Adiponectin and Thyroid Cancer: Insight into the Association between Adiponectin and Obesity

Yuanyuan Zhou, Ying Yang, Taicheng Zhou, Bai Li, Zhanjian Wang, Yuanyuan Zhou, Ying Yang, Taicheng Zhou, Bai Li, Zhanjian Wang

Abstract

In recent decades, the incidence and diagnosis of thyroid cancer have risen dramatically, and thyroid cancer has now become the most common endocrine cancer in the world. The onset of thyroid cancer is insidious, and its progression is slow and difficult to detect. Therefore, early prevention and treatment have important strategic significance. Moreover, an in-depth exploration of the pathogenesis of thyroid cancer is key to early prevention and treatment. Substantial evidence supports obesity as an independent risk factor for thyroid cancer. Adipose tissue dysfunction in the obese state is accompanied by dysregulation of a variety of adipocytokines. Adiponectin (APN) is one of the most pivotal adipocytokines, and its connection with obesity and obesity-related disease has gradually become a hot topic in research. Recently, the association between APN and thyroid cancer has received increasing attention. The purpose of this review is to systematically review previous studies, give prominence to APN, focus on the relationship between APN, obesity and thyroid cancer, and uncover the underlying pathogenic mechanisms.

Keywords: adiponectin; obesity; thyroid cancer.

Conflict of interest statement

Conflicts of Interest The authors declare that they have no competing interests.

copyright: © 2021 Zhou et al.

Figures

Figure 1.
Figure 1.
The anti-inflammatory properties of APN. APN exerts its anti-inflammatory properties in different cell types through mechanisms as follows. In adipocytes, a bi-directional regulation has been shown to exist between APN and pro-inflammatory factors, such as TNF-α, IL-6, and CRP. APN treated macrophages exhibited impaired function of phagocytic activity, suppressed foam cell transformation, negative regulation of NF-κB signaling, decreased expression of TNF-α or IL-6, and increased release of IL-10 and IL-1RA. In endothelial cells, APN can negatively regulate the expression of VCAM-1, E-selectin, ICAM-1, CRP, and IL-8, and suppress endothelial NF-κB activation and ROS production. The red arrow indicates promotion, and the blue arrow indicates inhibition. Abbreviations: APN adiponectin, TNF-α tumor necrosis factor-α, IL-6 interleukin-6, CRP C-reactive protein, NF-κB nuclear factor kappa-B, IL-10 interleukin-10, IL-1RA interleukin-1 receptor agonist, VCAM-1 vascular cell adhesion molecule 1, ICAM-1 intracellular adhesion molecule 1, IL-8 interleukin-8, ROS reactive oxygen species.
Figure 2.
Figure 2.
The possible mechanisms underlying obesity and thyroid cancer. Elevated serum TSH and other obesity-related comorbidities, including abnormal insulin resistance, chronic low degree inflammation, oxidative stress, abnormalities of the IGF system, and altered secretion of adipokines, could be the possible carcinogenic mechanism underlying obesity and thyroid cancer. Moreover, all of these factors may interplay with each other, forming a vicious cycle. Abbreviations: TSH thyroid-stimulating hormone, IGF insulin-like growth factor.
Figure 3.
Figure 3.
The directly and indirectly anti-cancer pathways of APN. Binding to AdipoR1/2, APN could initiate a series of directly anti-cancer pathways, including inhibition of PI3K/AKT, ERK1/2, Wnt/GSK3β/β-catenin, STAT3, USP2, and Bcl-2 signals; as well as activation of AMPK by LKB1, then up-regulates TSC2, p53/p21/p27 and promotes cell autophagy. Moreover, the tumorigenicity of the IGF axis has been confirmed to be partially through ERK and PI3K/AKT pathways, which are belonging to the downstream signaling of the APN. Since APN can increase insulin sensitivity, it is reasonable to believe that the complexity of the APN-related signaling network has been added by the possible indirect association between APN, insulin resistance, and the abnormal IGF system. The blue solid arrows and red solid arrows indicate inhibitory and stimulatory effects in the direct pathway, respectively. In turn, the blue dotted lines and red dotted lines indicate the inhibitory and stimulatory effects in the indirect pathway, respectively. Abbreviations: APN adiponectin, PI3K/AKT phosphatidylinositol 3-kinase/protein kinase B, ERK1/2 extracellular signal-regulated kinases 1 and 2 Wnt wingless-type protein, GSK3β glycogen synthase kinase 3β, STAT3 signal transducer and activator of transcription 3, USP2 ubiquitin-specific protease 2, AMPK 5’ adenosine monophosphate-activated protein kinase, LKB1 liver kinase B1, TSC2 tuberous sclerosis complex 2, IGF insulin-like growth factor.

References

    1. Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Pineros M, et al.. (2019). Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer, 144:1941-53.
    1. Morris LG, Myssiorek D (2010). Improved detection does not fully explain the rising incidence of well-differentiated thyroid cancer: a population-based analysis. Am J Surg, 200:454-61.
    1. Pagano L, Mele C, Arpaia D, Sama MT, Caputo M, Ippolito S, et al.. (2017). How do etiological factors can explain the different clinical features of patients with differentiated thyroid cancer and their histopathological findings? Endocrine, 56:129-37.
    1. Patel HH, Goyal N, Goldenberg D (2014). Imaging, genetic testing, and biomarker assessment of follicular cell-derived thyroid cancer. Ann Med, 46:409-16.
    1. Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R (2013). Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol, 2013:965212.
    1. Rosai J (2004). Poorly differentiated thyroid carcinoma: introduction to the issue, its landmarks, and clinical impact. Endocr Pathol, 15:293-6.
