentre, and active comparator controlled investigation of the effect of pioglitazone, metformin, and the combination of both on cardiovascular risk in patients with type 2 diabetes receiving stable basal insulin therapy: the PIOCOMB study. Cardiovasc Diabetol, 2011; 10(1):1–9. CrossRef

Herder C, Brunner EJ, Rathmann W, Strassburger K, Tabák AG, Schloot NC, Witte DR. Elevated levels of the anti-inflammatory interleukin-1 receptor antagonist precede the onset of type 2 diabetes: the Whitehall II study. Diabetes Care, 2009; 32(3):421–3. CrossRef

Hotamisligil GS. The role of TNFα and TNF receptors in obesity and insulin resistance. J Intern Med, 1999; 245(6):621–5. CrossRef

Jager DJ, Kooy A, Lehert PH, Bets D, Wulffele MG, Teerlink T, Scheffer PG, Schalkwijk CG, Donker AJ, Stehouwer CD. Effects of short-term treatment with metformin on markers of endothelial function and inflammatory activity in type 2 diabetes mellitus: a randomized, placebo-controlled trial. J Intern Med, 2005; 257(1):100–9.

Jager DJ, Kooy A, Schalkwijk C, Van Der Kolk J, Lehert P, Bets D, Wulffele MG, Donker AJ, Stehouwer CD. Long-term effects of metformin on endothelial function in type 2 diabetes: a randomized controlled trial. J Intern Med, 2014; 275(1):59–70. CrossRef

Kadoglou NP, Tsanikidis H, Kapelouzou A, Vrabas I, Vitta I, Karayannacos PE, Liapis CD, Sailer N. Effects of rosiglitazone and metformin treatment on apelin, visfatin, and ghrelin levels in patients with type 2 diabetes mellitus. Metabolism, 2010; 59(3):373–9. CrossRef

Kim HJ, Kang ES, Kim DJ, Kim SH, Ahn CW, Cha BS, Nam M, Chung CH, Lee KW, Nam CM, Lee HC. Effects of rosiglitazone and metformin on inflammatory markers and adipokines: decrease in interleukin-18 is an independent factor for the improvement of homeostasis model assessment-beta in type 2 diabetes mellitus. Clin Endocrinol, 2007; 66(2):282–9 CrossRef

Kiyici S, Ersoy C, Kaderli A, Fazlioglu M, Budak F, Duran C, Gul OO, Sigirli D, Baran I, Tuncel E, Erturk E. Effect of rosiglitazone, metformin and medical nutrition treatment on arterial stiffness, serum MMP-9 and MCP-1 levels in drug naive type 2 diabetic patients. Diabetes Res Clin Pract, 2009; 86(1):44–50. CrossRef

Knowler W. Prevention of type 2 diabetes with troglitazone in the Diabetes Prevention Program. Diabetes, 2005; 54(4):1150–6. CrossRef

Li J, Shen X. Effect of rosiglitazone on inflammatory cytokines and oxidative stress after intensive insulin therapy in patients with newly diagnosed type 2 diabetes. Diabetol Metab Syndr, 2019; 11(1):1–9. CrossRef

Lund SS, Tarnow L, Stehouwer CD, Schalkwijk CG, Teerlink T, Gram J, Winther K, Frandsen M, Smidt UM, Pedersen O, Parving HH. Impact of metformin versus repaglinide on non-glycaemic cardiovascular risk markers related to inflammation and endothelial dysfunction in non-obese patients with type 2 diabetes. Eur J Endocrinol, 2008; 158(5):631–42. CrossRef

Mo D, Liu S, Ma H, Tian H, Yu H, Zhang X, Tong N, Liao J, Ren Y. Effects of acarbose and metformin on the inflammatory state in newly diagnosed type 2 diabetes patients: a one-year randomized clinical study. Drug Des Devel Ther, 2019; 13:2769. CrossRef

