Back to Journals » Journal of Inflammation Research » Volume 18
Exploring Inflammatory Changes in the Peripheral Blood of Type 2 Diabetes Mellitus in China
Authors Li D, Zhang Z, Li W, Zhao C, Li X, Pan Z, Li Y
Received 15 October 2024
Accepted for publication 27 January 2025
Published 4 February 2025 Volume 2025:18 Pages 1679—1688
DOI https://doi.org/10.2147/JIR.S501105
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Dr Adam D Bachstetter
Dan Li,1,2,* Zhiru Zhang,3,* Wen Li,2 Chenhao Zhao,2 Xiaochen Li,2 Zhen Pan,2 Yang Li1
1Department of Clinical Laboratory, Children’s Hospital of Soochow University, Suzhou, Jiangsu, 215025, People’s Republic of China; 2Department of Clinical Laboratory, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, People’s Republic of China; 3Department of Human Resources, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, 22600, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Yang Li, Department of Clinical Laboratory, Children’s Hospital of Soochow University, Suzhou, Jiangsu, 215025, People’s Republic of China, Tel +86-512-80691230, Email [email protected]
Objective: Type 2 diabetes mellitus (T2DM) is a serious, chronic metabolic disease globally and its pathogenesis is not completely understood yet. This study aimed to thoroughly investigate the fluctuation of inflammatory markers in the peripheral blood of patients with T2DM, which are rarely reported.
Methods: Peripheral blood samples from patients with T2DM and healthy individuals, as well as their clinical information, were collected at the Second Affiliated Hospital of Soochow University. Flow cytometry was used to analyse the immune cells and cytokines in the peripheral blood. CCK-8 assay was performed to detect the viability of THP-1 cell after treatment with 5 m&Mgr; or 50 m&Mgr; glucose. Flow cytometry, Western Blotting and qPCR were used to analyse the apoptosis of monocytes or THP-1 cells.
Results: The numbers of white blood cells, lymphocytes, and neutrophils substantially increased with elevated IL-6 levels. There was a significant decrease in monocytes due to increased cell apoptosis caused by sustained high glucose stimulation. Hyperglycemia reduced monocyte viability and altered monocyte subgroups by increasing the number of intermediate and non-classical monocytes.
Conclusion: In summary, our work reveals that patients with T2DM do have variations of peripheral inflammation biomarkers, especially monocytes.
Keywords: Type 2 diabetes mellitus, peripheral inflammation, monocytes
Introduction
Diabetes mellitus (DM) is one of the major global diseases and type 2 diabetes mellitus (T2DM) accounts for approximately 95%.1 T2DM is an organ restricted disease, characterized by a relative β-cell deficit or insulin resistance that lead to inability of pancreatic β-cells to meet the demand for insulin.2 High glucose is one of its characteristics which has negative impacts on several organs, such as the brain, kidney, and eyes.3,4 However, to date, there have been no systematic studies on the effects of hyperglycemia on the inflammation in the peripheral blood of T2DM patients.5
Inflammation has been a global research topic and plays a pivotal role in T2DM progression, which is often associated with circulating pro-inflammatory cytokines and immune cells,6,7 but its mechanism still needs to be elucidated. Pro-inflammatory cytokines, like TNF-α, IL-1β and IL-17, are considered the central drivers of the inflammatory cascade, leading to β-cell dysfunction.8 Immune cells have been shown to be associated with diabetes risk and insulin resistance in some epidemiological studies, which mainly refer to white blood cells, such as circulating Th1 and Th17 lymphocytes, neutrophils, and monocyte-macrophages in the peripheral blood.9 In addition, previous studies also reported the increase of peripheral dendritic cells in T2DM patients.7
Monocytes, together with CD8+ T cells, are the first cells recruited to inflamed pancreatic tissues during the development of T2DM.10,11 Monocytes are divided into three subgroups and have a relatively unique immunological function. CD14+CD16− classical monocytes mainly show anti-inflammatory function. The CD14+CD16+ intermediate and CD14−CD16+ non-classical subgroups mainly have pro-inflammatory effects.12 Studies have shown that monocytes from patients with DM have reduced functions in phagocytosis, chemotaxis, and killing compared to cells from healthy groups.13,14 However, there have been few reports on the effects of high glucose levels on monocyte classification, cell viability, or survival.