    1. Roman S, Mehta P, Sosa JA (2009). Medullary thyroid cancer: early detection and novel treatments. Curr Opin Oncol, 21:5-10.
    1. Pappa T, Alevizaki M (2016). Management of hereditary medullary thyroid carcinoma. Endocrine, 53:7-17.
    1. McQueen AS, Bhatia KS (2015). Thyroid nodule ultrasound: technical advances and future horizons. Insights Imaging, 6:173-88.
    1. Rahbari R, Zhang L, Kebebew E (2010). Thyroid cancer gender disparity. Future Oncol, 6:1771-9.
    1. Baverstock K, Williams D (2006). The chernobyl accident 20 years on: an assessment of the health consequences and the international response. Environ Health Perspect, 114:1312-7.
    1. Williams D (2008). Twenty years' experience with post-Chernobyl thyroid cancer. Best Pract Res Clin Endocrinol Metab, 22:1061-73.
    1. Zimmermann MB, Galetti V (2015). Iodine intake as a risk factor for thyroid cancer: a comprehensive review of animal and human studies. Thyroid Res, 8:8.
    1. Santoro M, Grieco M, Melillo RM, Fusco A, Vecchio G (1995). Molecular defects in thyroid carcinomas: role of the RET oncogene in thyroid neoplastic transformation. Eur J Endocrinol, 133:513-22.
    1. Yamashita S, Mitsutake N (2007). Molecular mechanism of thyroid carcinogenesis. Nihon Rinsho, 65:1959-65.
    1. Bonora E, Tallini G, Romeo G (2010). Genetic Predisposition to Familial Nonmedullary Thyroid Cancer: An Update of Molecular Findings and State-of-the-Art Studies. J Oncol, 2010:385206.
    1. D'Avanzo B, La Vecchia C, Franceschi S, Negri E, Talamini R (1995). History of thyroid diseases and subsequent thyroid cancer risk. Cancer Epidemiol Biomarkers Prev, 4:193-9.
    1. Kitahara CM, Farkas DKR, Jorgensen JOL, Cronin-Fenton D, Sorensen HT (2018). Benign thyroid diseases and risk of thyroid cancer: a nationwide cohort study. J Clin Endocrinol Metab, 103:2216-24.
    1. NCD Risk Factor Collaboration (NCD-RisC) (2016). Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet, 387:1377-96.
    1. Mokdad AH, Forouzanfar MH, Daoud F, Mokdad AA, El Bcheraoui C, Moradi-Lakeh M, et al.. (2016). Global burden of diseases, injuries, and risk factors for young people's health during 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet, 387:2383-401.
    1. Kim R, Lee DH, Subramanian SV (2019). Understanding the obesity epidemic. BMJ, 366:l4409.
    1. Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M (2008). Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet, 371:569-78.
    1. Pazaitou-Panayiotou K, Polyzos SA, Mantzoros CS (2013). Obesity and thyroid cancer: epidemiologic associations and underlying mechanisms. Obes Rev, 14:1006-22.
    1. Marcello MA, Cunha LL, Batista FA, Ward LS (2014). Obesity and thyroid cancer. Endocr Relat Cancer, 21:T255-71.
    1. Santini F, Marzullo P, Rotondi M, Ceccarini G, Pagano L, Ippolito S, et al.. (2014). Mechanisms in endocrinology: the crosstalk between thyroid gland and adipose tissue: signal integration in health and disease. Eur J Endocrinol, 171:R137-52.
    1. Luo L, Liu M (2016). Adipose tissue in control of metabolism. J Endocrinol, 231:R77-99.
    1. Smitka K, Maresova D (2015). Adipose Tissue as an Endocrine Organ: An Update on Pro-inflammatory and Anti-inflammatory Microenvironment. Prague Med Rep, 116:87-111.
    1. Kajimura S (2017). Adipose tissue in 2016: Advances in the understanding of adipose tissue biology. Nat Rev Endocrinol, 13:69-70.
    1. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF (1995). A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem, 270:26746-9.
    1. Villarreal-Molina MT, Antuna-Puente B (2012). Adiponectin: anti-inflammatory and cardioprotective effects. Biochimie, 94:2143-9.
    1. Bobbert P, Scheibenbogen C, Jenke A, Kania G, Wilk S, Krohn S, et al.. (2011). Adiponectin expression in patients with inflammatory cardiomyopathy indicates favourable outcome and inflammation control. Eur Heart J, 32:1134-47.
    1. Jortay J, Senou M, Abou-Samra M, Noel L, Robert A, Many MC, et al.. (2012). Adiponectin and skeletal muscle: pathophysiological implications in metabolic stress. Am J Pathol, 181:245-56.
    1. Crawford LJ, Peake R, Price S, Morris TC, Irvine AE (2010). Adiponectin is produced by lymphocytes and is a negative regulator of granulopoiesis. J Leukoc Biol, 88:807-11.
    1. Wong GW, Wang J, Hug C, Tsao TS, Lodish HF (2004). A family of Acrp30/adiponectin structural and functional paralogs. Proc Natl Acad Sci U S A, 101:10302-7.
    1. Wang Y, Lam KS, Yau MH, Xu A (2008). Post-translational modifications of adiponectin: mechanisms and functional implications. Biochem J, 409:623-33.
    1. Takemura Y, Ouchi N, Shibata R, Aprahamian T, Kirber MT, Summer RS, et al.. (2007). Adiponectin modulates inflammatory reactions via calreticulin receptor-dependent clearance of early apoptotic bodies. J Clin Invest, 117:375-86.