Mugabo Y, Li L, Renier G. The connection between C-reactive protein (CRP) and diabetic vasculopathy. Focus on preclinical findings. Curr Diabetes Rev, 2010; 6(1):27–34. CrossRef

Natali A, Baldeweg S, Toschi E, Capaldo B, Barbaro D, Gastaldelli A, Yudkin JS, Ferrannini E. Vascular effects of improving metabolic control with metformin or rosiglitazone in type 2 diabetes. Diabetes Care, 2004; 27(6):1349–57. CrossRef

Padilha K, Venturini G, de Farias Pires T, Horimoto AR, Malagrino PA, Gois TC, Kiers B, Oliveira CM, de Oliveira Alvim R, Blatt C, Krieger JE. Serum metabolomics profile of type 2 diabetes mellitus in a Brazilian rural population. Metabolomics, 2016; 12(10):1–1. CrossRef

Pickup JC. Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care, 2004; 27(3):813–23. CrossRef

Pollack RM, Donath MY, LeRoith D, Leibowitz G. Anti-inflammatory agents in the treatment of diabetes and its vascular complications. Diabetes Care, 2016; 39:S244–52. CrossRef

Pradhan AD, Ridker PM. Do atherosclerosis and type 2 diabetes share a common inflammatory basis? Eur Heart J, 2002; 23(11):831–4. CrossRef

Ragonesi P. Vildagliptin added to metformin on b -cell function after a euglycemic hyperinsulinemic and hyperglycemic clamp in type 2 diabetes patients. Diabetes Technol Ther, 2012; 14(6):475–84. CrossRef

Ruan H, Miles PD, Ladd CM, Ross K, Golub TR, Olefsky JM, Lodish HF. Profiling gene transcription in vivo reveals adipose tissue as an immediate target of tumor necrosis factor-α: implications for insulin resistance. Diabetes, 2002; 51(11):3176–88. CrossRef

Saxena M, Srivastava N, Banerjee M. Association of IL-6, TNF-α and IL-10 gene polymorphisms with type 2 diabetes mellitus. Mol Biol Rep, 2013; 40(11):6271–9. CrossRef

Schiapaccassa A, Maranhão PA, de Souza MD, Panazzolo DG, Neto JF, Bouskela E, Kraemer-Aguiar LG. 30-days effects of vildagliptin on vascular function, plasma viscosity, inflammation, oxidative stress, and intestinal peptides on drug-naïve women with diabetes and obesity: a randomized head-to-head metformin-controlled study. Diabetol Metab Syndr, 2019; 11(1):1–9. CrossRef

Schulze MB, Rimm EB, Shai I, Rifai N, Hu FB. Relationship between adiponectin and glycemic control, blood lipids, and inflammatory markers in men with type 2 diabetes. Diabetes Care, 2004; 27(7):1680–7. CrossRef

Shoelson S. Inflammation and insulin resistance find the latest version?: review series inflammation and insulin resistance. J Clin Invest, 2006; 116:1793–801. CrossRef

Sjöholm Å, Nyström T. Inflammation and the etiology of type 2 diabetes. Diabetes Metab Res Rev, 2006; 22(1):4–10. CrossRef

Spranger J, Kroke A, Möhlig M, Hoffmann K, Bergmann MM, Ristow M, Boeing H, Pfeiffer AF. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes, 2003; 52(3):812–7. CrossRef

Stehouwer CD. Dysfunction, and chronic low-grade inflammation in diabetes. Diabetes, 1987; 51:1157–65. CrossRef

Tilg H, Moschen AR. Inflammatory mechanisms in the regulation of insulin resistance. Mol Med, 2008; 14(3):222–31. CrossRef

Tousoulis D, Koniari K, Antoniades C, Papageorgiou N, Miliou A, Noutsou M, Nikolopoulou A, Marinou K, Stefanadi E, Siasos G, Charakida M. Combined effects of atorvastatin and metformin on glucose-induced variations of inflammatory process in patients with diabetes mellitus. Int J Cardiol Cardiovasc, 2011; 149(1):46–9. CrossRef

