Back to Journals » Diabetes, Metabolic Syndrome and Obesity » Volume 12

Abdominal Fat Is Directly Associated With Inflammation In Persons With Type-2 Diabetes Regardless Of Glycemic Control – A Jordanian Study

Authors Bawadi H , Katkhouda R, Tayyem R , Kerkadi A, Bou Raad S , Subih H 

Received 4 May 2019

Accepted for publication 16 September 2019

Published 22 November 2019 Volume 2019:12 Pages 2411—2417

DOI https://doi.org/10.2147/DMSO.S214426

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 4

Editor who approved publication: Dr Konstantinos Tziomalos



Hiba Bawadi,1 Rami Katkhouda,2 Reema Tayyem,3 Abdelhamid Kerkadi,1 Samira Bou Raad,4 Hadil Subih5

1Human Nutrition Department, College of Health Sciences, QU Health, Qatar University, Doha, Qatar; 2Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, Jordan; 3Department of Clinical Nutrition and Dietetics, University of Jordan, Amman, Jordan; 4Department of Nutrition and Food Science, American University of Science and Technology, Beirut, Lebanon; 5Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, Jordan

Correspondence: Hiba Bawadi
Human Nutrition Department, College of Health Sciences, QU-Health, Qatar University, Building I06, University Street, PO Box 2713, Doha, Qatar
Tel +97444034801
Email [email protected]

Background and aim: Systemic inflammation is related to the progression of complications associated with diabetes. This study aimed to investigate the association between general and abdominal obesity and inflammation in patients with type-2 diabetes with or without glycemic control.
Methods: A total of 198 men (n=73) and women (n=125) diagnosed with type 2 diabetes participated in this study. General obesity markers, body mass index (BMI), and abdominal fat were assessed. Circulating concentrations of glycated hemoglobin (HbA1C), C-reactive protein (CRP), and serum interleukin-6 (IL-6) were determined. Poor glycemic control and good glycemic control were defined as having fasting HbA1C concentrations ≥7% and <7%, respectively. Multivariate adjusted analysis of covariance was used to determine the relation between BMI and abdominal fat and markers of inflammation in patients with good and poor glycemic control.
Results: Patients in <7% HbA1C category, those with high abdominal fat had ≈262% higher CRP and ≈30.6% higher IL-6 compared to those with low abdominal fat (p˂0.05). Patients in ≥7% HbA1C category, those with high abdominal fat had ≈41.4% higher CRP and ≈33.9% higher IL-6 compared to those with low abdominal fat (p˂0.05). Abdominal fat was directly related to CRP (p˂0.023) and IL-6 (p˂0.002) concentrations in both groups of type-2 diabetic patients with <7% and ≥7% HbA1C. In patients with ≥7% HbA1C, BMI was directly related to CRP (p˂0.02) and IL-6 (p˂0.047). Whereas in patients with <7% HbA1C, BMI was not associated with CRP or IL-6 concentrations.
Conclusion: High level of abdominal fat is associated with systemic inflammation in type-2 diabetes regardless of glycemic control. Abdominal fat is a better predictor (determinant) of inflammation than BMI in patients with type-2 diabetes with or without glycemic control.

Keywords: BMI, C-reactive protein, diabetes, IL-6, inflammation, obesity

Introduction

Type 2 diabetes mellitus (T2D) is a progressive, common disease characterized by hyperglycemia resulting from defects in insulin action. T2D is considered one of the costliest chronic diseases with strong association with many chronic diseases, such as renal failure, atherosclerotic vascular disease, hypertension, and dyslipidemia.13

Systemic inflammation is the continuous phenomenon of the immune response which can promote tissue damage.4 Chronic systemic inflammation arises due to the persistence of a pro-inflammatory substance. This environment could be the result of a pro-inflammatory stimulus or inadequate determination of the inflammatory response.5,6 The relationship between inflammation and diabetes is complicated. Intermediary role of inflammation has been established in many studies in the occurrence of T2D and its long-term complications.7 Several connections have been proposed. One suggests that inflammation plays a vital role in diabetes pathogenesis through the induction of β cell apoptosis in T2D by the activity of reactive oxygen species and inflammatory cytokines.811 A large body of research supports the finding that systemic inflammation is associated with insulin resistance.1214 Experiments conducted on animals and studies in humans displayed a strong association between low-grade inflammation and increased insulin resistance,15,16 leading to elevated glucose concentrations.