    1. Pajvani UB, Hawkins M, Combs TP, Rajala MW, Doebber T, Berger JP, et al.. (2004). Complex distribution, not absolute amount of adiponectin, correlates with thiazolidinedione-mediated improvement in insulin sensitivity. J Biol Chem, 279:12152-62.
    1. Fruebis J, Tsao TS, Javorschi S, Ebbets-Reed D, Erickson MR, Yen FT, et al.. (2001). Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc Natl Acad Sci U S A, 98:2005-10.
    1. Yamauchi T, Iwabu M, Okada-Iwabu M, Kadowaki T (2014). Adiponectin receptors: a review of their structure, function and how they work. Best Pract Res Clin Endocrinol Metab, 28:15-23.
    1. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, et al.. (2003). Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature, 423:762-9.
    1. Goldstein BJ, Scalia R (2004). Adiponectin: A novel adipokine linking adipocytes and vascular function. J Clin Endocrinol Metab, 89:2563-8.
    1. Ouchi N, Walsh K (2007). Adiponectin as an anti-inflammatory factor. Clin Chim Acta, 380:24-30.
    1. Achari AE, Jain SK (2017). Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int J Mol Sci, 18:1321.
    1. Balsan GA, Vieira JL, Oliveira AM, Portal VL (2015). Relationship between adiponectin, obesity and insulin resistance. Rev Assoc Med Bras (1992), 61:72-80.
    1. Li JM, Lu W, Ye J, Han Y, Chen H, Wang LS (2020). Association between expression of AMPK pathway and adiponectin, leptin, and vascular endothelial function in rats with coronary heart disease. Eur Rev Med Pharmacol Sci, 24:905-14.
    1. Katsiki N, Mantzoros C, Mikhailidis DP (2017). Adiponectin, lipids and atherosclerosis. Curr Opin Lipidol, 28:347-54.
    1. Tilg H, Moschen AR (2006). Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol, 6:772-83.
    1. Polito R, Nigro E, Messina A, Monaco ML, Monda V, Scudiero O, et al.. (2018). Adiponectin and Orexin-A as a Potential Immunity Link Between Adipose Tissue and Central Nervous System. Front Physiol, 9:982.
    1. Solinas G, Becattini B (2017). JNK at the crossroad of obesity, insulin resistance, and cell stress response. Mol Metab, 6:174-84.
    1. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, et al.. (2003). Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest, 112:1821-30.
    1. Breznik JA, Naidoo A, Foley KP, Schulz C, Lau TC, Loukov D, et al.. (2018). TNF, but not hyperinsulinemia or hyperglycemia, is a key driver of obesity-induced monocytosis revealing that inflammatory monocytes correlate with insulin in obese male mice. Physiol Rep, 6:e13937.
    1. Sierra-Honigmann MR, Nath AK, Murakami C, García-Cardeña G, Papapetropoulos A, Sessa WC, et al.. (1998). Biological action of leptin as an angiogenic factor. Science, 281:1683-6.
    1. Wellen KE, Hotamisligil GS (2003). Obesity-induced inflammatory changes in adipose tissue. J Clin Invest, 112:1785-8.
    1. Engin AB (2017). Adipocyte-Macrophage Cross-Talk in Obesity. Adv Exp Med Biol, 960:327-43.
    1. Guzik TJ, Skiba DS, Touyz RM, Harrison DG (2017). The role of infiltrating immune cells in dysfunctional adipose tissue. Cardiovasc Res, 113:1009-23.
    1. Maurizi G, Guardia L, Maurizi A, Poloni A (2018). Adipocytes properties and crosstalk with immune system in obesity-related inflammation. J Cell Physiol, 233:88-97.
    1. Thomas D, Apovian C (2017). Macrophage functions in lean and obese adipose tissue. Metabolism, 72:120-43.
    1. Yudkin JS, Stehouwer CD, Emeis JJ, Coppack SW (1999). C-reactive protein in healthy subjects: associations with obesity, insulin resistance, and endothelial dysfunction: a potential role for cytokines originating from adipose tissue? Arterioscler Thromb Vasc Biol, 19:972-8.
    1. Menezes AMB, Oliveira PD, Wehrmeister FC, Gonçalves H, Assunção MCF, Tovo-Rodrigues L, et al.. (2018). Association between interleukin-6, C-reactive protein and adiponectin with adiposity: Findings from the 1993 pelotas (Brazil) birth cohort at 18 and 22 years. Cytokine, 110:44-51.
    1. Tangvarasittichai S, Pongthaisong S, Tangvarasittichai O (2016). 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, 31:68-74.
    1. Bahceci M, Gokalp D, Bahceci S, Tuzcu A, Atmaca S, Arikan S (2007). The correlation between adiposity and adiponectin, tumor necrosis factor alpha, interleukin-6 and high sensitivity C-reactive protein levels. Is adipocyte size associated with inflammation in adults? J Endocrinol Invest, 30:210-4.
    1. Zohmangaihi D, Sharma SB, Madhu SV (2019). Adiponectin, IL-6 and hsCRP: interplay of inflammation with obesity and type 2 diabetes in indian population. J Diabetes Metab, 10:822.
    1. Ajuwon KM, Spurlock ME (2005). Adiponectin inhibits LPS-induced NF-kappaB activation and IL-6 production and increases PPARgamma2 expression in adipocytes. Am J Physiol Regul Integr Comp Physiol, 288:R1220-5.
    1. Maeda N, Shimomura I, Kishida K, Nishizawa H, Matsuda M, Nagaretani H, et al.. (2002). Diet-induced insulin resistance in mice lacking adiponectin/ACRP30. Nat Med, 8:731-7.