The impact of hyperglycemia on islet tissue and islet β cells is well known, but there is a lack of comprehensive understanding between the inflammation changes in peripheral blood of T2DM patients. In this study, we investigated the inflammatory alterations in the peripheral blood of patients with T2DM and found the variations of peripheral inflammation biomarkers, especially monocytes. Although the number of patients in this study may not be large enough and this study was just conducted in Suzhou area, these results will help to reveal the relationship between hyperglycemia and inflammation in T2DM.
Materials and Methods
Clinical Information of Patients With T2DM
Peripheral blood from sex-matched individuals with T2DM, based on the guidelines of the American Diabetes Association, was collected from the Department of Clinical Laboratory, Second Affiliated Hospital of Soochow University, from January to December 2023. Peripheral blood samples from 515 patients with T2DM and 494 healthy individuals were analyzed. This study was approved by the Ethics Committee of the Second Affiliated Hospital of Soochow University (JD-LK-2021–064-01) and was conducted in accordance with the Declaration of Helsinki.
We selected patients with T2DM with exclusion criteria as follows: patients with T1DM, severe hepatopulmonary dysfunction, severe renal insufficiency (glomerular filtration rate (eGFR) < 30 mL/1.73 mm2), and inflammatory disease; and patients who were unmatched for age and sex.
Flow Cytometry Analysis of Peripheral Immune Cell
Whole peripheral blood (50 µL) from patients with T2DM was stained for neutrophils, lymphocytes, and subsets of monocyte analyzed in flow cytometry. Surface markers included Pe-cy5.5-CD45, APC-CD14, PE-CD16, FITC-CD3, PE-Cy7-CD4, and APC-Cy7-CD8. Each sample was added to 1 mL RBC lysis buffer for 15 min and stained with the mixed markers for another 15 min at room temperature. Then removed the supernatant after centrifugation, and resuspended with 500 μL of Phosphate Buffer Saline (PBS) for analysis. All antibodies used for flow cytometry were purchased from BD Biosciences. The whole peripheral blood from healthy individuals were used as control.
Cytometric Bead Array of Cytokines
Serums from patients with T2DM in pro-coagulant collection tubes were analyzed for cytokines (IL-2, IL-4, IL-6, IL-10, TNF-α, IFN-γ, and IL-17A) by using a BD™ Cytometric Bead Array kit according to the manufacturer’s instruction. The serum from healthy individuals were used as control.
Cell Counting Kit 8 (CCK8) Assay
THP-1 cells were uniformly seeded in a 96-well plate (1× 104 cells/well). Six wells with THP-1 cells were treated with 50 mΜ glucose for 24, 48, and 72 h for cell viability using a cell counting kit (CCK)-8 (No. A311-01, Vazyme), according to the manufacturer’s protocol. Meanwhile, six wells with THP-1 cells without treated with 50 mΜ glucose were used as negative controls and six wells with only cell medium were used as blank controls. Optical density at 450 nm was determined using a FLUO Star Omega. Three independent experiments were performed at the same time.
Peripheral Monocyte Separation
Peripheral blood monocytes from patients with T2DM and healthy individuals were separated by using Ficoll-Paque PLUS density gradient medium (Sigma) according to the manufacturer’s instructions.
Cell Apoptosis
Monocytes or THP-1 cells (5× 105 per well) were cultured for 12 h respectively in the presence of 50 mM glucose. The cells were collected and stained with Annexin V and PI (No. A211, Vazyme), according to the manufacturer’s instructions. To analyze changes in protein levels, we lysed cultured THP-1 cells for Western blotting. Antibodies were purchased from Cell Signaling Technology. The mRNA levels of apoptosis-associated proteins (cleave-caspase-3, total caspase-3, and Bcl-2) were determined by using qPCR. The forward and reverse primers of Bcl-2, Bax, Bad, Bcl-xl, caspase-3 and GAPDH were synthesized by Tsingke Biotechnology (Table 1). Monocytes or THP-1 cells without treatment of 50 mM glucose were used as control.
![]() |
Table 1 Primers for qPCR |
Statistical Analysis
GraphPad Prism 8.0 software was used for statistical calculation. The results are expressed as the mean ± SD. Differences were assessed by Student’s t test. P < 0.05 was considered statistically significant.