Tousoulis D. Combined effects of atorvastatin and metformin on glucose-induced variations of inflammatory process in patients with diabetes mellitus. Int J Cardiol Cardiovasc, 2019; 149(1):46–49. CrossRef

Xie W, Du L. Diabetes is an inflammatory disease: evidence from traditional Chinese medicines. Diabetes Obes Metab, 2011; 13(4):289–301. CrossRef

Zhang WQ, Tian Y, Chen XM, Wang LF, Chen CC, Qiu CM. Liraglutide ameliorates beta-cell function, alleviates oxidative stress and inhibits low grade inflammation in young patients with new-onset type 2 diabetes. Diabetol Metab Syndr, 2018; 10(1):1–8. CrossRef

Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol, 2018; 14(2):88–98. CrossRef

Zubair M, Ahmad J. Role of growth factors and cytokines in diabetic foot ulcer healing: a detailed review. Rev Endocrine Metab Disord, 2019; 20(2):207–17. CrossRef

Reference

Abdulkadir A. Comparative effects of glibenclamide and metformin on c-reactive protein and oxidant/antioxidant status in patients with type II diabetes mellitus. SQU Med J, 2012; 12:55-61. https://doi.org/10.12816/0003088

Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, Wynshaw-Boris A, Poli G, Olefsky J, Karin M. IKK-β links inflammation to obesity-induced insulin resistance. Nat Med, 2005; 11(2):191-8. https://doi.org/10.1038/nm1185

Bulcao C, Ribeiro-Filho FF, Sanudo A, Ferreira SG. Effects of simvastatin and metformin on inflammation and insulin resistance in individuals with mild metabolic syndrome. Am J Cardiovasc Drugs, 2007; 7(3):219-24. https://doi.org/10.2165/00129784-200707030-00007

Caballero AE, Delgado A, Aguilar-Salinas CA, Herrera AN, Castillo JL, Cabrera T, Gomez-Perez FJ, Rull JA. The differential effects of metformin on markers of endothelial activation and inflammation in subjects with impaired glucose tolerance: a placebo-controlled, randomized clinical trial. J Clin Endocrinol Metab, 2004; 89(8):3943-8. https://doi.org/10.1210/jc.2004-0019

Carter AM, Bennett CE, Bostock JA, Grant PJ. Metformin reduces C-reactive protein but not complement factor C3 in overweight patients with type 2 diabetes mellitus. Diabetes Med, 2005; 22(9):1282-4. https://doi.org/10.1111/j.1464-5491.2005.01632.x

Chakraborty A, Chowdhury S, Bhattacharyya M. Effect of metformin on oxidative stress, nitrosative stress and inflammatory biomarkers in type 2 diabetes patients. Diabetes Res Clin Pract, 2011; 93(1):56-62. https://doi.org/10.1016/j.diabres.2010.11.030

Chaudhury A, Duvoor C, Reddy Dendi VS, Kraleti S, Chada A, Ravilla R, Marco A, Shekhawat NS, Montales MT, Kuriakose K, Sasapu A. Clinical review of antidiabetic drugs: implications for type 2 diabetes mellitus management. Front Endocrinol, 2017; 8:6. https://doi.org/10.3389/fendo.2017.00006

Chu NV, Kong AP, Kim DD, Armstrong D, Baxi S, Deutsch R, Caulfield M, Mudaliar SR, Reitz R, Henry RR, Reaven PD. Differential effects of metformin and troglitazone on cardiovascular risk factors in patients with type 2 diabetes. Diabetes Care, 2002; 25(3):542-9. https://doi.org/10.2337/diacare.25.3.542

Coughlan KA, Valentine RJ, Ruderman NB, Saha AK. AMPK activation: a therapeutic target for type 2 diabetes? Diabetes Metab Syndr Obes, 2014; 7:241. https://doi.org/10.2147/DMSO.S43731