Diabetes complications such as cardiovascular diseases, are often characterized by diabetes-induced systemic inflammation.17,18 Elevated inflammation characterized by increased concentration of IL-6 was significantly related to progression of microvascular complications of diabetes.19 Elevated inflammatory markers such as CRP and IL-6 are inversely related to nerve conduction velocities and directly related to peripheral neuropathy, even in non-diabetic persons.20 Despite the continuous work dedicated to gaining an in-depth understanding of the relation between inflammation and diabetes pathogenesis and progression, the role of glycemic control in inflammation is not clear in Jordanian population. Therefore, this study aimed to investigate the association between general and abdominal obesity and inflammatory biomarkers in patients with or without glycemic control.

Methods

This study was conducted in accordance with the Declaration of Helsinki. The protocol of this study was approved by the Institutional Review Board at Jordan University of Science and Technology (JUST). Patients were recruited from the outpatient endocrinology unit at King Abdullah University Hospital, JUST Health Center, and private endocrinology clinics in the north of Jordan.

Initial screening included 1500 patients diagnosed with T2D. Due to the multiple co-variation nature of the relationship of interest, about 87% of the initially screened patients were excluded from the study. The final study sample comprised 198 patients (73 male and 125 female) diagnosed with type 2 diabetes. Patients were excluded if they met one of the following criteria: 1) diagnosed with T2D for less than one year; 2) also diagnosed with either rheumatoid arthritis, cancer, diabetic foot, kidney diseases, or any chronic or acute inflammatory disease; 3) had a recent major or minor surgery; 4) on non-steroidal anti-inflammatory drugs for less than two weeks prior to the blood sample collection; 5) women on oral contraceptives; and 6) women who were pregnant. All 198 patients completed the study. All subjects signed informed consent after all study procedures were explained.

Blood Specimen Collection And Analysis

A 10-mL blood sample was collected by venipuncture by a registered nurse. For measurement of HbA1c, blood samples were collected in ethylenediaminetetraacetic acid tubes. Then HbA1C was measured in whole blood using the immune-inhibition test for the quantitative determination of glycosylated hemoglobin (Beckman Coulter AU analyzers). For the measurement of CRP and IL-6, blood was collected in Z-Clot activator tubes. Blood was allowed to clot before centrifugation for 15 mins at 1000×g. Aliquots of serum were stored at ≤ −22°C in sterile small tubes prior to biochemical assay. CRP and IL-6 concentrations were analyzed within a month after collection of blood. The immuno-turbidimetric test was used to determine CRP levels (Beckman Coulter AU analyzers). IL-6 was measured using a human immunoassay kit from R&D SYSTEMS through sandwich-type enzyme-linked immunosorbent assay. An Absorbance Microplate Reader was used to measure the optical density of IL-6 (BioTek ELx800).

Anthropometrics And Body Composition

Weight and height were measured following World Health Organization (WHO) procedures.21 Body weight was measured with the individuals wearing no shoes and light clothing. Height was measured using a measuring rod (Seca, Germany). Body mass index (BMI) was calculated using the ratio of weight (kilograms) to the square of height (meters) (kg/m2). The WHO BMI cutoff points were used to classify patients into different body weight categories.21

Patients’ abdominal fat percentage was determined using the bioelectrical impedance technique (TANITA, BC-418). The Segmental Body Composition Analyzer (TANITA, BC-418) used in this study was previously validated against hydro-densitometry in the assessment of body composition in healthy young adults.22 The cutoff points for abdominal fat percentage were gender-specific, based on which patients were classified into low, average, and high abdominal fat.23