Results
Clinical Characteristics of Patients With T2DM
The demographic and general characteristics of patients with T2DM are summarized in Table 2. Our data showed that among the 515 enrolled patients, the incidence in males was higher than that of females. Most patients (69.15%) suffered from this disease for more than five years. Half of the patients had high blood pressure or hyperglycemia. In addition, nearly 70% of patients have poor blood sugar control, which can be reflected by the fact that HbA1c is higher than 7%. We then performed a further analysis of the patients’ age (Figure 1 and Table 2). We found that the patients with diabetes were younger. Notably, among these 515 patients, the youngest was only 24 years old, and there were five individuals who were younger than 30 years old. Younger onset age may have a substantial association the current dietary habits of human beings.
![]() |
Table 2 Characteristics of T2DM |
![]() |
Figure 1 Age distribution of 515 patients with T2DM. |
T2DM Patients Have Variations in Peripheral Immune System
The harm of diabetes does not lie on hyperglycemia itself, but on a series of chronic inflammatory conditions that lead to high morbidity, disability, and mortality. In this study, we found that the total numbers of white blood cells (WBC), lymphocytes (Lym), and neutrophils (Neu) significantly increased, giving rise to inflammation (Figure 2A, left). Lymphocyte subsets showed no obvious changes (Supplementary Figure 1). Notably, the number and percentage of monocytes (Mon) were considerably significantly reduced (Figure 2A, right). IL-6 markedly increased, but IFN-γ decreased in the peripheral blood of patients with T2DM compared with those in healthy controls. The expression of other cytokines, such as IL-2, IL-4, IL-10, IL-12, IL-17A and TNF-α, was unchanged between patients with T2DM and healthy individuals (Figure 2B and 2C). These variations contribute to the peripheral immune inflammation of T2DM.
Hyperglycemia Affects the Differentiation and Cell Viability of Monocytes
Notably, the number of monocytes in peripheral blood of patients with T2DM decreased. Therefore, we focused on monocytes in future studies. We made distinctions between the following monocyte subgroups: classical monocytes (CM, CD14+CD16−), intermediate monocytes (IM, CD14+CD16+), and non-classical monocytes (nCM, CD14−CD16+). In the peripheral blood of patients with T2DM, the percentage of classical monocytes was decreased, indicating a weakened phagocytic ability. However, IM and nCM levels increased, especially in intermediate monocytes with sharp amplification, which has attracted much attention because of its pro-inflammatory function in the pathophysiology of cardiovascular diseases (Figure 3A). We also found that high concentrations of glucose (50 mM) reduced the viability of monocytic THP-1 cells (Figure 3B).
Monocyte Cell Apoptosis Augments in T2DM
To assess cell survival, we isolated monocytes from the peripheral blood of healthy controls and patients with T2DM, and then analyzed the combination of annexin V and propidium iodide (PI) after glucose stimulation. For the monocytes from T2DM patients, we found a high percentage of early apoptosis (Annexin-V+PI−) with 5 or 50 mM glucose stimulation in culture medium than those from healthy individuals (Figure 4A and 4B). This phenomenon can be explained by the irreversible apoptosis-prone state caused by long-term hyperglycemia. Monocytes from healthy controls showed no palpable changes in cell apoptosis when cultured with 5 or 50 mM glucose for 12 h in vitro, which may be due to the short stimulation time of high sugar. There were no differences in the percentage of Annexin-V−PI+ or Annexin-V+ PI+ cells among the four groups (Figure 4A and 4B). These monocytes were subjected to Western blotting to detect the levels of apoptosis-related proteins, cleave-caspase-3, total caspase-3, and Bcl-2. Gray analysis showed an increased expression of cleave-caspase-3 and total caspase-3 (Supplementary Figure 2). However, when we cultured the monocyte cell-line, THP-1 for 12 h, we found a significant increase in cell early apoptosis from 4% to 6% (Figure 4C). Meanwhile, mRNA expression of caspase-3 in THP-1 also markedly increased after culturing with 50 mM glucose (Figure 4D). In general, persistent hyperglycemia increased the apoptosis of monocytes.
