Subscribe to RSS
DOI: 10.1055/a-2715-0775
The Relationship Between Insulin Resistance and Cancer in Humans
Authors
Abstract
An independent association between insulin resistance and cancer has been consistently reported in humans. Patients with cancer display insulin resistance or its clinical manifestations, and this metabolic adaptation precedes the clinical diagnosis of cancer. Insulin resistance in cancer patients is associated with a metabolic switch from oxidative metabolism toward glycolysis that spares oxygen to be used in anabolic processes and facilitates the fast production of energy and intermediate metabolites required for the rapid proliferation of cancer cells. In malignant cells, glucose consumption via glycolysis occurs under normoxic conditions (aerobic glycolysis). Pathogenic mechanisms underlying insulin resistance in cancer patients include hypoxia-inducible factor-1 upregulation and overproduction of cytokines, such as interferon, interleukin-6, interleukin-18, and interleukin-1β. Deficit of 2-oxoglutarate (α-ketoglutarate) has been detected in cancer cells and may facilitate hypoxia-inducible factor-1 assembly and activity. Overproduction of cytokines in cancer patients follows activation of the immune system by abnormal nucleic acid variants. Anomalous DNA or RNA structures are recognized by immune sensors and stimulate signaling pathways that ultimately increase cytokine production. Likewise, interferon overproduction occurs in congenital disorders that feature ineffectively repaired DNA lesions, such as Werner syndrome, Bloom syndrome, mutations in DNA polymerase-δ1, and ataxia telangiectasia. These diseases cause simultaneous insulin resistance and a high tendency to develop cancer, highlighting the relationship between the two processes. Defectively repaired DNA injury endangers genomic integrity, predisposing to cancer, and activates the immune system to increase interferon production and subsequent insulin resistance. Hypoxia-inducible factor-1 and cytokines induce insulin resistance by suppressing peroxisome proliferator-activated-γ in the subcutaneous adipose tissue.
Keywords
insulin resistance - cancer - hypoxia inducible factor - peroxisome proliferator-activated-γ - interferon - DNA repairPublication History
Received: 16 May 2025
Accepted after revision: 30 September 2025
Article published online:
07 November 2025
© 2025. Thieme. All rights reserved.
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
-
References
- 1 Warburg O. On the origin of cancer cells. Science 1956; 123: 309-314
- 2 Lundholm K, Holm G, Scherstén T. Insulin resistance in patients with cancer. Cancer Res 1978; 38: 4665-4670
- 3 Yoshikawa T, Noguchi Y, Doi C. et al. Insulin resistance was connected with the alterations of substrate utilization in patients with cancer. Cancer Lett 1999; 141: 93-98
- 4 Bharadwaj S, Venkatraghavan L, Mariappan R. et al. Serum lactate as a potential biomarker of non-glial brain tumors. J Clin Neurosci 2015; 22: 1625-1627
- 5 Shih CC, Lee TS, Tsuang FY. et al. Pretreatment serum lactate level as a prognostic biomarker in patients undergoing supratentorial primary brain tumor resection. Oncotarget 2017; 8: 63715-63723
- 6 Branco M, Linhares P, Carvalho B. et al. Serum lactate levels are associated with glioma malignancy grade. Clin Neurol Neurosurg 2019; 186: 105546
- 7 Gobinath S, Gobishangar S, Thanenthiran AJ. et al. Challenging refractory type B lactic acidosis in gastric adenocarcinoma-a successfully managed case. J Surg Case Rep 2023; 2023: rjad412
- 8 Ma S, Lee H, Jo WY. et al. The Warburg effect in patients with brain tumors: a comprehensive analysis of clinical significance. J Neurooncol 2023; 165: 219-226
- 9 Lovejoy J, Newby FD, Gebhart SS. et al. Insulin resistance in obesity is associated with elevated basal lactate levels and diminished lactate appearance following intravenous glucose and insulin. Metabolism 1992; 41: 22-27
- 10 Crawford SO, Ambrose MS, Hoogeveen RC. et al. Association of lactate with blood pressure before and after rapid weight loss. Am J Hypertens 2008; 21: 1337-1342
- 11 Chondronikola M, Magkos F, Yoshino J. et al. Effect of Progressive Weight Loss on Lactate Metabolism: A Randomized Controlled Trial. Obesity (Silver Spring) 2018; 26: 683-688
- 12 Jones TE, Pories WJ, Houmard JA. et al. Plasma lactate as a marker of metabolic health: Implications of elevated lactate for impairment of aerobic metabolism in the metabolic syndrome. Surgery 2019; 166: 861-866
- 13 Broskey NT, Pories WJ, Jones TE. et al. The association between lactate and muscle aerobic substrate oxidation: Is lactate an early marker for metabolic disease in healthy subjects?. Physiol Rep 2021; 9: e14729
- 14 Jevtovic F, Zheng D, Lopez CA. et al. Greater reliance on glycolysis is associated with lower mitochondrial substrate oxidation and insulin sensitivity in infant myogenic MSCs. Am J Physiol Endocrinol Metab 2023; 325: E207-E213
- 15 Broskey NT, Pories WJ, DeMaria EJ. et al. Fasting plasma lactate as a possible early clinical marker for metabolic disease risk. Diabetes Metab Syndr 2024; 18: 102955
- 16 Juraschek SP, Selvin E, Miller ER. et al. Plasma lactate and diabetes risk in 8045 participants of the atherosclerosis risk in communities study. Ann Epidemiol 2013; 23: 791-796.e794
- 17 Juraschek SP, Shantha GP, Chu AY. et al. Lactate and risk of incident diabetes in a case-cohort of the atherosclerosis risk in communities (ARIC) study. PLoS One 2013; 8: e55113
- 18 Mongraw-Chaffin ML, Matsushita K, Brancati FL. et al. Diabetes medication use and blood lactate level among participants with type 2 diabetes: the atherosclerosis risk in communities carotid MRI study. PLoS One 2012; 7: e51237
- 19 Rohdenburg GI, Bernhard A, Krehbiel O. Sugar tolerance in cancer. JAMA 1919; 72: 1528-1530
- 20 Marks PA, Bishop JS. The glucose metabolism of patients with malignant disease and of normal subjects as studied by means of an intravenous glucose tolerance test. J Clin Invest 1957; 36: 254-264
- 21 Bishop JS, Marks PA. Studies on carbohydrate metabolism in patients with neoplastic disease. II. Response to insulin administration. J Clin Invest 1959; 38: 668-672
- 22 Makino T, Noguchi Y, Yoshikawa T. et al. Circulating interleukin 6 concentrations and insulin resistance in patients with cancer. Br J Surg 1998; 85: 1658-1662