    1. Yuan G, Chen X, Ma Q, Qiao J, Li R, Li X, et al.. (2007). C-reactive protein inhibits adiponectin gene expression and secretion in 3T3-L1 adipocytes. J Endocrinol, 194:275-81.
    1. Kappes A, Loffler G (2000). Influences of ionomycin, dibutyryl-cycloAMP and tumour necrosis factor-alpha on intracellular amount and secretion of apM1 in differentiating primary human preadipocytes. Horm Metab Res, 32:548-54.
    1. Fasshauer M, Kralisch S, Klier M, Lossner U, Bluher M, Klein J, et al.. (2003). Adiponectin gene expression and secretion is inhibited by interleukin-6 in 3T3-L1 adipocytes. Biochem Biophys Res Commun, 301:1045-50.
    1. Luo Y, Liu M (2016). Adiponectin: a versatile player of innate immunity. J Mol Cell Biol, 8:120-8.
    1. Yokota T, Oritani K, Takahashi I, Ishikawa J, Matsuyama A, Ouchi N, et al.. (2000). Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood, 96:1723-32.
    1. Furukawa K, Hori M, Ouchi N, Kihara S, Funahashi T, Matsuzawa Y, et al.. (2004). Adiponectin down-regulates acyl-coenzyme A: cholesterol acyltransferase-1 in cultured human monocyte-derived macrophages. Biochem Biophys Res Commun, 317:831-6.
    1. Yamaguchi N, Argueta JG, Masuhiro Y, Kagishita M, Nonaka K, Saito T, et al.. (2005). Adiponectin inhibits Toll-like receptor family-induced signaling. FEBS Lett, 579:6821-6.
    1. Wolf AM, Wolf D, Rumpold H, Enrich B, Tilg H (2004). Adiponectin induces the anti-inflammatory cytokines IL-10 and IL-1RA in human leukocytes. Biochem Biophys Res Commun, 323:630-5.
    1. Ouchi N, Kihara S, Arita Y, Maeda K, Kuriyama H, Okamoto Y, et al.. (1999). Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation, 100:2473-6.
    1. Devaraj S, Torok N, Dasu MR, Samols D, Jialal I (2008). Adiponectin decreases C-reactive protein synthesis and secretion from endothelial cells: evidence for an adipose tissue-vascular loop. Arterioscler Thromb Vasc Biol, 28:1368-74.
    1. Kobashi C, Urakaze M, Kishida M, Kibayashi E, Kobayashi H, Kihara S, et al.. (2005). Adiponectin inhibits endothelial synthesis of interleukin-8. Circ Res, 97:1245-52.
    1. Ouchi N, Kihara S, Arita Y, Okamoto Y, Maeda K, Kuriyama H, et al.. (2000). Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-kappaB signaling through a cAMP-dependent pathway. Circulation, 102:1296-301.
    1. Ouedraogo R, Wu X, Xu SQ, Fuchsel L, Motoshima H, Mahadev K, et al.. (2006). Adiponectin suppression of high-glucose-induced reactive oxygen species in vascular endothelial cells: evidence for involvement of a cAMP signaling pathway. Diabetes, 55:1840-6.
    1. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, et al.. (2012). Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun, 425:560-4.
    1. Yang WS, Lee WJ, Funahashi T, Tanaka S, Matsuzawa Y, Chao CL, et al.. (2001). Weight reduction increases plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab, 86:3815-9.
    1. Abdella NA, Mojiminiyi OA (2018). Clinical Applications of Adiponectin Measurements in Type 2 Diabetes Mellitus: Screening, Diagnosis, and Marker of Diabetes Control. Dis Markers, 2018:5187940.
    1. Yadav A, Kataria MA, Saini V, Yadav A (2013). Role of leptin and adiponectin in insulin resistance. Clin Chim Acta, 417:80-4.
    1. Wu Q, Chen Y, Chen S, Wu X, Nong W (2020). Correlation between adiponectin, chemerin, vascular endothelial growth factor and epicardial fat volume in patients with coronary artery disease. Exp Ther Med, 19:1095-102.
    1. Wu CC, Chang CS, Hsu CC, Wang CP, Tsai IT, Lu YC, et al.. (2020). Elevated Plasma Adiponectin Levels Are Associated with Abnormal Corrected QT Interval in Patients with Stable Angina. Int Heart J, 61:29-38.
    1. Kim DH, Kim C, Ding EL, Townsend MK, Lipsitz LA (2013). Adiponectin levels and the risk of hypertension: a systematic review and meta-analysis. Hypertension, 62:27-32.
    1. Rasmussen MS, Lihn AS, Pedersen SB, Bruun JM, Rasmussen M, Richelsen B (2006). Adiponectin receptors in human adipose tissue: effects of obesity, weight loss, and fat depots. Obesity, 14:28-35.
    1. Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K (2016). Body Fatness and Cancer--Viewpoint of the IARC Working Group. N Engl J Med, 375:794-8.
    1. Gu L, Cao C, Fu J, Li Q, Li DH, Chen MY (2018). Serum adiponectin in breast cancer: a meta-analysis. Medicine, 97:e11433.
    1. Yu Z, Tang S, Ma H, Duan H, Zeng Y (2019). Association of serum adiponectin with breast cancer: a meta-analysis of 27 case-control studies. Medicine, 98:e14359.
    1. Li ZJ, Yang XL, Yao Y, Han WQ, Li BO (2016). Circulating adiponectin levels and risk of endometrial cancer: Systematic review and meta-analysis. Exp Ther Med, 11:2305-13.