Statistical Analysis

Statistical Package for Social Sciences (SPSS, version 19, Chicago: SPSS Inc.) software was used in data analysis. Characteristics of subjects’ variables were described using frequency distribution for categorical variables and mean and standard deviation for continuous variables. Patients were classified as having good glycemic control if they had <7% HbA1C. If a patient had ≥7% HbA1C, that patient was categorized as having poor glycemic control. Multivariate-adjusted analysis of variance (MANOVA) was used to examine the differences in mean serum levels of CRP and IL-6 in patients with good and poor glycemic control. Analysis was adjusted for age, gender, use of lipid-lowering drugs, and diabetes duration. Least significant difference post-hoc MANOVA was conducted to determine which two means differ within the BMI and abdominal fat categories. A p-value of <0.05 was considered the cutoff level for statistical significance.

Results

Table 1 shows characteristics of the study participants. The majority of the participants were >50 years old. More than 2/3 of the sample (75.8%) were nonsmokers. The glycemic control was poor for 66.2% of the patients. Lipid lowering drugs were used by 19.7% of the participants. 7.1% of the participants had a normal BMI. 27.8% were overweight, and 65.1% were obese. Only 7.5% of the participants had abdominal fat at low level, while 68.7% presented with normal level, and 20.2% had high fat level. We performed an additional analysis on gender and age differences regarding inflation. We found no significant difference between men and women (except CRP in poor glycemic group) and between older and younger persons regarding CRP and IL-6 concentrations in both glycemic groups (data not shown). However, the analysis was adjusted for these variables.

Table 1 Participants’ Characteristics – Glycemic Control (N=198)

Table 2 shows the association between BMI and abdominal fat and inflammatory biomarkers in patients with controlled glycemia. Patients with good glycemic control (<7% HbA1C) with high abdominal fat had ~262% higher CRP and ~30.6% higher IL-6 compared to those with low abdominal fat (p˂0.05). Among patients with good glycemic control, no significant association was found between BMI and inflammatory markers (CRP or IL-6).

Table 2 Relation Between BMI And Abdominal Fat And Inflammatory Biomarkers In Patients With Type-2 Diabetes With Good Glycemic Control (<7% HbA1C)a

Table 3 shows the relation between BMI and abdominal fat and inflammatory biomarkers in patients with uncontrolled glycemia. Patients with poor glycemic control (≥7% HbA1C), with high abdominal fat had ≈41.4% higher CRP and ≈33.9% higher IL-6 compared to those with low abdominal fat (p˂0.05). Patients with poor glycemic control with obesity grade III had ≈206% higher CRP and ≈50.2% higher IL-6 compared to those with normal BMI.

Table 3 Relation Between BMI And Abdominal Fat And Biomarkers Of Inflammation In Patients With Type-2 Diabetes With Poor Glycemic Control (≥7% HbA1C)a

Discussion

An interesting finding of the current study is that the inflammatory markers studied (CRP, IL-6) were higher among patients with higher BMI; however, this finding was observed only among patients with poor glycemic control. In other words, obese patients with controlled diabetes (HbA1c ˂7%) had less systemic inflammation despite having diabetes and obesity. This might be explained by the impact of a consistent state of hyperglycemia (HbA1c ˃7%), which may lead to up-regulation of acute phase inflammatory markers such as amyloid A3 (SAA3) and pro-inflammatory cytokines such as IL-6, by adipose tissue under the stimulation of high blood glucose.24 Elevation of IL-6 may in turn stimulate the production of CRP by the hepatocytes.25,26 Ray et al stated that elevated levels of glucose resulting from obesity can create stress in pancreatic islets, adipose tissue, liver, and muscle.27 Consequently, there is a surge in local production and release of cytokines and chemokines, such as IL-6 and TNF-α. These changes enhance further production and release of cytokines and chemokines.27 Another mechanism through which high glucose increases the complications associated with diabetes is production of reactive oxygen species (ROS). These ROS suppress insulin production by inhibiting the transcription factors of insulin in β-cells of the pancreas. ROS cause endothelial dysfunction and induce synthesis of chemokines such as MCP-1, ICAM 1, and IL-1. This further upregulates the ROS production, leading to diabetes induced atherosclerosis.28,29 Therefore, when serum glucose is controlled, even in the context of obesity, there could be a downplay of stress which in turn decreases the previously mentioned heightened release of cytokines and chemokines.