- 23 Yoshikawa T, Noguchi Y, Doi C. et al. Insulin resistance in patients with cancer: relationships with tumor site, tumor stage, body-weight loss, acute-phase response, and energy expenditure. Nutrition 2001; 17: 590-593
- 24 Màrmol JM, Carlsson M, Raun SH. et al. Insulin resistance in patients with cancer: a systematic review and meta-analysis. Acta Oncol 2023; 62: 364-371
- 25 Park SM, Lim MK, Jung KW. et al. Prediagnosis smoking, obesity, insulin resistance, and second primary cancer risk in male cancer survivors: National Health Insurance Corporation Study. J Clin Oncol 2007; 25: 4835-4843
- 26 Loh WJ, North BV, Johnston DG. et al. Insulin resistance-related biomarker clustering and subclinical inflammation as predictors of cancer mortality during 21.5 years of follow-up. Cancer Causes Control 2010; 21: 709-718
- 27 Perseghin G, Calori G, Lattuada G. et al. Insulin resistance/hyperinsulinemia and cancer mortality: the Cremona study at the 15th year of follow-up. Acta Diabetol 2012; 49: 421-428
- 28 Pan K, Nelson RA, Wactawski-Wende J. et al. Insulin Resistance and Cancer-Specific and All-Cause Mortality in Postmenopausal Women: The Women's Health Initiative. J Natl Cancer Inst 2020; 112: 170-178
- 29 Copeland GP, Leinster SJ, Davis JC. et al. Insulin resistance in patients with colorectal cancer. Br J Surg 1987; 74: 1031-1035
- 30 Farahani H, Mahmoudi T, Asadi A. et al. Insulin Resistance and Colorectal Cancer Risk: the Role of Elevated Plasma Resistin Levels. J Gastrointest Cancer 2020; 51: 478-483
- 31 Hu X, Feng F, Li X. et al. Gene polymorphisms related to insulin resistance and gene-environment interaction in colorectal cancer risk. Ann Hum Biol 2015; 42: 560-568
- 32 Liu T, Zhang Q, Wang Y. et al. Association between the TyG index and TG/HDL-C ratio as insulin resistance markers and the risk of colorectal cancer. BMC Cancer 2022; 22: 1007
- 33 Li W, Liu T, Qian L. et al. Insulin resistance and inflammation mediate the association of abdominal obesity with colorectal cancer risk. Front Endocrinol (Lausanne) 2022; 13: 983160
- 34 Son M, Moon SY, Koh M. et al. Association between Surrogate Markers of Insulin Resistance and the Incidence of Colorectal Cancer in Korea: A Nationwide Population-Based Study. J Clin Med 2024; 13: 1628
- 35 Bruning PF, Bonfrèr JM, van Noord PA. et al. Insulin resistance and breast-cancer risk. Int J Cancer 1992; 52: 511-516
- 36 Gonullu G, Ersoy C, Ersoy A. et al. Relation between insulin resistance and serum concentrations of IL-6 and TNF-alpha in overweight or obese women with early stage breast cancer. Cytokine 2005; 31: 264-269
- 37 Garmendia ML, Pereira A, Alvarado ME. et al. Relation between insulin resistance and breast cancer among Chilean women. Ann Epidemiol 2007; 17: 403-409
- 38 Capasso I, Esposito E, Pentimalli F. et al. Homeostasis model assessment to detect insulin resistance and identify patients at high risk of breast cancer development: National Cancer Institute of Naples experience. J Exp Clin Cancer Res 2013; 32: 14
- 39 Griffith KA, Chung SY, Zhu S. et al. Insulin Resistance and Inflammation in Black Women with and without Breast Cancer: Cause for Concern. Ethn Dis 2016; 26: 513-520
- 40 Kachhawa P, Kachhawa K, Agrawal D. et al. Association of Dyslipidemia, Increased Insulin Resistance, and Serum CA 15-3 with Increased Risk of Breast Cancer in Urban Areas of North and Central India. J Midlife Health 2018; 9: 85-91
- 41 Panigoro SS, Sutandyo N, Witjaksono F. et al. The Association Between Triglyceride-Glucose Index as a Marker of Insulin Resistance and the Risk of Breast Cancer. Front Endocrinol (Lausanne) 2021; 12: 745236
- 42 Pan K, Chlebowski RT, Mortimer JE. et al. Insulin resistance and breast cancer incidence and mortality in postmenopausal women in the Women's Health Initiative. Cancer 2020; 126: 3638-3647
- 43 Hernandez AV, Guarnizo M, Miranda Y. et al. Association between insulin resistance and breast carcinoma: a systematic review and meta-analysis. PLoS One 2014; 9: e99317
- 44 Jung SY, Mancuso N, Papp J. et al. Post genome-wide gene-environment interaction study: The effect of genetically driven insulin resistance on breast cancer risk using Mendelian randomization. PLoS One 2019; 14: e0218917
- 45 Healy LA, Ryan AM, Carroll P. et al. Metabolic syndrome, central obesity and insulin resistance are associated with adverse pathological features in postmenopausal breast cancer. Clin Oncol (R Coll Radiol) 2010; 22: 281-288
- 46 Duggan C, Irwin ML, Xiao L. et al. Associations of insulin resistance and adiponectin with mortality in women with breast cancer. J Clin Oncol 2011; 29: 32-39
- 47 Argirion I, Weinstein SJ, Mannisto S. et al. Serum Insulin, Glucose, Indices of Insulin Resistance, and Risk of Lung Cancer. Cancer Epidemiol Biomarkers Prev 2017; 26: 1519-1524
- 48 Wang G, Zhu Z, Wang Y. et al. The association between METS-IR, an indirect index for insulin resistance, and lung cancer risk. Eur J Public Health 2024; 34: 800-805
- 49 Liu J, Wang R, Tan S. et al. Association between insulin resistance, metabolic syndrome and its components and lung cancer: a systematic review and meta-analysis. Diabetol Metab Syndr 2024; 16: 63
- 50 Hsing AW, Gao YT, Chua S. et al. Insulin resistance and prostate cancer risk. J Natl Cancer Inst 2003; 95: 67-71
- 51 Albanes D, Weinstein SJ, Wright ME. et al. Serum insulin, glucose, indices of insulin resistance, and risk of prostate cancer. J Natl Cancer Inst 2009; 101: 1272-1279
- 52 Lutz SZ, Hennenlotter J, Franko A. et al. Diabetes and the Prostate: Elevated Fasting Glucose, Insulin Resistance and Higher Levels of Adrenal Steroids in Prostate Cancer. J Clin Med 2022; 11: 6762
- 53 Stocks T, Lukanova A, Rinaldi S. et al. Insulin resistance is inversely related to prostate cancer: a prospective study in Northern Sweden. Int J Cancer 2007; 120: 2678-2686
- 54 Yun SJ, Min BD, Kang HW. et al. Elevated insulin and insulin resistance are associated with the advanced pathological stage of prostate cancer in Korean population. J Korean Med Sci 2012; 27: 1079-1084
- 55 Jochems SHJ, Fritz J, Häggström C. et al. Prediagnostic markers of insulin resistance and prostate cancer risk and death: A pooled study. Cancer Med 2023; 12: 13732-13744
- 56 Wang J, Apizi A, Tao N. et al. Association between the metabolic score for insulin resistance and prostate cancer: a cross-sectional study in Xinjiang. PeerJ 2024; 12: e17827