Discussion
DM is a persistent metabolic disorder that affects millions of people worldwide. T2DM accounts for approximately 95% of diabetes cases, and its pathogenesis may be influenced by complex genetic and environmental factors that are not completely revealed yet.15,16 At present, it can only be maintained by drugs but still has high morbidity, mortality and complications, which are primarily attributed to sustained hyperglycemia and insulin resistance.17 As a chronic condition, T2DM tends to increase the risk of several other diseases caused by macrovascular and microvascular damage. Moreover, it also has negative effects on several organs, such as brain, kidney, heart and eyes.18 T2DM is a serious, chronic metabolic disease, therefore, it is expected to have a higher number of white blood cells, lymphocytes, and neutrophils in the peripheral blood of patients with T2DM. The levels of the pro-inflammatory cytokine IL-6 also significantly increased. However, the number of monocytes decreased and further analysis revealed its impact on monocyte differentiation and cell viability. Hyperglycemia induces monocyte apoptosis by increasing the expression of caspase-3. In summary, patients with T2DM do have variations of inflammatory biomarkers in their peripheral blood.
Various inflammatory markers are associated with glucose-related disorders.19 Immune cells in the peripheral blood mainly refer to white blood cells, such as circulating lymphocytes, neutrophils, and monocyte-macrophages.20 Studies have shown that the risk of diabetes increases with an increased number of white blood cell, which is positively correlated with insulin resistance.21 Neutrophil dysregulation, characterized by effector dysfunction and inadequate cell death processes that lead to uncontrolled inflammatory responses, can serve as an inflammatory biomarker in patients with diabetes.22 Other observations also indicate that eosinophils in diabetes, both in the blood circulation and renal tissue, might be associated with the development and severity of diabetic nephropathy.23 In this study, we found increased leukocytes, lymphocytes, and neutrophils in the peripheral blood of T2DM, but no change in eosinophils or basophils. The increased lymphocytes is closely related to the secretion of cytokines, while the increase of neutrophils may be due to a compensatory increase in dysfunction and inadequate cell death processes. These two cell types account for a large proportion of peripheral blood. Therefore, they are considered the main cells contributing to peripheral immune inflammation.
The peripheral blood is important for the circulation and redistribution of lymphocytes to tissues and organs. Lymphocytes, especially CD8+ T cells, were the first cells found in the inflamed pancreatic tissue during disease development. CD8+ T lymphocytes contribute to persistent low-grade inflammation by secreting pro-inflammatory cytokines and producing cytotoxic molecules, such as granzyme B and perforin.24 Circulating lymphocytes are increased in patients with T2DM in this study, but the percentages of lymphocyte subsets that were mainly divided into CD4+T, CD8+T, NK, and B cells showed no obvious change. Meanwhile, inflammatory responses do not depend on single mediator, but on various markers. Cytokines are one of these markers.25 We found increased IL-6 levels, which indicates a state of chronic inflammation in the peripheral blood of T2DM patients. In patients with type 1 diabetes mellitus (T1DM), islet autoantigens can stimulate lymphocytes to produce more IL-6, IFN-γ and TNF-α.26 This was slightly different from what was observed in T2DM.
Monocyte-macrophages were reported as the main effector cells that reduced insulin signaling. Patients with hyperglycemia had more macrophages that infiltrated the adipose tissue and islets, causing inflammation.27 In this study, we found monocytes were the only cells that were decreased in the peripheral blood of T2DM patients. The reduced number of circulating monocytes in the peripheral blood may have been due to increased tissue settlement. To analyze the reasons for the diminished monocyte counts, experiments demonstrated that hyperglycemia reduced the cell activity and increased the apoptosis of monocytes, which is partially consistent with previous reports that monocytes from patients with T2DM have reduced chemotaxis, phagocytosis, and killing compared to cells from healthy groups. In view of the particularity of this phenotype and the importance of their function, we focused on the analysis of monocyte subsets. CD14+CD16− CMs are involved in phagocytosis, pathogen elimination and anti-inflammatory effects. CD14+CD16+ IMs play an antigen-presenting role that affects CD4+ T cell proliferation and differentiation. CD14−CD16+ nCM are primarily involved in immune surveillance and tissue repair. The latter two are pro-inflammatory cells.12 In the present study, the percentage of CM in the peripheral blood from patients with T2DM decreased, while the percentage of IM and nCM increased. However, we did not explore how high glucose affects the differentiation of monocytes. That will be the key points of our future researches.