A few studies investigated the relation between obesity and inflammation among patients with diabetes according to their glycemic control. Mohan et al conducted a study on Asian Indian patients with diabetes, and demonstrated higher CRP levels among participants with increased HbA1c and tertiles of body fat.30 Zaciragic et al reported a direct relation between serum CRP concentrations and BMI in T2D.31 The same relationship was documented by Rawson et al. Other obesity markers such as total fat percentage were also investigated in the literature in relation to inflammation among patients with diabetes.32 The findings of our study are in line with previously published work that showed an association between circulatory markers of chronic inflammation (CRP, IL-6) and body fat percentage.3336 Other studies failed to find an association between inflammatory response and obesity among patients with diabetes, however.37 Investigators interpret this controversy by the nonlinearity that may exist between the cytokine levels and BMI of diabetic patients.

The scenario was different when examining abdominal fat percentage. Regardless of the glycemic control of the patients, abdominal fat percentage was associated with an increased level of serum inflammatory biomarkers (CRP, IL-6). These results were supported by findings from numerous studies that depicted a positive correlation between CRP and abdominal body fat.31,33,36 Rexrode et al and Park et al also reported a very clear positive trend concerning CRP levels.38,39 A study was conducted on elderly type 2 diabetic patients by Pedersen et al in 2003.40 They found that high circulatory inflammatory cytokines (IL-6 and TNF-α) were associated with increased abdominal fat mass in patients with good and poor glycemic control. The exact explanation of the relation between abdominal fat and inflammatory response is not yet fully understood. In vivo and in vitro studies detected amplified IL-6 production by subcutaneous adipose cells with defects in insulin action.41 The distinctive role of intraperitoneal adipocytes as an active gland results in the secretion of several hormones and several adipocytokines.42 Once these cytokines are secreted by adipocytes, this results in a boost in the CRP production via hepatocytes.25,26

It is worth mentioning that although absolute HbA1c levels were used as indicators of glycemic control, glucose variability was not measured in this study. Hoffman et al reported on 17 adolescents with type 1 diabetes, stating that increased glucose variability is associated with increased inflammation in this group,43 suggesting that pathogenesis in type-1 diabetes is different from T2D. It would be interesting to determine whether patients in the non-controlled glycemia group had increased instances of intermittent hyperglycemia or whether it was sustained hyperglycemia. Animal studies have suggested that intermittent hyperglycemia has a more marked effect than sustained hyperglycemia in inducing oxidative stress and increasing inflammation;44 however, studies in human adults have been less conclusive. Many studies in human adults have found no association between glucose variability and consequences of inflammation, such as microvascular or macrovascular complications, arterial stiffness, and carotid intimal thickness.43 On the other hand, other studies stated that fluctuations in glucose, especially during post prandial periods, heightened the triggering effect on oxidative stress compared to sustained hyperglycemia.45

The major strength of this study is the stringent procedure we used for the selection of the participants to rule out any acute response to inflammation. Due to the cross-sectional nature of the study, results should not be viewed in terms of cause and effect relationship. Due to the small sample size in this study, results may not be applicable to the population at large in Jordan. For the same reason, we were unable to determine the relationship between body composition and inflammation using various hyperglycemic levels. Future studies should use more accurate body composition measurement methods such as DEXA rather than bioelectrical impedance methods that were used in this study.