- 57 Li L, Meng F, Xu D. et al. Synergism between the metabolic syndrome components and cancer incidence: results from a prospective nested case-control study based on the China Health and Retirement Longitudinal Study (CHARLS). BMJ Open 2022; 12: e061362
- 58 Stocks T, Rapp K, Bjørge T. et al. Blood glucose and risk of incident and fatal cancer in the metabolic syndrome and cancer project (me-can): analysis of six prospective cohorts. PLoS Med 2009; 6: e1000201
- 59 Borena W, Stocks T, Jonsson H. et al. Serum triglycerides and cancer risk in the metabolic syndrome and cancer (Me-Can) collaborative study. Cancer Causes Control 2011; 22: 291-299
- 60 Chen W, Lu F, Liu SJ. et al. Cancer risk and key components of metabolic syndrome: a population-based prospective cohort study in Chinese. Chin Med J (Engl) 2012; 125: 481-485
- 61 Stocks T, Van Hemelrijck M, Manjer J. et al. Blood pressure and risk of cancer incidence and mortality in the Metabolic Syndrome and Cancer Project. Hypertension 2012; 59: 802-810
- 62 Liu T, Liu CA, Zhang QS. et al. Association of the age of onset of metabolic syndrome with the risk of all cancer types. Diabetes Metab Syndr 2023; 17: 102896
- 63 Lai YJ, Yen YF, Chen LJ. et al. Modification of incident cancer risk with changes in metabolic syndrome status: A prospective cohort study in Taiwan. Ann Epidemiol 2024; 91: 65-73
- 64 Deng L, Liu T, Liu CA. et al. The association of metabolic syndrome score trajectory patterns with risk of all cancer types. Cancer 2024; 130: 2150-2159
- 65 Esposito K, Chiodini P, Colao A. et al. Metabolic syndrome and risk of cancer: a systematic review and meta-analysis. Diabetes Care 2012; 35: 2402-2411
- 66 Jaggers JR, Sui X, Hooker SP. et al. Metabolic syndrome and risk of cancer mortality in men. Eur J Cancer 2009; 45: 1831-1838
- 67 Lee JS, Cho SI, Park HS. Metabolic syndrome and cancer-related mortality among Korean men and women. Ann Oncol 2010; 21: 640-645
- 68 Watanabe J, Kakehi E, Kotani K. et al. Metabolic syndrome is a risk factor for cancer mortality in the general Japanese population: the Jichi Medical School Cohort Study. Diabetol Metab Syndr 2019; 11: 3
- 69 Nguyen TV, Arisawa K, Katsuura-Kamano S. et al. Associations of metabolic syndrome and metabolically unhealthy obesity with cancer mortality: The Japan Multi-Institutional Collaborative Cohort (J-MICC) study. PLoS One 2022; 17: e0269550
- 70 Van Hoang D, Inoue Y, Fukunaga A. et al. Metabolic syndrome and the risk of severe cancer events: a longitudinal study in Japanese workers. BMC Cancer 2023; 23: 555
- 71 Maiti B, Kundranda MN, Spiro TP. et al. The association of metabolic syndrome with triple-negative breast cancer. Breast Cancer Res Treat 2010; 121: 479-483
- 72 Cho WK, Choi DH, Park W. et al. Effect of Body Mass Index on Survival in Breast Cancer Patients According to Subtype, Metabolic Syndrome, and Treatment. Clin Breast Cancer 2018; 18: e1141-e1147
- 73 Taroeno-Hariadi KW, Putra YR, Ariesta NF. et al. The Association between Abdominal Obesity, Metabolic Syndrome and Survival Outcomes in Patients with Breast Cancer. Asian Pac J Cancer Prev 2022; 23: 3157-3165
- 74 Chlebowski RT, Aragaki AK, Pan K. et al. Breast cancer incidence and mortality by metabolic syndrome and obesity: The Women's Health Initiative. Cancer 2024; 130: 3147-3156
- 75 Chen Y, Kong W, Liu M. et al. Metabolic syndrome and risk of colorectal cancer: A Mendelian randomization study. Heliyon 2024; 10: e23872
- 76 Zhan ZQ, Chen YZ, Huang ZM. et al. Metabolic syndrome, its components, and gastrointestinal cancer risk: a meta-analysis of 31 prospective cohorts and Mendelian randomization study. J Gastroenterol Hepatol 2024; 39: 630-641
- 77 Yuan C, Shu X, Hu Z. et al. Genetic prediction of the relationship between metabolic syndrome and colorectal cancer risk: a Mendelian randomization study. Diabetol Metab Syndr 2024; 16: 109
- 78 Esposito K, Chiodini P, Capuano A. et al. Colorectal cancer association with metabolic syndrome and its components: a systematic review with meta-analysis. Endocrine 2013; 44: 634-647
- 79 Hu D, Zhang M, Zhang H. et al. Prediction of Metabolic Syndrome for the Survival of Patients With Digestive Tract Cancer: A Meta-Analysis. Front Oncol 2019; 9: 281
- 80 Han F, Wu G, Zhang S. et al. The association of Metabolic Syndrome and its Components with the Incidence and Survival of Colorectal Cancer: A Systematic Review and Meta-analysis. Int J Biol Sci 2021; 17: 487-497
- 81 Shen X, Wang Y, Zhao R. et al. Metabolic syndrome and the risk of colorectal cancer: a systematic review and meta-analysis. Int J Colorectal Dis 2021; 36: 2215-2225
- 82 Lu B, Qian JM, Li JN. The metabolic syndrome and its components as prognostic factors in colorectal cancer: A meta-analysis and systematic review. J Gastroenterol Hepatol 2023; 38: 187-196
- 83 Esposito K, Chiodini P, Capuano A. et al. Effect of metabolic syndrome and its components on prostate cancer risk: meta-analysis. J Endocrinol Invest 2013; 36: 132-139
- 84 Bhandari R, Kelley GA, Hartley TA. et al. Metabolic syndrome is associated with increased breast cancer risk: a systematic review with meta-analysis. Int J Breast Cancer 2014; 2014: 189384
- 85 Guo M, Liu T, Li P. et al. Association Between Metabolic Syndrome and Breast Cancer Risk: An Updated Meta-Analysis of Follow-Up Studies. Front Oncol 2019; 9: 1290
- 86 Zhao P, Xia N, Zhang H. et al. The Metabolic Syndrome Is a Risk Factor for Breast Cancer: A Systematic Review and Meta-Analysis. Obes Facts 2020; 13: 384-396
- 87 Li P, Wang T, Zeng C. et al. Association between metabolic syndrome and prognosis of breast cancer: a meta-analysis of follow-up studies. Diabetol Metab Syndr 2020; 12: 10
- 88 Sin S, Lee CH, Choi SM. et al. Metabolic Syndrome and Risk of Lung Cancer: An Analysis of Korean National Health Insurance Corporation Database. J Clin Endocrinol Metab 2020; 105: e4102-e4111
- 89 Zhang Z, Liu Q, Huang C. et al. Association Between Metabolic Syndrome and the Risk of Lung Cancer: A Meta-Analysis. Horm Metab Res 2023; 55: 846-854
- 90 Li M, Cao SM, Dimou N. et al. Association of Metabolic Syndrome With Risk of Lung Cancer: A Population-Based Prospective Cohort Study. Chest 2024; 165: 213-223
- 91 Lund Håheim L. Metabolic syndrome and prostate cancer. Expert Rev Endocrinol Metab 2007; 2: 633-640
