open access

Vol 74, No 5 (2023)
Review paper
Submitted: 2023-04-30
Accepted: 2023-07-14
Published online: 2023-10-09
Get Citation

Adipose tissue as a cause of endocrine dysfunction

Alina Ewa Kuryłowicz12
·
Pubmed: 37902012
·
Endokrynol Pol 2023;74(5):468-479.
Affiliations
  1. Department of Human Epigenetics, Mossakowski Medical Research Centre PAS, Warsaw, Poland
  2. Department of General Medicine and Geriatric Cardiology, Medical Centre of Postgraduate Education Warsaw, Poland

open access

Vol 74, No 5 (2023)
Review Article
Submitted: 2023-04-30
Accepted: 2023-07-14
Published online: 2023-10-09

Abstract

Adipose tissue is a large hormonally active organ that secretes several substances (adipokines), and an important site for the synthesis and metabolism of steroid hormones. With energy balance, the secretory and metabolic activity of adipose tissue determines the normal function of many organs, including the endocrine glands. However, in the course of overweight and obesity, adverse changes occur in the structure and function of adipocytes. Obesity-related adipose tissue dysfunction translates into a change in the profile of secreted adipokines, and it impairs steroidogenesis. These phenomena contribute to the development of obesity-related complications, which also affect the major tropic axes regulating the endocrine glands. However, there is increasing evidence that weight reduction is an effective treatment for obesity-related adipose tissue dysfunction, thereby restoring endocrine function.

This narrative review presents the impact of adipose tissue on endocrine gland activity both in the physiological state and in obesity-related dysfunction. It also discusses how functional (related to excess adiposity) changes in the endocrine system can be restored with effective treatment of obesity.

Abstract

Adipose tissue is a large hormonally active organ that secretes several substances (adipokines), and an important site for the synthesis and metabolism of steroid hormones. With energy balance, the secretory and metabolic activity of adipose tissue determines the normal function of many organs, including the endocrine glands. However, in the course of overweight and obesity, adverse changes occur in the structure and function of adipocytes. Obesity-related adipose tissue dysfunction translates into a change in the profile of secreted adipokines, and it impairs steroidogenesis. These phenomena contribute to the development of obesity-related complications, which also affect the major tropic axes regulating the endocrine glands. However, there is increasing evidence that weight reduction is an effective treatment for obesity-related adipose tissue dysfunction, thereby restoring endocrine function.

This narrative review presents the impact of adipose tissue on endocrine gland activity both in the physiological state and in obesity-related dysfunction. It also discusses how functional (related to excess adiposity) changes in the endocrine system can be restored with effective treatment of obesity.

Get Citation

Keywords

adipose tissue; adipokines; hormonal homeostasis; obesity; adipose tissue dysfunction; lifestyle interventions; glucagon-like peptide 1 receptor agonists (GLP-1RAs); bariatric surgery

About this article
Title

Adipose tissue as a cause of endocrine dysfunction

Journal

Endokrynologia Polska

Issue

Vol 74, No 5 (2023)

Article type

Review paper

Pages

468-479

Published online

2023-10-09

Page views

981

Article views/downloads

683

DOI

10.5603/ep.95378

Pubmed

37902012

Bibliographic record

Endokrynol Pol 2023;74(5):468-479.

Keywords

adipose tissue
adipokines
hormonal homeostasis
obesity
adipose tissue dysfunction
lifestyle interventions
glucagon-like peptide 1 receptor agonists (GLP-1RAs)
bariatric surgery