In conclusion, the findings of the current study indicate that abdominal fat is related to increased inflammation regardless of glycemic control in Jordanian sample. General obesity is associated with systemic inflammation only among patients with poor glycemic control. Patients should maintain HbA1C concentrations below 7% to reduce the glucose induced inflammatory markers, as this has been linked to complications associated with diabetes. Longitudinal studies are needed to decipher the complex relationship between body composition and inflammation and its overall impact on progression of diabetes.

Acknowledgments

The authors would like to thank the Deanship of Research at JUST for providing financial support for this project. Authors would also like to thank Ms. Fawzieh Hammad for her help during the data collection. The collaboration of the volunteers was also greatly appreciated. Thanks are due to Dr Vijay Ganji for his efforts in revising this manuscript. The publication of this article was funded by the Qatar National Library

Author Contributions

All authors contributed toward data analysis, drafting and revising the paper, gave final approval of the version to be published and agree to be accountable for all aspects of the work.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Ajlouni K, Khader Y, Batieha A, Ajlouni H, El-Khateeb M. An increase in prevalence of diabetes mellitus in Jordan over 10 years. J Diabetes Complications. 2008;22(5):317–324. doi:10.1016/j.jdiacomp.2007.01.004

2. Khader YS, Batieha A, El-Khateeb M, Al Omari M, Ajlouni K. Prevalence of dyslipidemia and its associated factors among Jordanian adults. J Clin Lipidol. 2010;4(1):53–58. doi:10.1016/j.jacl.2009.12.004

3. Mubarak FM, Froelicher ES, Jaddou HY, Ajlouni KM. Hypertension among 1000 patients with type 2 diabetes attending a national diabetes center in Jordan. Ann Saudi Med. 2008;28(5):346–351. doi:10.5144/0256-4947.2008.346

4. Gabay C. Interleukin-6 and chronic inflammation. Arthritis Res Ther. 2006;8(2):S3. doi:10.1186/ar1917

5. Baniyash M. Chronic inflammation, immunosuppression and cancer: new insights and outlook. Semin Cancer Biol. 2006;16(1):80–88. doi:10.1016/j.semcancer.2005.12.002

6. Porta C, Larghi P, Rimoldi M, et al. Cellular and molecular pathways linking inflammation and cancer. Immunobiology. 2009;214(9–10):761–777. doi:10.1016/j.imbio.2009.06.014

7. Wang X, Bao W, Liu J, et al. Inflammatory markers and risk of type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2013;36(1):166–175. doi:10.2337/dc12-0702

8. Donath M, Ehses J, Maedler K, et al. Mechanisms of β-cell death in type 2 diabetes. Diabetes. 2005;54(2):108–113. doi:10.2337/diabetes.54.suppl_2.S108

9. Maedler K, Sergeev P, Ris F, et al. Glucose-induced β cell production of IL-1β contributes to glucotoxicity in human pancreatic islets. J Clin Invest. 2002;110(6):851–860. doi:10.1172/JCI15318

10. Meier J. Beta cell mass in diabetes: a realistic therapeutic target? Diabetologia. 2008;51(5):703–713. doi:10.1007/s00125-008-0936-9

11. Robertson R. Chronic oxidative stress as a cenetral mechanism for glucose toxicity in pancreatic islet beta cells in diabetes. J Biol Chem. 2004;279(41):42351–42354. doi:10.1074/jbc.R400019200

12. Freeman D, Norrie J, Caslake M, et al. C-reactive protein is an independent predictor of risk for the development of diabetes in the West of Scotland Coronary Prevention Study. Diabetes. 2002;51(5):1596–1600. doi:10.2337/diabetes.51.5.1596

13. Osborn O, Olefsky J. The cellular and signaling networks linking the immune system and metabolism in disease. Nat Med. 2012;18(3):363–374. doi:10.1038/nm.2627

14. Tanigaki K, Vongpatanasin W, Barrera J, et al. C-reactive protein causes insulin resistance in mice through Fcγ receptor IIB–mediated inhibition of skeletal muscle glucose delivery. Diabetes. 2013;62(3):721–731. doi:10.2337/db12-0133