- 92 Xiang YZ, Xiong H, Cui ZL. et al. The association between metabolic syndrome and the risk of prostate cancer, high-grade prostate cancer, advanced prostate cancer, prostate cancer-specific mortality and biochemical recurrence. J Exp Clin Cancer Res 2013; 32: 9
- 93 Gacci M, Russo GI, De Nunzio C. et al. Meta-analysis of metabolic syndrome and prostate cancer. Prostate Cancer Prostatic Dis 2017; 20: 146-155
- 94 Motterle G, Zecchini G, Mandato FG. et al. Metabolic syndrome and risk of prostate cancer: a systematic review and meta-analysis. Panminerva Med 2022; 64: 337-343
- 95 Childs KS, Randall RE, Goodbourn S. LGP2 plays a critical role in sensitizing mda-5 to activation by double-stranded RNA. PLoS One 2013; 8: e64202
- 96 Duic I, Tadakuma H, Harada Y. et al. Viral RNA recognition by LGP2 and MDA5, and activation of signaling through step-by-step conformational changes. Nucleic Acids Res 2020; 48: 11664-11674
- 97 Wang Y, Samuel CE. Adenosine deaminase ADAR1 increases gene expression at the translational level by decreasing protein kinase PKR-dependent eIF-2alpha phosphorylation. J Mol Biol 2009; 393: 777-787
- 98 Chung H, Calis JJA, Wu X. et al. Human ADAR1 Prevents Endogenous RNA from Triggering Translational Shutdown. Cell 2018; 172: 811-824.e814
- 99 Gannon HS, Zou T, Kiessling MK. et al. Identification of ADAR1 adenosine deaminase dependency in a subset of cancer cells. Nat Commun 2018; 9: 5450
- 100 Liu H, Golji J, Brodeur LK. et al. Tumor-derived IFN triggers chronic pathway agonism and sensitivity to ADAR loss. Nat Med 2019; 25: 95-102
- 101 Ishizuka JJ, Manguso RT, Cheruiyot CK. et al. Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade. Nature 2019; 565: 43-48
- 102 George CX, Samuel CE. Human RNA-specific adenosine deaminase ADAR1 transcripts possess alternative exon 1 structures that initiate from different promoters, one constitutively active and the other interferon inducible. Proc Natl Acad Sci U S A 1999; 96: 4621-4626
- 103 Bazak L, Haviv A, Barak M. et al. A-to-I RNA editing occurs at over a hundred million genomic sites, located in a majority of human genes. Genome Res 2014; 24: 365-376
- 104 Paul SK, Oshima M, Patil A. et al. Retrotransposons in Werner syndrome-derived macrophages trigger type I interferon-dependent inflammation in an atherosclerosis model. Nat Commun 2024; 15: 4772
- 105 Cosentino GP, Venkatesan S, Serluca FC. et al. Double-stranded-RNA-dependent protein kinase and TAR RNA-binding protein form homo- and heterodimers in vivo. Proc Natl Acad Sci U S A 1995; 92: 9445-9449
- 106 Vattem KM, Staschke KA, Zhu S. et al. Inhibitory sequences in the N-terminus of the double-stranded-RNA-dependent protein kinase, PKR, are important for regulating phosphorylation of eukaryotic initiation factor 2alpha (eIF2alpha). Eur J Biochem 2001; 268: 1143-1153
- 107 Patel RC, Sen GC. PACT, a protein activator of the interferon-induced protein kinase, PKR. EMBO J 1998; 17: 4379-4390
- 108 Zhou YJ, Zhou H, Li Y. et al. NOD1 activation induces innate immune responses and insulin resistance in human adipocytes. Diabetes Metab 2012; 38: 538-543
- 109 Yi-Jun Z, Ai L, Yu-Ling S. et al. Nucleotide-binding oligomerization domain-1 ligand induces inflammation and attenuates glucose uptake in human adipocytes. Chin Med Sci J 2012; 27: 147-152
- 110 Lappas M. NOD1 expression is increased in the adipose tissue of women with gestational diabetes. J Endocrinol 2014; 222: 99-112
- 111 Zhou YJ, Liu C, Li CL. et al. Increased NOD1, but not NOD2, activity in subcutaneous adipose tissue from patients with metabolic syndrome. Obesity (Silver Spring) 2015; 23: 1394-1400
- 112 Zhang T, Tian J, Fan J. et al. Exercise training-attenuated insulin resistance and liver injury in elderly pre-diabetic patients correlates with NLRP3 inflammasome. Front Immunol 2023; 14: 1082050
- 113 Goossens GH, Blaak EE, Theunissen R. et al. Expression of NLRP3 inflammasome and T cell population markers in adipose tissue are associated with insulin resistance and impaired glucose metabolism in humans. Mol Immunol 2012; 50: 142-149
- 114 Esser N, L'Homme L, De Roover A. et al. Obesity phenotype is related to NLRP3 inflammasome activity and immunological profile of visceral adipose tissue. Diabetologia 2013; 56: 2487-2497
- 115 Bando S, Fukuda D, Soeki T. et al. Expression of NLRP3 in subcutaneous adipose tissue is associated with coronary atherosclerosis. Atherosclerosis 2015; 242: 407-414
- 116 Kursawe R, Dixit VD, Scherer PE. et al. A Role of the Inflammasome in the Low Storage Capacity of the Abdominal Subcutaneous Adipose Tissue in Obese Adolescents. Diabetes 2016; 65: 610-618
- 117 Hunter CA, Timans J, Pisacane P. et al. Comparison of the effects of interleukin-1 alpha, interleukin-1 beta and interferon-gamma-inducing factor on the production of interferon-gamma by natural killer. Eur J Immunol 1997; 27: 2787-2792
- 118 Tominaga K, Yoshimoto T, Torigoe K. et al. IL-12 synergizes with IL-18 or IL-1beta for IFN-gamma production from human T cells. Int Immunol 2000; 12: 151-160
- 119 Cooper MA, Fehniger TA, Ponnappan A. et al. Interleukin-1beta costimulates interferon-gamma production by human natural killer cells. Eur J Immunol 2001; 31: 792-801
- 120 Micallef MJ, Ohtsuki T, Kohno K. et al. Interferon-gamma-inducing factor enhances T helper 1 cytokine production by stimulated human T cells: synergism with interleukin-12 for interferon-gamma production. Eur J Immunol 1996; 26: 1647-1651
- 121 Xiong D, Feng R, Yang S. et al. Antitumor activity of interleukin-18 on A549 human lung cancer cell line. J Cancer Res Ther 2019; 15: 1635-1641
- 122 Choudhury RP, Birks JS, Mani V. et al. Arterial Effects of Canakinumab in Patients With Atherosclerosis and Type 2 Diabetes or Glucose Intolerance. J Am Coll Cardiol 2016; 68: 1769-1780
- 123 Ridker PM, MacFadyen JG, Thuren T. et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 2017; 390: 1833-1842
- 124 Lanka N, Acharya P, Virani S. et al. Safety and Efficacy of Canakinumab for the Prevention and Control of Type 2 Diabetes Mellitus and Its Complications: A Systematic Review. Cureus 2024; 16: e67065
- 125 Li X, Shu C, Yi G. et al. Cyclic GMP-AMP synthase is activated by double-stranded DNA-induced oligomerization. Immunity 2013; 39: 1019-1031
- 126 Zhang K, Wang S, Gou H. et al. Crosstalk Between Autophagy and the cGAS-STING Signaling Pathway in Type I Interferon Production. Front Cell Dev Biol 2021; 9: 748485