    1. Lin T, Zhao X, Kong WM (2015). Association between adiponectin levels and endometrial carcinoma risk: evidence from a dose-response meta-analysis. BMJ Open, 5:e008541.
    1. Gong TT, Wu QJ, Wang YL, Ma XX (2015). Circulating adiponectin, leptin and adiponectin-leptin ratio and endometrial cancer risk: Evidence from a meta-analysis of epidemiologic studies. Int J Cancer, 137:1967-78.
    1. Busch EL, Crous-Bou M, Prescott J, Downing MJ, Rosner BA, Mutter GL, et al.. (2018). Adiponectin, Leptin, and Insulin-Pathway Receptors as Endometrial Cancer Subtyping Markers. Horm Cancer, 9:33-9.
    1. Wang XH, Zhang Y, Liu LZ, Shang CG (2018). Effects of metformin and adiponectin on endometrial cancer cells growth. Beijing Da Xue Xue Bao Yi Xue Ban, 50:767-73.
    1. Hoffmann M, Gogola J, Ptak A (2018). Adiponectin Reverses the Proliferative Effects of Estradiol and IGF-1 in Human Epithelial Ovarian Cancer Cells by Downregulating the Expression of Their Receptors. Horm Cancer, 9:166-74.
    1. Slomian GJ, Nowak D, Buczkowska M, Glogowska-Gruszka A, Slomian SP, Roczniak W, et al.. (2019). The role of adiponectin and leptin in the treatment of ovarian cancer patients. Endokrynol Pol, 70:57-63.
    1. Hu X, Hu C, Zhang C, Zhang M, Long S, Cao Z (2019). Role of Adiponectin in prostate cancer. Int Braz J Urol, 45:220-8.
    1. Angel CZ, Iguacel I, Mullee A, Guha N, Wasson R, McKenna DJ, et al.. (2019). Appetite-regulating hormones-leptin, adiponectin and ghrelin-and the development of prostate cancer: a systematic review and exploratory meta-analysis. Prostate Cancer Prostatic Dis, 23:11-23.
    1. Schmid D, Ricci C, Behrens G, Leitzmann MF (2015). Adiposity and risk of thyroid cancer: a systematic review and meta-analysis. Obes Rev, 16:1042-54.
    1. Kitahara CM, Pfeiffer RM, Sosa JA, Shiels MS (2019). Impact of overweight and obesity on U.S. papillary thyroid cancer incidence trends (1995-2015). J Natl Cancer Inst, 112:810-7.
    1. Ma J, Huang M, Wang L, Ye W, Tong Y, Wang H (2015). Obesity and risk of thyroid cancer: evidence from a meta-analysis of 21 observational studies. Med Sci Monit, 21:283-91.
    1. He Q, Sun H, Li F, Liang N (2019). Obesity and risk of differentiated thyroid cancer: A large-scale case-control study. Clin Endocrinol (Oxf), 91:869-78.
    1. Zhao S, Jia X, Fan X, Zhao L, Pang P, Wang Y, et al.. (2019). Association of obesity with the clinicopathological features of thyroid cancer in a large, operative population: A retrospective case-control study. Medicine (Baltimore), 98:e18213.
    1. Jin QF, Fang QG, Qi JX, Li P (2019). Impact of BMI on Complications and Satisfaction in Patients With Papillary Thyroid Cancer and Lateral Neck Metastasis. Cancer Control, 26:1073274819853831.
    1. Son H, Lee H, Kang K, Lee I (2018). The risk of thyroid cancer and obesity: A nationwide population-based study using the Korea National Health Insurance Corporation cohort database. Surg Oncol, 27:166-71.
    1. Engeland A, Tretli S, Akslen LA, Bjorge T (2006). Body size and thyroid cancer in two million Norwegian men and women. Br J Cancer, 95:366-70.
    1. Oh SW, Yoon YS, Shin SA (2005). Effects of excess weight on cancer incidences depending on cancer sites and histologic findings among men: Korea National Health Insurance Corporation Study. J Clin Oncol, 23:4742-54.
    1. Kitahara CM, Platz EA, Freeman LE, Hsing AW, Linet MS, Park Y, et al.. (2011). Obesity and thyroid cancer risk among U.S. men and women: a pooled analysis of five prospective studies. Cancer Epidemiol Biomarkers Prev, 20:464-72.
    1. Kwon H, Han KD, Park CY (2019). Weight change is significantly associated with risk of thyroid cancer: A nationwide population-based cohort study. Sci Rep, 9:1546.
    1. Leitzmann MF, Brenner A, Moore SC, Koebnick C, Park Y, Hollenbeck A, et al.. (2010). Prospective study of body mass index, physical activity and thyroid cancer. Int J Cancer, 126:2947-56.
    1. Ali A, Mirza Y, Faizan U, Zahid N, Awan MS (2018). Association of Obesity and Thyroid Cancer at a Tertiary Care Hospital in Pakistan. Cureus, 10:e2364.
    1. Rinaldi S, Lise M, Clavel-Chapelon F, Boutron-Ruault MC, Guillas G, Overvad K, et al.. (2012). Body size and risk of differentiated thyroid carcinomas: findings from the EPIC study. Int J Cancer, 131:E1004-14.
    1. Han JM, Kim TY, Jeon MJ, Yim JH, Kim WG, Song DE, et al.. (2013). Obesity is a risk factor for thyroid cancer in a large, ultrasonographically screened population. Eur J Endocrinol, 168:879-86.