15. Heilbronn L, Campbell L. Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity. Curr Pharm Des. 2008;14(12):1225–1230. doi:10.2174/138161208784246153

16. Schenk S, Saberi M, Olefsky J. Insulin sensitivity: modulation by nutrients and inflammation. J Clin Invest. 2008;118(9):2992–3002. doi:10.1172/JCI34260

17. Dandona P, Aljada A, Chaudhuri A, Bandyopadhyay A. The potential influence of inflammation and insulin resistance on the pathogenesis and treatment of atherosclerosis-related complications in type 2 diabetes. J Clin Endocrinol Metab. 2003;88(6):2422–2429. doi:10.1210/jc.2003-030178

18. Lu H, Raptis M, Black E, Stan M, Amar S, Graves D. Influence of diabetes on the exacerbation of an inflammatory response in cardiovascular tissue. Endocrinology. 2004;145(11):4934–4939. doi:10.1210/en.2004-0737

19. Wegner M, Araszkiewicz A, Piorunska-Stolzmann M, Wierusz-Wysocka B, Zozulinska-Ziolkiewicz D. Association between IL-6 concentration and diabetes-related variables in DM1 patients with and without microvascular complications. Inflammation. 2013;36(3):723–728. doi:10.1007/s10753-013-9598-y

20. Rashad N, El-Shabrawy R, Sabry H, Fathy H, Said D, Yousef M. Interleukin-6 and hs-CRP as early diagnostic biomarkers for obesity-related peripheral polyneuropathy in non-diabetic patients. Egypt J Immunol. 2018;25(2):153–165.

21. World Health Organization. Obesity: Preventing and Managing the Global Epidemic. World Health Organization; 2000.

22. Rutherford W, Diemer G, Scott E. Comparison of Bioelectrical Impedance and Skinfolds with Hydrodensitometry in the Assessment of Body Composition in Healthy Young Adults. J Res. 2011;6(2):56–60.

23. Gallagher D, Heymsfield S, Heo M, Jebb S, Murgatroyd P, Sakamoto Y. Healthy percentage body fat ranges: an approach for developing guidelines based on body mass index. Am J Clin Nutr. 2000;72(3):694–701. doi:10.1093/ajcn/72.3.694

24. Lin Y, Rajala M, Berger J, Moller D, Barzilai N, Scherer P. Hyperglycemia-induced production of acute phase reactants in adipose tissue. J Biol Chem. 2001;276(45):42077–42083. doi:10.1074/jbc.M107101200

25. Bulló M, Casas-Agustench P, Amigó-Correig P, Aranceta J, Salas-Salvadó J. Inflammation, obesity and comorbidities: the role of diet. Public Health Nutr. 2007;10(10A):1164–1172. doi:10.1017/S1368980007000663

26. Yudkin J, Kumari M, Humphries S, Mohamed-Ali V. Inflammation, obesity, stress and coronary heart disease: is interleukin-6 the link? Atherosclerosis. 2000;148(2):209–214. doi:10.1016/s0021-9150(99)00463-3

27. Ray I, Mahata S, De R. Obesity: an immunometabolic perspective. Front Endocrinol (Lausanne). 2016;7(157). doi:10.3389/fendo.2016.00089

28. Yuan T, Yang T, Chen H, et al. New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol. 2018;20:247–260.

29. Halim M, Halim A. The effects of inflammation, aging and oxidative stress on the pathogenesis of diabetes mellitus (type 2 diabetes). Diabet Metab Syndr. 2019;13:1165–1172. doi:10.1016/j.dsx.2019.01.040

30. Mohan V, Deepa R, Velmurugan K, Premalatha G. Association of C‐reactive protein with body fat, diabetes and coronary artery disease in Asian Indians: the Chennai Urban Rural Epidemiology Study (CURES‐6). Diabet Med. 2005;22(7):863–870. doi:10.1111/j.1464-5491.2005.01541.x