- 127 Yuan F, Dutta T, Wang L. et al. Human DNA Exonuclease TREX1 Is Also an Exoribonuclease That Acts on Single-stranded RNA. J Biol Chem 2015; 290: 13344-13353
- 128 Orebaugh CD, Fye JM, Harvey S. et al. The TREX1 C-terminal region controls cellular localization through ubiquitination. J Biol Chem 2013; 288: 28881-28892
- 129 Barefield C, Karlseder J. The BLM helicase contributes to telomere maintenance through processing of late-replicating intermediate structures. Nucleic Acids Res 2012; 40: 7358-7367
- 130 Xue C, Daley JM, Xue X. et al. Single-molecule visualization of human BLM helicase as it acts upon double- and single-stranded DNA substrates. Nucleic Acids Res 2019; 47: 11225-11237
- 131 Kamath-Loeb AS, Johansson E, Burgers PM. et al. Functional interaction between the Werner Syndrome protein and DNA polymerase delta. Proc Natl Acad Sci U S A 2000; 97: 4603-4608
- 132 Brzostek-Racine S, Gordon C, Van Scoy S. et al. The DNA damage response induces IFN. J Immunol 2011; 187: 5336-5345
- 133 Gul E, Sayar EH, Gungor B. et al. Type I IFN-related NETosis in ataxia telangiectasia and Artemis deficiency. J Allergy Clin Immunol 2018; 142: 246-257
- 134 Honjo S, Yokote K, Fujishiro T. et al. Early amelioration of insulin resistance and reduction of interleukin-6 in Werner syndrome using pioglitazone. J Am Geriatr Soc 2008; 56: 173-174
- 135 Aono K, Maezawa Y, Kato H. et al. Recommendations for cancer screening and surveillance in patients with Werner syndrome. Geriatr Gerontol Int 2024; 24: 1085-1087
- 136 Goh KJ, Chen JH, Rocha N. et al. Human pluripotent stem cell-based models suggest preadipocyte senescence as a possible cause of metabolic complications of Werner and Bloom Syndromes. Sci Rep 2020; 10: 7490
- 137 Sugrañes TA, Flanagan M, Thomas C. et al. Age of first cancer diagnosis and survival in Bloom syndrome. Genet Med 2022; 24: 1476-1484
- 138 Weedon MN, Ellard S, Prindle MJ. et al. An in-frame deletion at the polymerase active site of POLD1 causes a multisystem disorder with lipodystrophy. Nat Genet 2013; 45: 947-950
- 139 Palles C, Cazier JB, Howarth KM. et al. Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat Genet 2013; 45: 136-144
- 140 van Os NJ, Roeleveld N, Weemaes CM. et al. Health risks for ataxia-telangiectasia mutated heterozygotes: a systematic review, meta-analysis and evidence-based guideline. Clin Genet 2016; 90: 105-117
- 141 Connelly PJ, Smith N, Chadwick R. et al. Recessive mutations in the cancer gene Ataxia Telangiectasia Mutated (ATM), at a locus previously associated with metformin response, cause dysglycaemia and insulin resistance. Diabet Med 2016; 33: 371-375
- 142 Paulino TL, Rafael MN, Hix S. et al. Is age a risk factor for liver disease and metabolic alterations in ataxia Telangiectasia patients?. Orphanet J Rare Dis 2017; 12: 136
- 143 Lindahl H, Svensson E, Danielsson A. et al. The clinical spectrum of ataxia telangiectasia in a cohort in Sweden. Heliyon 2024; 10: e26073
- 144 Siddoo-Atwal C, Haas AL, Rosin MP. Elevation of interferon beta-inducible proteins in ataxia telangiectasia cells. Cancer Res 1996; 56: 443-447
- 145 Gratia M, Rodero MP, Conrad C. et al. Bloom syndrome protein restrains innate immune sensing of micronuclei by cGAS. J Exp Med 2019; 216: 1199-1213
- 146 Kate WD, Fanta M, Weinfeld M. Loss of the DNA repair protein, polynucleotide kinase/phosphatase, activates the type 1 interferon response independent of ionizing radiation. Nucleic Acids Res 2024; 52: 9630-9653
- 147 Indulekha K, Surendar J, Mohan V. High sensitivity C-reactive protein, tumor necrosis factor-α, interleukin-6, and vascular cell adhesion molecule-1 levels in Asian Indians with metabolic syndrome and insulin resistance (CURES-105). J Diabetes Sci Technol 2011; 5: 982-988
- 148 Mitrou P, Lambadiari V, Maratou E. et al. Skeletal muscle insulin resistance in morbid obesity: the role of interleukin-6 and leptin. Exp Clin Endocrinol Diabetes 2011; 119: 484-489
- 149 Kern PA, Ranganathan S, Li C. et al. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol Endocrinol Metab 2001; 280: E745-E751
- 150 De Filippo G, Rendina D, Moccia F. et al. Interleukin-6, soluble interleukin-6 receptor/interleukin-6 complex and insulin resistance in obese children and adolescents. J Endocrinol Invest 2015; 38: 339-343
- 151 Almuraikhy S, Kafienah W, Bashah M. et al. Interleukin-6 induces impairment in human subcutaneous adipogenesis in obesity-associated insulin resistance. Diabetologia 2016; 59: 2406-2416
- 152 Tarkun I, Cetinarslan B, Türemen E. et al. Association between Circulating Tumor Necrosis Factor-Alpha, Interleukin-6, and Insulin Resistance in Normal-Weight Women with Polycystic Ovary Syndrome. Metab Syndr Relat Disord 2006; 4: 122-128
- 153 Vgontzas AN, Trakada G, Bixler EO. et al. Plasma interleukin 6 levels are elevated in polycystic ovary syndrome independently of obesity or sleep apnea. Metabolism 2006; 55: 1076-1082
- 154 Tangvarasittichai S, Pongthaisong S, Tangvarasittichai O. Tumor Necrosis Factor-Α, Interleukin-6, C-Reactive Protein Levels and Insulin Resistance Associated with Type 2 Diabetes in Abdominal Obesity Women. Indian J Clin Biochem 2016; 31: 68-74
- 155 Bruun JM, Verdich C, Toubro S. et al. Association between measures of insulin sensitivity and circulating levels of interleukin-8, interleukin-6 and tumor necrosis factor-alpha. Effect of weight loss in obese men. Eur J Endocrinol 2003; 148: 535-542
- 156 Agarwal A, Hegde A, Yadav C. et al. Association of Interleukin-6 and Myeloperoxidase with Insulin Resistance in Impaired Fasting Glucose Subjects. Indian J Clin Biochem 2017; 32: 33-38
- 157 Deepa R, Velmurugan K, Arvind K. et al. Serum levels of interleukin 6, C-reactive protein, vascular cell adhesion molecule 1, and monocyte chemotactic protein 1 in relation to insulin resistance and glucose intolerance--the Chennai Urban Rural Epidemiology Study (CURES). Metabolism 2006; 55: 1232-1238
- 158 Taniguchi A, Fukushima M, Ohya M. et al. Interleukin 6, adiponectin, leptin, and insulin resistance in nonobese Japanese type 2 diabetic patients. Metabolism 2006; 55: 258-262
- 159 Fischer CP, Perstrup LB, Berntsen A. et al. Elevated plasma interleukin-18 is a marker of insulin-resistance in type 2 diabetic and non-diabetic humans. Clin Immunol 2005; 117: 152-160