Conclusion
In this study, we investigated the inflammatory alterations in the peripheral blood of patients with T2DM and found the variations of some peripheral inflammatory cytokines and immune cells, especially monocytes. Although the number of patients in this study may not be large enough and this study was just conducted in Suzhou area, these results will help to reveal the relationship between hyperglycemia and inflammation in T2DM.
Data Sharing Statement
All data generated or analysed during this study are included in this article. Further enquiries can be directed to the corresponding author.
Ethics Statement
Informed consent was obtained from all individual participants included in the study. All of the experimental procedures were approved by the ethics committee of the Second Affiliated Hospital of Soochow University (JD-LK-2021-064-01).
Informed Consent
Written informed consent was obtained from all the patients or their guardians.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
This work was supported by the High-level Innovative and Entrepreneurial Talents Introduction Program of Jiangsu Province (2020–30186, 2020–30191), Government Scholarship for Studying Abroad of Jiangsu Province (2024–087), Jiangsu Health International Exchange Program (2024–038), the Medical Research Project of Jiangsu Commission of Health (M2020027), Gusu Health Talents Program of Suzhou (2024–094, 2021–020 and GSWS2022034), National Tutorial System Project of Suzhou Health Young Backbone Talents (Qngg2022011), the Science and Technology Program of Suzhou (SYW2024103, SKJY2021092, SYS2020163, SYSD2019120), Discipline Construction Program of the Second Affiliated Hospital of Soochow University (XKTJ-TD202001), Key Research Program Cultivation Project of Children’s Hospital of Soochow University (2023ZDPY02).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Jeremiah SS, Moin ASM, Butler AE. Virus-induced diabetes mellitus: revisiting infection etiology in light of SARS-CoV-2. Metabolism. 2024;156:
2. Karaca M, Magnan C, Kargar C. Functional pancreatic beta-cell mass: involvement in type 2 diabetes and therapeutic intervention. Diabetes Metab. 2009;35(2):77–84. doi:10.1016/j.diabet.2008.09.007
3. Wanner C, Marx N. SGLT2 inhibitors: the future for treatment of type 2 diabetes mellitus and other chronic diseases. Diabetologia. 2018;61(10):2134–2139. doi:10.1007/s00125-018-4678-z
4. Parkman HP, Wilson LA, Farrugia G, et al. NIDDK Gastroparesis Clinical Research Consortium (GpCRC). delayed gastric emptying associates with diabetic complications in diabetic patients with symptoms of gastroparesis. Am J Gastroenterol. 2019;114(11):1778–1794. doi:10.14309/ajg.0000000000000410
5. Qin W, Sun L, Dong M, et al. Regulatory T cells and diabetes mellitus. Hum Gene Ther. 2021;32(17–18):875–881. doi:10.1089/hum.2021.024
6. Inayat H, Azim MK, Baloch AA. Analysis of inflammatory gene expression profile of peripheral blood leukocytes in Type 2 diabetes. Immunol Invest. 2019;48(6):618–631. doi:10.1080/08820139.2019.1586917
7. Cinkajzlová A, Mráz M, Haluzík M. Adipose tissue immune cells in obesity, type 2 diabetes mellitus and cardiovascular diseases. J Endocrinol. 2021;252(1):R1–R22. doi:10.1530/JOE-21-0159
8. Tong HV, Luu NK, Son HA, et al. Adiponectin and pro-inflammatory cytokines are modulated in Vietnamese patients with type 2 diabetes mellitus. J Diabetes Investig. 2017;8(3):295–305. doi:10.1111/jdi.12579
9. Nam HW, Cho YJ, Lim JA, et al. Functional status of immune cells in patients with long-lasting type 2 diabetes mellitus. Clin Exp Immunol. 2018;194(1):125–136. doi:10.1111/cei.13187
10. Šiklová M, Krauzová E, Svobodová B, et al. Circulating monocyte and lymphocyte populations in healthy first-degree relatives of Type 2 diabetic patients at fasting and during short-term hyperinsulinemia. Mediators Inflamm. 2019;2019:1491083. doi:10.1155/2019/1491083