Dehghan A, Van Hoek M, Sijbrands EJ, Stijnen T, Hofman A, Witteman JC. Risk of type 2 diabetes attributable to C-reactive protein and other risk factors. Diabetes Care, 2007; 30(10):2695-9. https://doi.org/10.2337/dc07-0348

Derosa G, Salvadeo SA, D'Angelo A, Fogari E, Ragonesi PD, Ciccarelli L, Piccinni MN, Ferrari I, Gravina A, Maffioli P, Cicero AF. Rosiglitazone therapy improves insulin resistance parameters in overweight and obese diabetic patients intolerant to metformin. Arch Med Res, 2008; 39(4):412-9. https://doi.org/10.1016/j.arcmed.2007.12.009

Derosa G, Franzetti IG, Querci F, Carbone A, Ciccarelli L, Piccinni MN, Fogari E, Maffioli P. Exenatide plus metformin compared with metformin alone on β-cell function in patients with type 2 diabetes. Diabet Med, 2012; 29(12):1515-23. https://doi.org/10.1111/j.1464-5491.2012.03699.x

Derosa G, Carbone A, D'Angelo A, Querci F, Fogari E, Cicero AF, Maffioli P. Variations in inflammatory biomarkers following the addition of sitagliptin in patients with type 2 diabetes not controlled with metformin. Intern Med, 2013; 52(19):2179-87. https://doi.org/10.2169/internalmedicine.52.8175

Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov, 2014; 13(6):465-76. https://doi.org/10.1038/nrd4275

Duncan BB, Schmidt MI, Pankow JS, Ballantyne CM, Couper D, Vigo A, Hoogeveen R, Folsom AR, Heiss G. Low-grade systemic inflammation and the development of type 2 diabetes: the atherosclerosis risk in communities study. Diabetes, 2003; 52(7):1799-805. https://doi.org/10.2337/diabetes.52.7.1799

Dunmore SJ, Brown JE. The role of adipokines in b-cell failure of type 2 diabetes. J Endocrinol, 2013; 216(1):37-45. https://doi.org/10.1530/JOE-12-0278

Eguchi K, Nagai R. Islet inflammation in type 2 diabetes and physiology. J Clin Invest, 2017; 127(1):14-23. https://doi.org/10.1172/JCI88877

Engelmann J, Manuwald U, Rubach C, Kugler J, Birkenfeld AL, Hanefeld M, Rothe U. Determinants of mortality in patients with type 2 diabetes: a review. Rev Endocr Metab Disord, 2016; 17(1):129-37. https://doi.org/10.1007/s11154-016-9349-0

Erem C?, Ozbas HM, Nuhoglu I, Deger OR, Civan NA, Ersoz HO. Comparison of effects of gliclazide, metformin and pioglitazone monotherapies on glycemic control and cardiovascular risk factors in patients with newly diagnosed uncontrolled type 2 diabetes mellitus. Exp Clin Endocrinol Diabetes, 2014; 122(05):295-302. https://doi.org/10.1055/s-0034-1370989

Eriksson A, Attvall S, Bonnier M, Eriksson JW, Rosander B, Karlsson FA. Short-term effects of metformin in type 2 diabetes. Diabetes Obes Metab, 2007; 9(4):483-9. https://doi.org/10.1111/j.1463-1326.2006.00624.x

Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract, 2014; 105(2):141-50. https://doi.org/10.1016/j.diabres.2014.04.006

Esteghamati A, Eskandari D, Mirmiranpour H, Noshad S, Mousavizadeh M, Hedayati M, Nakhjavani M. Effects of metformin on markers of oxidative stress and antioxidant reserve in patients with newly diagnosed type 2 diabetes: a randomized clinical trial. Clin Nutr, 2013; 32(2):179-85. https://doi.org/10.1016/j.clnu.2012.08.006