- 160 Shiny A, Regin B, Balachandar V. et al. Convergence of innate immunity and insulin resistance as evidenced by increased nucleotide oligomerization domain (NOD) expression and signaling in monocytes from patients with type 2 diabetes. Cytokine 2013; 64: 564-570
- 161 Gillies N, Pendharkar SA, Asrani VM. et al. Interleukin-6 is associated with chronic hyperglycemia and insulin resistance in patients after acute pancreatitis. Pancreatology 2016; 16: 748-755
- 162 Bi X, Ai H, Wu Q. et al. Insulin Resistance Is Associated with Interleukin 1β (IL-1β) in Non-Diabetic Hemodialysis Patients. Med Sci Monit 2018; 24: 897-902
- 163 Dogra M, Jaggi S, Aggarwal D. et al. Role of interleukin-6 and insulin resistance as screening markers for metabolic syndrome in patients of chronic obstructive pulmonary disease. A hospital-based cross-sectional study. Monaldi Arch Chest Dis 2021; 92
- 164 Rotter V, Nagaev I, Smith U. Interleukin-6 (IL-6) induces insulin resistance in 3T3-L1 adipocytes and is, like IL-8 and tumor necrosis factor-alpha, overexpressed in human fat cells from insulin-resistant subjects. J Biol Chem 2003; 278: 45777-45784
- 165 Bugge A, El-Naaman B, McMurray RG. et al. Inflammatory markers and clustered cardiovascular disease risk factors in Danish adolescents. Horm Res Paediatr 2012; 78: 288-296
- 166 Spranger J, Kroke A, Möhlig M. et al. 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: 812-817
- 167 Ridker PM, Libby P, MacFadyen JG. et al. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS). Eur Heart J 2018; 39: 3499-3507
- 168 Luque-Ramírez M, Escobar-Morreale HF. Treatment of polycystic ovary syndrome (PCOS) with metformin ameliorates insulin resistance in parallel with the decrease of serum interleukin-6 concentrations. Horm Metab Res 2010; 42: 815-820
- 169 Sergi D, Sanz JM, Lazzer S. et al. Interleukin-18 Is a Potential Biomarker Linking Dietary Fatty Acid Quality and Insulin Resistance: Results from a Cross-Sectional Study in Northern Italy. Nutrients 2023; 15: 1782
- 170 Hung J, McQuillan BM, Chapman CM. et al. Elevated interleukin-18 levels are associated with the metabolic syndrome independent of obesity and insulin resistance. Arterioscler Thromb Vasc Biol 2005; 25: 1268-1273
- 171 Kabakchieva P, Gateva A, Velikova T. et al. Elevated levels of interleukin-18 are associated with several indices of general and visceral adiposity and insulin resistance in women with polycystic ovary syndrome. Arch Endocrinol Metab 2022; 66: 3-11
- 172 Zhang YF, Yang YS, Hong J. et al. Elevated serum levels of interleukin-18 are associated with insulin resistance in women with polycystic ovary syndrome. Endocrine 2006; 29: 419-423
- 173 Flisiak-Jackiewicz M, Bobrus-Chociej A, Tarasów E. et al. Predictive Role of Interleukin-18 in Liver Steatosis in Obese Children. Can J Gastroenterol Hepatol 2018; 2018: 3870454
- 174 Leick L, Lindegaard B, Stensvold D. et al. Adipose tissue interleukin-18 mRNA and plasma interleukin-18: effect of obesity and exercise. Obesity (Silver Spring) 2007; 15: 356-363
- 175 Bruun JM, Stallknecht B, Helge JW. et al. Interleukin-18 in plasma and adipose tissue: effects of obesity, insulin resistance, and weight loss. Eur J Endocrinol 2007; 157: 465-471
- 176 Lee HM, Kim JJ, Kim HJ. et al. Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes 2013; 62: 194-204
- 177 Dalmas E, Venteclef N, Caer C. et al. T cell-derived IL-22 amplifies IL-1β-driven inflammation in human adipose tissue: relevance to obesity and type 2 diabetes. Diabetes 2014; 63: 1966-1977
- 178 Ridker PM, Everett BM, Thuren T. et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med 2017; 377: 1119-1131
- 179 Everett BM, Donath MY, Pradhan AD. et al. Anti-Inflammatory Therapy With Canakinumab for the Prevention and Management of Diabetes. J Am Coll Cardiol 2018; 71: 2392-2401
- 180 Adeva-Andany MM, Carneiro-Freire N, Castro-Quintela E. et al. Interferon Upregulation Associates with Insulin Resistance in Humans. Curr Diabetes Rev 2024; 21: E180324228061
- 181 Koivisto VA, Pelkonen R, Cantell K. Effect of interferon on glucose tolerance and insulin sensitivity. Diabetes 1989; 38: 641-647
- 182 Abdel-Moneim A, Semmler M, Abdel-Reheim ES. et al. Association of glycemic status and interferon-γ production with leukocytes and platelet indices alterations in type2 diabetes. Diabetes Metab Syndr 2019; 13: 1963-1969
- 183 Surendar J, Mohan V, Rao MM. et al. Increased levels of both Th1 and Th2 cytokines in subjects with metabolic syndrome (CURES-103). Diabetes Technol Ther 2011; 13: 477-482
- 184 Kalafati M, Kutmon M, Evelo CT. et al. An interferon-related signature characterizes the whole blood transcriptome profile of insulin-resistant individuals-the CODAM study. Genes Nutr 2021; 16: 22
- 185 McGillicuddy FC, Chiquoine EH, Hinkle CC. et al. Interferon gamma attenuates insulin signaling, lipid storage, and differentiation in human adipocytes via activation of the JAK/STAT pathway. J Biol Chem 2009; 284: 31936-31944
- 186 Bradley D, Smith AJ, Blaszczak A. et al. Interferon gamma mediates the reduction of adipose tissue regulatory T cells in human obesity. Nat Commun 2022; 13: 5606
- 187 Lin TY, Chiu CJ, Kuan CH. et al. IL-29 promoted obesity-induced inflammation and insulin resistance. Cell Mol Immunol 2020; 17: 369-379
- 188 Lagathu C, Yvan-Charvet L, Bastard JP. et al. Long-term treatment with interleukin-1beta induces insulin resistance in murine and human adipocytes. Diabetologia 2006; 49: 2162-2173
- 189 Gao D, Madi M, Ding C. et al. Interleukin-1β mediates macrophage-induced impairment of insulin signaling in human primary adipocytes. Am J Physiol Endocrinol Metab 2014; 307: E289-E304
- 190 Zhang HS, Du GY, Zhang ZG. et al. NRF2 facilitates breast cancer cell growth via HIF1α-mediated metabolic reprogramming. Int J Biochem Cell Biol 2018; 95: 85-92
- 191 Shamis SAK, McMillan DC, Edwards J. The relationship between hypoxia-inducible factor 1α (HIF-1α) and patient survival in breast cancer: Systematic review and meta-analysis. Crit Rev Oncol Hematol 2021; 159: 103231
- 192 Zhao Z, Mu H, Li Y. et al. Clinicopathological and prognostic value of hypoxia-inducible factor-1α in breast cancer: a meta-analysis including 5177 patients. Clin Transl Oncol 2020; 22: 1892-1906
- 193 Lu Z, Bi Y, Jiang J. et al. Exploring the prognostic and therapeutic value of HIF1A in lung adenocarcinoma. Heliyon 2024; 10: e37739