    1. Brindel P, Doyon F, Rachedi F, Boissin JL, Sebbag J, Shan L, et al.. (2009). Anthropometric factors in differentiated thyroid cancer in French Polynesia: a case-control study. Cancer Causes Control, 20:581-90.
    1. Clavel-Chapelon F, Guillas G, Tondeur L, Kernaleguen C, Boutron-Ruault MC (2010). Risk of differentiated thyroid cancer in relation to adult weight, height and body shape over life: the French E3N cohort. Int J Cancer, 126:2984-90.
    1. Roger PP, Dumont JE (1987). Thyrotropin is a potent growth factor for normal human thyroid cells in primary culture. Biochem Biophys Res Commun, 149:707-11.
    1. Pacifico L, Anania C, Ferraro F, Andreoli GM, Chiesa C (2012). Thyroid function in childhood obesity and metabolic comorbidity. Clin Chim Acta, 413:396-405.
    1. Kitahara CM, Platz EA, Ladenson PW, Mondul AM, Menke A, de González AB (2012). Body fatness and markers of thyroid function among U.S. men and women. PLoS One, 7:e34979.
    1. Aypak C, Türedi O, Yüce A, Görpelioğlu S (2013). Thyroid-stimulating hormone (TSH) level in nutritionally obese children and metabolic co-morbidity. J Pediatr Endocrinol Metab, 26:703-8.
    1. Ayturk S, Gursoy A, Kut A, Anil C, Nar A, Tutuncu NB (2009). Metabolic syndrome and its components are associated with increased thyroid volume and nodule prevalence in a mild-to-moderate iodine-deficient area. Eur J Endocrinol, 161:599-605.
    1. Michalaki MA, Vagenakis AG, Leonardou AS, Argentou MN, Habeos IG, Makri MG, et al.. (2006). Thyroid function in humans with morbid obesity. Thyroid, 16:73-8.
    1. Haymart MR, Repplinger DJ, Leverson GE, Elson DF, Sippel RS, Jaume JC, et al.. (2008). Higher serum thyroid stimulating hormone level in thyroid nodule patients is associated with greater risks of differentiated thyroid cancer and advanced tumor stage. J Clin Endocrinol Metab, 93:809-14.
    1. Gul K, Ozdemir D, Dirikoc A, Oguz A, Tuzun D, Baser H, et al.. (2010). Are endogenously lower serum thyroid hormones new predictors for thyroid malignancy in addition to higher serum thyrotropin? Endocrine, 37:253-60.
    1. Fussey JM, Beaumont RN, Wood AR, Vaidya B, Smith J, Tyrrell J (2020). Mendelian randomization supports a causative effect of TSH on thyroid carcinoma. Endocr Relat Cancer, 27:551-9.
    1. Haugen BR, Alexander EK, Bible KC, Doherty GM, Mandel SJ, Nikiforov YE, et al.. (2016). 2015 American Thyroid Association Management Guidelines for Adult Patients with Thyroid Nodules and Differentiated Thyroid Cancer: The American Thyroid Association Guidelines Task Force on Thyroid Nodules and Differentiated Thyroid Cancer. Thyroid, 26:1-133.
    1. Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M (2019). Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism, 92:121-35.
    1. Iyengar NM, Gucalp A, Dannenberg AJ, Hudis CA (2016). Obesity and Cancer Mechanisms: Tumor Microenvironment and Inflammation. J Clin Oncol, 34:4270-6.
    1. Stone TW, McPherson M, Darlington LG (2018). Obesity and cancer: existing and new hypotheses for a causal connection. EBioMedicine, 30:14-28.
    1. Mitsiades N, Pazaitou-Panayiotou K, Aronis KN, Moon HS, Chamberland JP, Liu X, et al.. (2011). Circulating adiponectin is inversely associated with risk of thyroid cancer: in vivo and in vitro studies. J Clin Endocrinol Metab, 96:E2023-8.
    1. Mele C, Sama MT, Bisoffi AA, Caputo M, Bullara V, Mai S, et al.. (2019). Circulating adipokines and metabolic setting in differentiated thyroid cancer. Endocr Connect, 8:997-1006.
    1. Warakomski J, Romuk E, Jarzab B, Krajewska J, Sieminska L (2018). Concentrations of Selected Adipokines, Interleukin-6, and Vitamin D in Patients with Papillary Thyroid Carcinoma in Respect to Thyroid Cancer Stages. Int J Endocrinol, 2018:4921803.
    1. Dossus L, Franceschi S, Biessy C, Navionis AS, Travis RC, Weiderpass E, et al.. (2018). Adipokines and inflammation markers and risk of differentiated thyroid carcinoma: the EPIC study. Int J Cancer, 142:1332-42.
    1. Kwon H, Park SE, Yun JS, Park CY (2020). Serum Adiponectin and Progranulin Level in Patients with Benign Thyroid Nodule or Papillary Thyroid Cancer. Endocrinol Metab (Seoul), 35:396-406.
    1. Abooshahab R, Yaghmaei P, Ghadaksaz HG, Hedayati M (2016). Lack of Association between Serum Adiponectin/Leptin Levels and Medullary Thyroid Cancer. Asian Pac J Cancer Prev, 17:3861-4.
    1. Pazaitou-Panayiotou K, Panagiotou G, Polyzos SA, Mantzoros CS (2016). SERUM ADIPONECTIN AND INSULIN-LIKE GROWTH FACTOR 1 IN PREDOMINANTLY FEMALE PATIENTS WITH THYROID CANCER: ASSOCIATION WITH THE HISTOLOGIC CHARACTERISTICS OF THE TUMOR. Endocr Pract, 22:68-75.