31. Začiragić A, Hadžović-Džuvo A, Valjevac A, Avdagić N, Mulabegović N. Serum C-reactive protein concentration and measures of adiposity in patients with type 2 diabetes mellitus. Bosn J Basic Med Sci. 2007;7(4):322–327. doi:10.17305/bjbms.2007.3021

32. Rawson E, Freedson P, Osganian S, Matthews C, Reed G, Ockene I. Body mass index, but not physical activity, is associated with C-reactive protein. Med Sci Sports Exerc. 2003;35(7):1160–1166. doi:10.1249/01.MSS.0000074565.79230.AB

33. Axelsson J, Rashid Qureshi A, Suliman M, et al. Truncal fat mass as a contributor to inflammation in end-stage renal disease. Am J Clin Nutr. 2004;80(5):1222–1229. doi:10.1093/ajcn/80.5.1222

34. Festa A, D’Agostino R, Williams K, et al. The relation of body fat mass and distribution to markers of chronic inflammation. Int J Obes. 2001;25(10):1407–1415. doi:10.1038/sj.ijo.0801792

35. Vega GL, Adams-Huet B, Peshock R, Willett D, Shah B, Grundy S. Influence of body fat content and distribution on variation in metabolic risk. J Clin Endocrinol Metab. 2006;91(11):4459–4466. doi:10.1210/jc.2006-0814

36. Hammett C, Oxenham H, Baldi J, et al. Effect of six months’ exercise training on C-reactive protein levels in healthy elderly subjects. J Am Coll Cardiol. 2004;44(12):2411–2413. doi:10.1016/j.jacc.2004.09.030

37. Nadeem A, Naveed A, Hussain M, Raza S. Correlation of inflammatory markers with type 2 Diabetes Mellitus in Pakistani patients. J Postgrad Med Inst. 2013;27(3):367–373.

38. Park H, Park J, Yu R. Relationship of obesity and visceral adiposity with serum concentrations of CRP, TNF-α and IL-6. Diabetes Res Clin Pract. 2005;69(1):29–35. doi:10.1016/j.diabres.2004.11.007

39. Rexrode K, Pradhan A, Manson J, Buring J, Ridker P. Relationship of total and abdominal adiposity with CRP and IL-6 in women. Ann Epidemiol. 2003;13(10):674–682.

40. Pedersen M, Bruunsgaard H, Weis N, et al. Circulating levels of TNF-alpha and IL-6-relation to truncal fat mass and muscle mass in healthy elderly individuals and in patients with type-2 diabetes. Mech Ageing Dev. 2003;124(4):495–502. doi:10.1016/s0047-6374(03)00027-7

41. Bastard J, Maachi M, Van Nhieu J, et al. Adipose tissue IL-6 content correlates with resistance to insulin activation of glucose uptake both in vivo and in vitro. J Clin Endocrinol Metab. 2002;87(5):2048–2049. doi:10.1210/jcem.87.5.8450

42. Lafontan M, Berlan M. Do regional differences in adipocyte biology provide new pathophysiological insights? Trends Pharmacol Sci. 2003;24(6):276–283. doi:10.1016/S0165-6147(03)00132-9

43. Hoffman R, Dye A, Huang H, Bauer J. Glycemic variability predicts inflammation in adolescents with type 1 diabetes. J Pediatr Endocrinol Metab. 2016;29(10):1129–1133. doi:10.1515/jpem-2016-0139

44. Dal S, Jeandidier N, Seyfritz E, et al. Featured article: oxidative stress status and liver tissue defenses in diabetic rats during intensive subcutaneous insulin therapy. Exp Biol Med. 2016;241(2):184–192. doi:10.1177/1535370215603837

45. Monnier L, Mas E, Ginet C, et al. Activation of oxidative stress by acute glucose fluctuations compared with sustained chronic hyperglycemia in patients with type 2 diabetes. Jama. 2006;295(14):1681–1687. doi:10.1001/jama.295.14.1681

Creative Commons License © 2019 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.