- 194 Yang SL, Ren QG, Wen L. et al. Clinicopathological and prognostic significance of hypoxia-inducible factor-1 alpha in lung cancer: a systematic review with meta-analysis. J Huazhong Univ Sci Technolog Med Sci 2016; 36: 321-327
- 195 Ren W, Mi D, Yang K. et al. The expression of hypoxia-inducible factor-1α and its clinical significance in lung cancer: a systematic review and meta-analysis. Swiss Med Wkly 2013; 143: w13855
- 196 Ping W, Jiang WY, Chen WS. et al. Expression and significance of hypoxia inducible factor-1α and lysyl oxidase in non-small cell lung cancer. Asian Pac J Cancer Prev 2013; 14: 3613-3618
- 197 Yohena T, Yoshino I, Takenaka T. et al. Upregulation of hypoxia-inducible factor-1alpha mRNA and its clinical significance in non-small cell lung cancer. J Thorac Oncol 2009; 4: 284-290
- 198 Giatromanolaki A, Koukourakis MI, Sivridis E. et al. Relation of hypoxia inducible factor 1 alpha and 2 alpha in operable non-small cell lung cancer to angiogenic/molecular profile of tumours and survival. Br J Cancer 2001; 85: 881-890
- 199 Zhang J, Wu Y, Lin YH. et al. Prognostic value of hypoxia-inducible factor-1 alpha and prolyl 4-hydroxylase beta polypeptide overexpression in gastric cancer. World J Gastroenterol 2018; 24: 2381-2391
- 200 Chen L, Shi Y, Yuan J. et al. HIF-1 alpha overexpression correlates with poor overall survival and disease-free survival in gastric cancer patients post-gastrectomy. PLoS One 2014; 9: e90678
- 201 Lin S, Ma R, Zheng XY. et al. Meta-analysis of immunohistochemical expression of hypoxia inducible factor-1α as a prognostic role in gastric cancer. World J Gastroenterol 2014; 20: 1107-1113
- 202 Chen J, Li T, Liu Q. et al. Clinical and prognostic significance of HIF-1alpha, PTEN, CD44v6, and survivin for gastric cancer: a meta-analysis. PLoS One 2014; 9: e91842
- 203 Zhu CL, Huang Q, Liu CH. et al. Prognostic value of HIF-1α expression in patients with gastric cancer. Mol Biol Rep 2013; 40: 6055-6062
- 204 Shibaji T, Nagao M, Ikeda N. et al. Prognostic significance of HIF-1 alpha overexpression in human pancreatic cancer. Anticancer Res 2003; 23: 4721-4727
- 205 Zoa A, Yang Y, Huang W. et al. High expression of hypoxia-inducible factor 1-alpha predicts poor prognosis in pancreatic ductal adenocarcinoma: a meta-analysis and database validation protocol. Transl Cancer Res 2022; 11: 3080-3091
- 206 Pipinikas CP, Carter ND, Corbishley CM. et al. HIF-1alpha mRNA gene expression levels in improved diagnosis of early stages of prostate cancer. Biomarkers 2008; 13: 680-691
- 207 Thomas R, Kim MH. HIF-1 alpha: a key survival factor for serum-deprived prostate cancer cells. Prostate 2008; 68: 1405-1415
- 208 Durślewicz J, Wybierała AM, Szczepanek S. et al. RUVBL1 in Clear-Cell Renal Cell Carcinoma: Unraveling Prognostic Significance and Correlation with HIF1A. Cancers (Basel) 2024; 16: 1273
- 209 Ding ZN, Dong ZR, Chen ZQ. et al. Effects of hypoxia-inducible factor-1α and hypoxia-inducible factor-2α overexpression on hepatocellular carcinoma survival: A systematic review with meta-analysis. J Gastroenterol Hepatol 2021; 36: 1487-1496
- 210 Yang SL, Liu LP, Jiang JX. et al. The correlation of expression levels of HIF-1α and HIF-2α in hepatocellular carcinoma with capsular invasion, portal vein tumor thrombi and patients' clinical outcome. Jpn J Clin Oncol 2014; 44: 159-167
- 211 Sivridis E, Giatromanolaki A, Gatter KC. et al. Association of hypoxia-inducible factors 1alpha and 2alpha with activated angiogenic pathways and prognosis in patients with endometrial carcinoma. Cancer 2002; 95: 1055-1063
- 212 Zhao YX, Yang Z, Ma LB. et al. HIF1A overexpression predicts the high lymph node metastasis risk and indicates a poor prognosis in papillary thyroid cancer. Heliyon 2023; 9: e14714
- 213 Lin X, Su H, Huo J. et al. The association of hypoxia-inducible factor-1α and hypoxia-inducible factor-2α protein expression with clinicopathological characteristics in papillary thyroid carcinoma: A meta-analysis. Medicine (Baltimore) 2023; 102: e34045
- 214 Liu YM, Ying SP, Huang YR. et al. Expression of HIF-1α and HIF-2α correlates to biological and clinical significance in papillary thyroid carcinoma. World J Surg Oncol 2016; 14: 30
- 215 Shao N, Han Y, Song L. et al. Clinical significance of hypoxia-inducible factor 1α, and its correlation with p53 and vascular endothelial growth factor expression in resectable esophageal squamous cell carcinoma. J Cancer Res Ther 2020; 16: 269-275
- 216 Zhao X, Tang YP, Wang CY. et al. Prognostic values of STAT3 and HIF-1alpha in esophageal squamous cell carcinoma. Eur Rev Med Pharmacol Sci 2019; 23: 3351-3357
- 217 Ping W, Sun W, Zu Y. et al. Clinicopathological and prognostic significance of hypoxia-inducible factor-1α in esophageal squamous cell carcinoma: a meta-analysis. Tumour Biol 2014; 35: 4401-4409
- 218 Zhang L, Ye SB, Li ZL. et al. Increased HIF-1alpha expression in tumor cells and lymphocytes of tumor microenvironments predicts unfavorable survival in esophageal squamous cell carcinoma patients. Int J Clin Exp Pathol 2014; 7: 3887-3897
- 219 Sun G, Hu W, Lu Y. et al. A meta-analysis of HIF-1α and esophageal squamous cell carcinoma (ESCC) risk. Pathol Oncol Res 2013; 19: 685-693
- 220 Sumera S, Ali A, Yousafzai YM. et al. Overexpression of Hypoxia-Inducible Factor-1α and Its Relation with Aggressiveness and Grade of Oral Squamous Cell Carcinoma. Diagnostics (Basel) 2023; 13: 451
- 221 Zhou J, Huang S, Wang L. et al. Clinical and prognostic significance of HIF-1α overexpression in oral squamous cell carcinoma: a meta-analysis. World J Surg Oncol 2017; 15: 104
- 222 Eckert AW, Schutze A, Lautner MH. et al. HIF-1alpha is a prognostic marker in oral squamous cell carcinomas. Int J Biol Markers 2010; 25: 87-92
- 223 Kang FW, Gao Y, Que L. et al. Hypoxia-inducible factor-1α overexpression indicates poor clinical outcomes in tongue squamous cell carcinoma. Exp Ther Med 2013; 5: 112-118
- 224 Mohamed AA, Eble MJ, Dahl E. et al. Prognostic implications of HIF-1α expression in anal squamous cell carcinoma treated with intensity-modulated radiotherapy (IMRT). Clin Transl Radiat Oncol 2024; 49: 100853
- 225 Luo D, Ren H, Zhang W. et al. Clinicopathological and prognostic value of hypoxia-inducible factor-1α in patients with bone tumor: a systematic review and meta-analysis. J Orthop Surg Res 2019; 14: 56