Everett BM, Cook NR, Rifai N, Ridker PM. Effects of initiating insulin and metformin on glycemic control and inflammatory biomarkers among patients with type 2 diabetes: the LANCET randomized trial. JAMA, 2009; 302(11):1186-94. https://doi.org/10.1001/jama.2009.1347

Fève B, Bastard JP. The role of interleukins in insulin resistance and type 2 diabetes mellitus. Nat Rev Endocrinol, 2009; 5(6):305-11 https://doi.org/10.1038/nrendo.2009.62

Foretz M, Guigas B, Viollet B. Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat Rev Endocrinol, 2019; 15(10):569-89. https://doi.org/10.1038/s41574-019-0242-2

Forst T, Michelson G, Ratter F, Weber MM, Anders S, Mitry M, Wilhelm B, Pfützner A. Addition of liraglutide in patients with type 2 diabetes well controlled on metformin monotherapy improves several markers of vascular function. Diabet Med, 2012; 29(9):1115-8. https://doi.org/10.1111/j.1464-5491.2012.03589.x

Greevenbroek MM, Schalkwijk CG, Stehouwer CD. Obesity-associated low-grade inflammation in type 2 diabetes mellitus: causes and consequences. Neth J Med, 2013; 71(4):174-87.

Hanefeld M, Pfützner A, Forst T, Kleine I, Fuchs W. Double-blind, randomized, multicentre, and active comparator controlled investigation of the effect of pioglitazone, metformin, and the combination of both on cardiovascular risk in patients with type 2 diabetes receiving stable basal insulin therapy: the PIOCOMB study. Cardiovasc Diabetol, 2011; 10(1):1-9. https://doi.org/10.1186/1475-2840-10-65

Herder C, Brunner EJ, Rathmann W, Strassburger K, Tabák AG, Schloot NC, Witte DR. Elevated levels of the anti-inflammatory interleukin-1 receptor antagonist precede the onset of type 2 diabetes: the Whitehall II study. Diabetes Care, 2009; 32(3):421-3. https://doi.org/10.2337/dc08-1161

Hotamisligil GS. The role of TNFα and TNF receptors in obesity and insulin resistance. J Intern Med, 1999; 245(6):621-5. https://doi.org/10.1046/j.1365-2796.1999.00490.x

Jager DJ, Kooy A, Lehert PH, Bets D, Wulffele MG, Teerlink T, Scheffer PG, Schalkwijk CG, Donker AJ, Stehouwer CD. Effects of short-term treatment with metformin on markers of endothelial function and inflammatory activity in type 2 diabetes mellitus: a randomized, placebo-controlled trial. J Intern Med, 2005; 257(1):100-9. Jager DJ, Kooy A, Schalkwijk C, Van Der Kolk J, Lehert P, Bets D, Wulffele MG, Donker AJ, Stehouwer CD. Long-term effects of metformin on endothelial function in type 2 diabetes: a randomized controlled trial. J Intern Med, 2014; 275(1):59-70. https://doi.org/10.1111/j.1365-2796.2004.01420.x

Kadoglou NP, Tsanikidis H, Kapelouzou A, Vrabas I, Vitta I, Karayannacos PE, Liapis CD, Sailer N. Effects of rosiglitazone and metformin treatment on apelin, visfatin, and ghrelin levels in patients with type 2 diabetes mellitus. Metabolism, 2010; 59(3):373-9. https://doi.org/10.1016/j.metabol.2009.08.005

Kim HJ, Kang ES, Kim DJ, Kim SH, Ahn CW, Cha BS, Nam M, Chung CH, Lee KW, Nam CM, Lee HC. Effects of rosiglitazone and metformin on inflammatory markers and adipokines: decrease in interleukin-18 is an independent factor for the improvement of homeostasis model assessment-beta in type 2 diabetes mellitus. Clin Endocrinol, 2007; 66(2):282-9 https://doi.org/10.1111/j.1365-2265.2006.02723.x