    1. Tumminia A, Vinciguerra F, Parisi M, Graziano M, Sciacca L, Baratta R, et al.. (2019). Adipose tissue, obesity and adiponectin: role in endocrine cancer risk. Int J Mol Sci, 20:2863.
    1. Baillargeon J, Platz EA, Rose DP, Pollock BH, Ankerst DP, Haffner S, et al.. (2006). Obesity, adipokines, and prostate cancer in a prospective population-based study. Cancer Epidemiol Biomarkers Prev, 15:1331-5.
    1. Ikeda A, Nakagawa T, Kawai K, Onozawa M, Hayashi T, Matsushita Y, et al.. (2015). Serum adiponectin concentration in 2,939 Japanese men undergoing screening for prostate cancer. Prostate Int, 3:87-92.
    1. Housa D, Vernerová Z, Herácek J, Procházka B, Cechák P, Kuncová J, et al.. (2008). Adiponectin as a potential marker of prostate cancer progression: studies in organ-confined and locally advanced prostate cancer. Physiol Res, 57:451-8.
    1. Menzaghi C, Trischitta V (2018). The Adiponectin Paradox for All-Cause and Cardiovascular Mortality. Diabetes, 67:12-22.
    1. Duggan C, Irwin ML, Xiao L, Henderson KD, Smith AW, Baumgartner RN, et al.. (2011). Associations of insulin resistance and adiponectin with mortality in women with breast cancer. J Clin Oncol, 29:32-9.
    1. Chong DQ, Mehta RS, Song M, Kedrin D, Meyerhardt JA, Ng K, et al.. (2015). Prediagnostic Plasma Adiponectin and Survival among Patients with Colorectal Cancer. Cancer Prev Res (Phila), 8:1138-45.
    1. Cheng SP, Liu CL, Hsu YC, Chang YC, Huang SY, Lee JJ (2013). Expression and biologic significance of adiponectin receptors in papillary thyroid carcinoma. Cell Biochem Biophys, 65:203-10.
    1. Herzig S, Shaw RJ (2018). AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol, 19:121-35.
    1. Kahn BB, Alquier T, Carling D, Hardie DG (2005). AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab, 1:15-25.
    1. Long YC, Zierath JR (2006). AMP-activated protein kinase signaling in metabolic regulation. J Clin Invest, 116:1776-83.
    1. Taliaferro-Smith L, Nagalingam A, Zhong D, Zhou W, Saxena NK, Sharma D (2009). LKB1 is required for adiponectin-mediated modulation of AMPK-S6K axis and inhibition of migration and invasion of breast cancer cells. Oncogene, 28:2621-33.
    1. Shaw RJ (2009). LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol, 196:65-80.
    1. Saxena NK, Sharma D (2010). Metastasis suppression by adiponectin: LKB1 rises up to the challenge. Cell Adh Migr, 4:358-62.
    1. Shackelford DB, Shaw RJ (2009). The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer, 9:563-75.
    1. Sugiyama M, Takahashi H, Hosono K, Endo H, Kato S, Yoneda K, et al.. (2009). Adiponectin inhibits colorectal cancer cell growth through the AMPK/mTOR pathway. Int J Oncol, 34:339-44.
    1. Motoshima H, Goldstein BJ, Igata M, Araki E (2006). AMPK and cell proliferation--AMPK as a therapeutic target for atherosclerosis and cancer. J Physiol, 574:63-71.
    1. Luo Z, Saha AK, Xiang X, Ruderman NB (2005). AMPK, the metabolic syndrome and cancer. Trends Pharmacol Sci, 26:69-76.
    1. Kim AY, Lee YS, Kim KH, Lee JH, Lee HK, Jang SH, et al.. (2010). Adiponectin represses colon cancer cell proliferation via AdipoR1- and -R2-mediated AMPK activation. Mol Endocrinol, 24:1441-52.
    1. Chung SJ, Nagaraju GP, Nagalingam A, Muniraj N, Kuppusamy P, Walker A, et al.. (2017). ADIPOQ/adiponectin induces cytotoxic autophagy in breast cancer cells through STK11/LKB1-mediated activation of the AMPK-ULK1 axis. Autophagy, 13:1386-403.
    1. Pinal N, Calleja M, Morata G (2019). Pro-apoptotic and pro-proliferation functions of the JNK pathway of Drosophila: roles in cell competition, tumorigenesis and regeneration. Open Biol, 9:180256.
    1. Papa S, Choy PM, Bubici C (2019). The ERK and JNK pathways in the regulation of metabolic reprogramming. Oncogene, 38:2223-40.
    1. Fathi N, Rashidi G, Khodadadi A, Shahi S, Sharifi S (2018). STAT3 and apoptosis challenges in cancer. Int J Biol Macromol, 117:993-1001.
    1. Laudisi F, Cherubini F, Monteleone G, Stolfi C (2018). STAT3 interactors as potential therapeutic targets for cancer treatment. Int J Mol Sci, 19:1787.
    1. Bowman T, Garcia R, Turkson J, Jove R (2000). STATs in oncogenesis. Oncogene, 19:2474-88.
    1. Beales ILP, Garcia-Morales C, Ogunwobi OO, Mutungi G (2014). Adiponectin inhibits leptin-induced oncogenic signalling in oesophageal cancer cells by activation of PTP1B. Mol Cell Endocrinol, 382:150-8.
    1. Messaggio F, Mendonsa AM, Castellanos J, Nagathihalli NS, Gorden L, Merchant NB, et al.. (2017). Adiponectin receptor agonists inhibit leptin induced pSTAT3 and in vivo pancreatic tumor growth. Oncotarget, 8:85378-91.