- 226 Lin W, Yin CY, Yu Q. et al. Expression of glucose transporter-1, hypoxia inducible factor-1α and beclin-1 in head and neck cancer and their implication. Int J Clin Exp Pathol 2018; 11: 3708-3717
- 227 Gong L, Zhang W, Zhou J. et al. Prognostic value of HIFs expression in head and neck cancer: a systematic review. PLoS One 2013; 8: e75094
- 228 Koukourakis MI, Giatromanolaki A, Sivridis E. et al. Hypoxia-inducible factor (HIF1A and HIF2A), angiogenesis, and chemoradiotherapy outcome of squamous cell head-and-neck cancer. Int J Radiat Oncol Biol Phys 2002; 53: 1192-1202
- 229 Schrijvers ML, van der Laan BF, de Bock GH. et al. Overexpression of intrinsic hypoxia markers HIF1alpha and CA-IX predict for local recurrence in stage T1-T2 glottic laryngeal carcinoma treated with radiotherapy. Int J Radiat Oncol Biol Phys 2008; 72: 161-169
- 230 Huang Y, Yang Y, Chen X. et al. Downregulation of malic enzyme 3 facilitates progression of gastric carcinoma via regulating intracellular oxidative stress and hypoxia-inducible factor-1α stabilization. Cell Mol Life Sci 2024; 81: 375
- 231 Tseng CW, Kuo WH, Chan SH. et al. Transketolase Regulates the Metabolic Switch to Control Breast Cancer Cell Metastasis via the α-Ketoglutarate Signaling Pathway. Cancer Res 2018; 78: 2799-2812
- 232 Liu T, Zhao L, Chen W. et al. Inactivation of von Hippel-Lindau increases ovarian cancer cell aggressiveness through the HIF1α/miR-210/VMP1 signaling pathway. Int J Mol Med 2014; 33: 1236-1242
- 233 Zia MK, Rmali KA, Watkins G. et al. The expression of the von Hippel-Lindau gene product and its impact on invasiveness of human breast cancer cells. Int J Mol Med 2007; 20: 605-611
- 234 Karasaki Y, Kashiwazaki H. The difference of mutation in the peroxisome proliferator activated receptor gamma2 gene among people at high altitudes and low altitudes in Bolivia. J UOEH 2005; 27: 243-248
- 235 Wang B, Wood IS, Trayhurn P. Hypoxia induces leptin gene expression and secretion in human preadipocytes: differential effects of hypoxia on adipokine expression by preadipocytes. J Endocrinol 2008; 198: 127-134
- 236 Liu XL, Zhang FL, Zhou ZY. et al. Analysis of two sequence variants in peroxisome proliferator activated receptor gamma gene in Tajik population at high altitudes and Han population at low altitudes in China. Mol Biol Rep 2010; 37: 179-184
- 237 Li X, Kimura H, Hirota K. et al. Hypoxia reduces the expression and anti-inflammatory effects of peroxisome proliferator-activated receptor-gamma in human proximal renal tubular cells. Nephrol Dial Transplant 2007; 22: 1041-1051
- 238 Thorn CE, Knight B, Pastel E. et al. Adipose tissue is influenced by hypoxia of obstructive sleep apnea syndrome independent of obesity. Diabetes Metab 2017; 43: 240-247
- 239 El Amine Z, Mauger JF, Imbeault P. Human Preadipocytes Differentiated under Hypoxia following PCB126 Exposure during Proliferation: Effects on Differentiation, Glucose Uptake and Adipokine Profile. Cells 2023; 12: 2326
- 240 Foley JE, Kashiwagi A, Verso MA. et al. Improvement in in vitro insulin action after one month of insulin therapy in obese noninsulin-dependent diabetics. Measurements of glucose transport and metabolism, insulin binding, and lipolysis in isolated adipocytes. J Clin Invest 1983; 72: 1901-1909
- 241 Maianu L, Keller SR, Garvey WT. Adipocytes exhibit abnormal subcellular distribution and translocation of vesicles containing glucose transporter 4 and insulin-regulated aminopeptidase in type 2 diabetes mellitus: implications regarding defects in vesicle trafficking. J Clin Endocrinol Metab 2001; 86: 5450-5456
- 242 Hammarstedt A, Sopasakis VR, Gogg S. et al. Improved insulin sensitivity and adipose tissue dysregulation after short-term treatment with pioglitazone in non-diabetic, insulin-resistant subjects. Diabetologia 2005; 48: 96-104
- 243 Kampmann U, Christensen B, Nielsen TS. et al. GLUT4 and UBC9 protein expression is reduced in muscle from type 2 diabetic patients with severe insulin resistance. PLoS One 2011; 6: e27854
- 244 Agrawal M, Yeo CR, Shabbir A. et al. Fat storage-inducing transmembrane protein 2 (FIT2) is less abundant in type 2 diabetes, and regulates triglyceride accumulation and insulin sensitivity in adipocytes. FASEB J 2019; 33: 430-440
- 245 Koester AM, Geiser A, Bowman PRT. et al. GLUT4 translocation and dispersal operate in multiple cell types and are negatively correlated with cell size in adipocytes. Sci Rep 2022; 12: 20535
- 246 Bril F, Ezeh U, Amiri M. et al. Adipose Tissue Dysfunction in Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2023; 109: 10-24
- 247 Arner P, Arner E, Hammarstedt A. et al. Genetic predisposition for Type 2 diabetes, but not for overweight/obesity, is associated with a restricted adipogenesis. PLoS One 2011; 6: e18284
- 248 Karnieli E, Barzilai A, Rafaeloff R. et al. Distribution of glucose transporters in membrane fractions isolated from human adipose cells. Relation to cell size. J Clin Invest 1986; 78: 1051-1055
- 249 Catalano PM, Nizielski SE, Shao J. et al. Downregulated IRS-1 and PPARgamma in obese women with gestational diabetes: relationship to FFA during pregnancy. Am J Physiol Endocrinol Metab 2002; 282: E522-E533
- 250 Liao W, Nguyen MT, Yoshizaki T. et al. Suppression of PPAR-gamma attenuates insulin-stimulated glucose uptake by affecting both GLUT1 and GLUT4 in 3T3-L1 adipocytes. Am J Physiol Endocrinol Metab 2007; 293: E219-E227
- 251 Van Meijel RLJ, Wang P, Bouwman F. et al. The Effects of Mild Intermittent Hypoxia Exposure on the Abdominal Subcutaneous Adipose Tissue Proteome in Overweight and Obese Men: A First-in-Human Randomized, Single-Blind, and Cross-Over Study. Front Physiol 2021; 12: 791588
- 252 Adeva-Andany MM, Dominguez-Montero A, Castro-Quintela E. et al. Hypoxia-Induced Insulin Resistance Mediates the Elevated Cardiovascular Risk in Patients with Obstructive Sleep Apnea: A Comprehensive Review. Rev Cardiovasc Med 2024; 25: 231
- 253 Larsen JJ, Hansen JM, Olsen NV. et al The effect of altitude hypoxia on glucose homeostasis in men. J Physiol 1997; 504 Pt 1 241-249
- 254 Oltmanns KM, Gehring H, Rudolf S. et al. Hypoxia causes glucose intolerance in humans. Am J Respir Crit Care Med 2004; 169: 1231-1237
- 255 Louis M, Punjabi NM. Effects of acute intermittent hypoxia on glucose metabolism in awake healthy volunteers. J Appl Physiol (1985) 2009; 106: 1538-1544