11. Corbi SCT, de Vasconcellos JF, Bastos AS, et al. Circulating lymphocytes and monocytes transcriptomic analysis of patients with type 2 diabetes mellitus, dyslipidemia and periodontitis. Sci Rep. 2020;10(1):8145. doi:10.1038/s41598-020-65042-9
12. Narasimhan PB, Marcovecchio P, Hamers AAJ, Hedrick CC. Nonclassical monocytes in health and disease. Annu Rev Immunol. 2019;26(37):439–456. doi:10.1146/annurev-immunol-042617-053119
13. Pardali E, Makowski LM, Leffers M, Borgscheiper A, Waltenberger J. BMP-2 induces human mononuclear cell chemotaxis and adhesion and modulates monocyte-to-macrophage differentiation. J Cell Mol Med. 2018;22(11):5429–5438. doi:10.1111/jcmm.13814
14. Paccosi S, Pala L, Cresci B, et al. Insulin resistance and obesity affect monocyte-derived dendritic cell phenotype and function. Diabet Res Clin Pract. 2020;170:108528. doi:10.1016/j.diabres.2020.108528
15. Nilsson E, Ling C. DNA methylation links genetics, fetal environment, and an unhealthy lifestyle to the development of type 2 diabetes. Clin Clin Epigenet. 2017;39(1):105. doi:10.1186/s13148-017-0399-2
16. Singh S, Kriti M, A KS, et al. Deciphering the complex interplay of risk factors in type 2 diabetes mellitus: a comprehensive review. Metabol Open. 2024;19(22):100287. doi:10.1016/j.metop.2024.100287
17. Ferrannini E, DeFronzo RA. Impact of glucose-lowering drugs on cardiovascular disease in type 2 diabetes. Eur Heart J. 2015;36(34):2288–2296. doi:10.1093/eurheartj/ehv239
18. Kanazawa A, Aida M, Yoshida Y, et al. Effects of synbiotic supplementation on chronic inflammation and the gut microbiota in obese patients with Type 2 diabetes mellitus: a randomized controlled study. Nutrients. 2021;13(2):558. doi:10.3390/nu13020558
19. Dabravolski SA, Orekhova VA, Baig MS, et al. The role of mitochondrial mutations and chronic inflammation in diabetes. Int J mol Sci. 2021;22(13):6733. doi:10.3390/ijms22136733
20. Pezhman L, Tahrani A, Chimen M. Dysregulation of leukocyte trafficking in Type 2 diabetes: mechanisms and potential therapeutic avenues. Front Cell Dev Biol. 2021;22(9):624184. doi:10.3389/fcell.2021.624184
21. Park JM, Lee HS, Park JY, Jung DH, Lee JW. White blood cell count as a predictor of incident Type 2 diabetes mellitus among non-obese adults: a longitudinal 10-year analysis of the Korean genome and epidemiology study. J Inflamm Res. 2021;1(14):1235–1242. doi:10.2147/JIR.S300026
22. Keeter WC, Moriarty AK, Galkina EV. Role of neutrophils in type 2 diabetes and associated atherosclerosis. Int J Biochem Cell Biol. 2021;141:106098. doi:10.1016/j.biocel.2021.106098
23. Moussa K, Gurung P, Adams-Huet B, Devaraj S, Jialal I. Increased eosinophils in adipose tissue of metabolic syndrome. J Diabetes Complications. 2019;33(8):535–538. doi:10.1016/j.jdiacomp.2019.05.010
24. Lei L, Cui L, Mao Y, et al. Augmented CD25 and CD69 expression on circulating CD8+ T cells in type 2 diabetes mellitus with albuminuria. Diabetes Metab. 2017;43(4):382–384. doi:10.1016/j.diabet.2016.10.002
25. Fadaei R, Bagheri N, Heidarian E, et al. Serum levels of IL-32 in patients with type 2 diabetes mellitus and its relationship with TNF-α and IL-6. Cytokine. 2020;125:154832. doi:10.1016/j.cyto.2019.154832
26. Amin K, Qadr SH, Hassan Hussein R, Ali KM, Rahman HS. Levels of cytokines and GADA in type I and II diabetic patients. Prim Care Diabet. 2020;14(1):61–67. doi:10.1016/j.pcd.2019.03.008
27. Ying W, Fu W, Lee YS, Olefsky JM. The role of macrophages in obesity-associated islet inflammation and β-cell abnormalities. Nat Rev Endocrinol. 2020;16(2):81–90. doi:10.1038/s41574-019-0286-3
© 2025 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, 3.0) 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.