    1. Karim R, Tse G, Putti T, Scolyer R, Lee S (2004). The significance of the Wnt pathway in the pathology of human cancers. Pathology, 36:120-8.
    1. Wang Y, Lam JB, Lam KS, Liu J, Lam MC, Hoo RL, et al.. (2006). Adiponectin modulates the glycogen synthase kinase-3beta/beta-catenin signaling pathway and attenuates mammary tumorigenesis of MDA-MB-231 cells in nude mice. Cancer Res, 66:11462-70.
    1. Ding F, Xiao H, Wang M, Xie X, Hu F (2014). The role of the ubiquitin-proteasome pathway in cancer development and treatment. Front Biosci, 19:886-95.
    1. Qu Q, Mao Y, Xiao G, Fei X, Wang J, Zhang Y, et al.. (2015). USP2 promotes cell migration and invasion in triple negative breast cancer cell lines. Tumour Biol, 36:5415-23.
    1. Young MJ, Hsu KC, Lin TE, Chang WC, Hung JJ (2019). The role of ubiquitin-specific peptidases in cancer progression. J Biomed Sci, 26:42.
    1. Shan J, Zhao W, Gu W (2009). Suppression of cancer cell growth by promoting cyclin D1 degradation. Mol Cell, 36:469-76.
    1. Lee JT, Shan J, Gu W (2010). Targeting the degradation of cyclin D1 will help to eliminate oncogene addiction. Cell Cycle, 9:857-8.
    1. Nepal S, Shrestha A, Park PH (2015). Ubiquitin specific protease 2 acts as a key modulator for the regulation of cell cycle by adiponectin and leptin in cancer cells. Mol Cell Endocrinol, 412:44-55.
    1. Dieudonne MN, Bussiere M, Dos Santos E, Leneveu MC, Giudicelli Y, Pecquery R (2006). Adiponectin mediates antiproliferative and apoptotic responses in human MCF7 breast cancer cells. Biochem Biophys Res Commun, 345:271-9.
    1. Gallagher EJ, LeRoith D (2015). Obesity and Diabetes: The Increased Risk of Cancer and Cancer-Related Mortality. Physiol Rev, 95:727-48.
    1. Arcidiacono B, Iiritano S, Nocera A, Possidente K, Nevolo MT, Ventura V, et al.. (2012). Insulin resistance and cancer risk: an overview of the pathogenetic mechanisms. Exp Diabetes Res, 2012:789174.
    1. Le Roith D (2003). The insulin-like growth factor system. Exp Diabesity Res, 4:205-12.
    1. Elbaz M, Ahirwar D, Ravi J, Nasser MW, Ganju RK (2017). Novel role of cannabinoid receptor 2 in inhibiting EGF/EGFR and IGF-I/IGF-IR pathways in breast cancer. Oncotarget, 8:29668-78.
    1. Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO, Demir IE (2018). Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Mol Cancer, 17:66.
    1. Mourmouras N, Philippou A, Christopoulos P, Kostoglou K, Grivaki C, Konstantinidis C, et al.. (2018). Differential Expression of IGF-I Transcripts in Bladder Cancer. Anticancer Res, 38:3453-9.
    1. Karagiannis AK, Philippou A, Tseleni-Balafouta S, Zevolis E, Nakouti T, Tsopanomichalou-Gklotsou M, et al.. (2019). IGF-IEc Expression Is Associated With Advanced Differentiated Thyroid Cancer. Anticancer Res, 39:2811-9.
    1. Manzella L, Massimino M, Stella S, Tirrò E, Pennisi MS, Martorana F, et al.. (2019). Activation of the IGF axis in thyroid cancer: implications for tumorigenesis and treatment. Int J Mol Sci, 20:3258.
    1. Belfiore A, Pandini G, Vella V, Squatrito S, Vigneri R (1999). Insulin/IGF-I hybrid receptors play a major role in IGF-I signaling in thyroid cancer. Biochimie, 81:403-7.
    1. Schmidt JA, Allen NE, Almquist M, Franceschi S, Rinaldi S, Tipper SJ, et al.. (2014). Insulin-like growth factor-i and risk of differentiated thyroid carcinoma in the European prospective investigation into cancer and nutrition. Cancer Epidemiol Biomarkers Prev, 23:976-85.
    1. Vigneri R, Goldfine ID, Frittitta L (2016). Insulin, insulin receptors, and cancer. J Endocrinol Invest, 39:1365-76.
    1. Pandini G, Frasca F, Mineo R, Sciacca L, Vigneri R, Belfiore A (2002). Insulin/insulin-like growth factor I hybrid receptors have different biological characteristics depending on the insulin receptor isoform involved. J Biol Chem, 277:39684-95.
    1. Vella V, Pandini G, Sciacca L, Mineo R, Vigneri R, Pezzino V, et al.. (2002). A novel autocrine loop involving IGF-II and the insulin receptor isoform-A stimulates growth of thyroid cancer. J Clin Endocrinol Metab, 87:245-54.
    1. Morgan SJ, Neumann S, Marcus-Samuels B, Gershengorn MC (2016). Thyrotropin and Insulin-Like Growth Factor 1 Receptor Crosstalk Upregulates Sodium-Iodide Symporter Expression in Primary Cultures of Human Thyrocytes. Thyroid, 26:1794-803.
    1. Tas F, Bilgin E, Tastekin D, Erturk K, Duranyildiz D (2016). Serum IGF-1 and IGFBP-3 levels as clinical markers for patients with lung cancer. Biomed Rep, 4:609-14.

Source: PubMed

3
Prenumerera