Kiyici S, Ersoy C, Kaderli A, Fazlioglu M, Budak F, Duran C, Gul OO, Sigirli D, Baran I, Tuncel E, Erturk E. Effect of rosiglitazone, metformin and medical nutrition treatment on arterial stiffness, serum MMP-9 and MCP-1 levels in drug naive type 2 diabetic patients. Diabetes Res Clin Pract, 2009; 86(1):44-50. https://doi.org/10.1016/j.diabres.2009.07.004

Knowler W. Prevention of type 2 diabetes with troglitazone in the Diabetes Prevention Program. Diabetes, 2005; 54(4):1150-6. https://doi.org/10.2337/diabetes.54.4.1150

Li J, Shen X. Effect of rosiglitazone on inflammatory cytokines and oxidative stress after intensive insulin therapy in patients with newly diagnosed type 2 diabetes. Diabetol Metab Syndr, 2019; 11(1):1-9. https://doi.org/10.1186/s13098-019-0432-z

Lund SS, Tarnow L, Stehouwer CD, Schalkwijk CG, Teerlink T, Gram J, Winther K, Frandsen M, Smidt UM, Pedersen O, Parving HH. Impact of metformin versus repaglinide on non-glycaemic cardiovascular risk markers related to inflammation and endothelial dysfunction in non-obese patients with type 2 diabetes. Eur J Endocrinol, 2008; 158(5):631-42. https://doi.org/10.1530/EJE-07-0815

Mo D, Liu S, Ma H, Tian H, Yu H, Zhang X, Tong N, Liao J, Ren Y. Effects of acarbose and metformin on the inflammatory state in newly diagnosed type 2 diabetes patients: a one-year randomized clinical study. Drug Des Devel Ther, 2019; 13:2769. https://doi.org/10.2147/DDDT.S208327

Mugabo Y, Li L, Renier G. The connection between C-reactive protein (CRP) and diabetic vasculopathy. Focus on preclinical findings. Curr Diabetes Rev, 2010; 6(1):27-34. https://doi.org/10.2174/157339910790442628

Natali A, Baldeweg S, Toschi E, Capaldo B, Barbaro D, Gastaldelli A, Yudkin JS, Ferrannini E. Vascular effects of improving metabolic control with metformin or rosiglitazone in type 2 diabetes. Diabetes Care, 2004; 27(6):1349-57. https://doi.org/10.2337/diacare.27.6.1349

Padilha K, Venturini G, de Farias Pires T, Horimoto AR, Malagrino PA, Gois TC, Kiers B, Oliveira CM, de Oliveira Alvim R, Blatt C, Krieger JE. Serum metabolomics profile of type 2 diabetes mellitus in a Brazilian rural population. Metabolomics, 2016; 12(10):1-1. https://doi.org/10.1007/s11306-016-1107-5

Pickup JC. Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care, 2004; 27(3):813-23. https://doi.org/10.2337/diacare.27.3.813

Pollack RM, Donath MY, LeRoith D, Leibowitz G. Anti-inflammatory agents in the treatment of diabetes and its vascular complications. Diabetes Care, 2016; 39:S244-52. https://doi.org/10.2337/dcS15-3015

Pradhan AD, Ridker PM. Do atherosclerosis and type 2 diabetes share a common inflammatory basis? Eur Heart J, 2002; 23(11):831-4. https://doi.org/10.1053/euhj.2001.3052

Ragonesi P. Vildagliptin added to metformin on b -cell function after a euglycemic hyperinsulinemic and hyperglycemic clamp in type 2 diabetes patients. Diabetes Technol Ther, 2012; 14(6):475-84. https://doi.org/10.1089/dia.2011.0278

Ruan H, Miles PD, Ladd CM, Ross K, Golub TR, Olefsky JM, Lodish HF. Profiling gene transcription in vivo reveals adipose tissue as an immediate target of tumor necrosis factor-α: implications for insulin resistance. Diabetes, 2002; 51(11):3176-88. https://doi.org/10.2337/diabetes.51.11.3176

Saxena M, Srivastava N, Banerjee M. Association of IL-6, TNF-α and IL-10 gene polymorphisms with type 2 diabetes mellitus. Mol Biol Rep, 2013; 40(11):6271-9. https://doi.org/10.1007/s11033-013-2739-4

Schiapaccassa A, Maranhão PA, de Souza MD, Panazzolo DG, Neto JF, Bouskela E, Kraemer-Aguiar LG. 30-days effects of vildagliptin on vascular function, plasma viscosity, inflammation, oxidative stress, and intestinal peptides on drug-naïve women with diabetes and obesity: a randomized head-to-head metformin-controlled study. Diabetol Metab Syndr, 2019; 11(1):1-9. https://doi.org/10.1186/s13098-019-0466-2

Schulze MB, Rimm EB, Shai I, Rifai N, Hu FB. Relationship between adiponectin and glycemic control, blood lipids, and inflammatory markers in men with type 2 diabetes. Diabetes Care, 2004; 27(7):1680-7. https://doi.org/10.2337/diacare.27.7.1680

Shoelson S. Inflammation and insulin resistance find the latest version : review series inflammation and insulin resistance. J Clin Invest, 2006; 116:1793-801. https://doi.org/10.1172/JCI29069

Sjöholm Å, Nyström T. Inflammation and the etiology of type 2 diabetes. Diabetes Metab Res Rev, 2006; 22(1):4-10. https://doi.org/10.1002/dmrr.568

Spranger J, Kroke A, Möhlig M, Hoffmann K, Bergmann MM, Ristow M, Boeing H, Pfeiffer AF. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)- Potsdam Study. Diabetes, 2003; 52(3):812-7. https://doi.org/10.2337/diabetes.52.3.812

Stehouwer CD. Dysfunction, and chronic low-grade inflammation in diabetes. Diabetes, 1987; 51:1157-65. https://doi.org/10.2337/diabetes.51.4.1157

Tilg H, Moschen AR. Inflammatory mechanisms in the regulation of insulin resistance. Mol Med, 2008; 14(3):222-31. https://doi.org/10.2119/2007-00119.Tilg

Tousoulis D, Koniari K, Antoniades C, Papageorgiou N, Miliou A, Noutsou M, Nikolopoulou A, Marinou K, Stefanadi E, Siasos G, Charakida M. Combined effects of atorvastatin and metformin on glucose-induced variations of inflammatory process in patients with diabetes mellitus. Int J Cardiol Cardiovasc, 2011; 149(1):46-9. https://doi.org/10.1016/j.ijcard.2009.11.038

Tousoulis D. Combined effects of atorvastatin and metformin on glucose-induced variations of inflammatory process in patients with diabetes mellitus. Int J Cardiol Cardiovasc, 2019; 149(1):46-49. https://doi.org/10.1016/j.ijcard.2009.11.038

Xie W, Du L. Diabetes is an inflammatory disease: evidence from traditional Chinese medicines. Diabetes Obes Metab, 2011; 13(4):289-301. https://doi.org/10.1111/j.1463-1326.2010.01336.x

Zhang WQ, Tian Y, Chen XM, Wang LF, Chen CC, Qiu CM. Liraglutide ameliorates beta-cell function, alleviates oxidative stress and inhibits low grade inflammation in young patients with new-onset type 2 diabetes. Diabetol Metab Syndr, 2018; 10(1):1-8. https://doi.org/10.1186/s13098-018-0392-8

Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol, 2018; 14(2):88-98. https://doi.org/10.1038/nrendo.2017.151

Zubair M, Ahmad J. Role of growth factors and cytokines in diabetic foot ulcer healing: a detailed review. Rev Endocrine Metab Disord, 2019; 20(2):207-17. https://doi.org/10.1007/s11154-019-09492-1

Article Metrics
92 Views 93 Downloads 185 Total

Year

Month

Related Search

By author names