Authors

Alina Ewa Kuryłowicz

References (114)
  1. Cypess AM, Cypess AM. Reassessing Human Adipose Tissue. N Engl J Med. 2022; 386(8): 768–779.
  2. Li J, Papadopoulos V, Vihma V. Steroid biosynthesis in adipose tissue. Steroids. 2015; 103: 89–104.
  3. Morrison CD. Leptin signaling in brain: A link between nutrition and cognition? Biochim Biophys Acta. 2009; 1792(5): 401–408.
  4. Watanobe H, Habu S. Leptin regulates growth hormone-releasing factor, somatostatin, and alpha-melanocyte-stimulating hormone but not neuropeptide Y release in rat hypothalamus in vivo: relation with growth hormone secretion. J Neurosci. 2002; 22(14): 6265–6271.
  5. Saleri R, Giustina A, Tamanini C, et al. Leptin stimulates growth hormone secretion via a direct pituitary effect combined with a decreased somatostatin tone in a median eminence-pituitary perifusion study. Neuroendocrinology. 2004; 79(4): 221–228.
  6. Ozata M, Ozdemir IC, Licinio J. Human leptin deficiency caused by a missense mutation: multiple endocrine defects, decreased sympathetic tone, and immune system dysfunction indicate new targets for leptin action, greater central than peripheral resistance to the effects of leptin, and spontaneous correction of leptin-mediated defects. J Clin Endocrinol Metab. 1999; 84(10): 3686–3695.
  7. Witkowska-Sędek E, Rumińska M, Stelmaszczyk-Emmel A, et al. The associations between the growth hormone/insulin-like growth factor-1 axis, adiponectin, resistin and metabolic profile in children with growth hormone deficiency before and during growth hormone treatment. Acta Biochim Pol. 2018; 65(2): 333–340.
  8. Childs GV, Odle AK, MacNicol MC, et al. The Importance of Leptin to Reproduction. Endocrinology. 2021; 162(2).
  9. Jin L, Burguera BG, Couce ME, et al. Leptin and leptin receptor expression in normal and neoplastic human pituitary: evidence of a regulatory role for leptin on pituitary cell proliferation. J Clin Endocrinol Metab. 1999; 84(8): 2903–2911.
  10. Cioffi JA, Shafer AW, Zupancic TJ, et al. Novel B219/OB receptor isoforms: possible role of leptin in hematopoiesis and reproduction. Nat Med. 1996; 2(5): 585–589.
  11. Tena-Sempere M, Manna PR, Zhang FP, et al. Developmental and hormonal regulation of leptin receptor (Ob-R) messenger ribonucleic acid expression in rat testis. Biol Reprod. 2001; 64(2): 634–643.
  12. Boutari C, Pappas PD, Mintziori G, et al. The effect of underweight on female and male reproduction. Metabolism. 2020; 107: 154229.
  13. Reverchon M, Cornuau M, Ramé C, et al. Resistin decreases insulin-like growth factor I-induced steroid production and insulin-like growth factor I receptor signaling in human granulosa cells. Fertil Steril. 2013; 100(1): 247–55.e1.
  14. Munir I, Yen HW, Baruth T, et al. Resistin stimulation of 17alpha-hydroxylase activity in ovarian theca cells in vitro: relevance to polycystic ovary syndrome. J Clin Endocrinol Metab. 2005; 90(8): 4852–4857.
  15. Maillard V, Elis S, Desmarchais A, et al. Visfatin and resistin in gonadotroph cells: expression, regulation of LH secretion and signalling pathways. Reprod Fertil Dev. 2017; 29(12): 2479–2495.
  16. Ziora KT, Oswiecimska JM, Swietochowska E, et al. Assessment of serum levels resistin in girls with anorexia nervosa. Part II. Relationships between serum levels of resistin and thyroid, adrenal and gonadal hormones. Neuro Endocrinol Lett. 2011; 32(5): 697–703.
  17. Raeisi T, Rezaie H, Darand M, et al. Circulating resistin and follistatin levels in obese and non-obese women with polycystic ovary syndrome: A systematic review and meta-analysis. PLoS One. 2021; 16(3): e0246200.
  18. Lagaly DV, Aad PY, Grado-Ahuir JA, et al. Role of adiponectin in regulating ovarian theca and granulosa cell function. Mol Cell Endocrinol. 2008; 284(1-2): 38–45.
  19. Rodriguez-Pacheco F, Martinez-Fuentes AJ, Tovar S, et al. Regulation of pituitary cell function by adiponectin. Endocrinology. 2007; 148(1): 401–410.
  20. Choubey M, Ranjan A, Bora PS, et al. Role of adiponectin as a modulator of testicular function during aging in mice. Biochim Biophys Acta Mol Basis Dis. 2019; 1865(2): 413–427.
  21. Reverchon M, Rame C, Bunel A, et al. VISFATIN (NAMPT) Improves In Vitro IGF1-Induced Steroidogenesis and IGF1 Receptor Signaling Through SIRT1 in Bovine Granulosa Cells. Biol Reprod. 2016; 94(3): 54.
  22. Jeremy M, Gurusubramanian G, Roy VK. Localization pattern of visfatin (NAMPT) in d-galactose induced aged rat testis. Ann Anat. 2017; 211: 46–54.
  23. Ortiga-Carvalho TM, Oliveira KJ, Soares BA, et al. The role of leptin in the regulation of TSH secretion in the fed state: in vivo and in vitro studies. J Endocrinol. 2002; 174(1): 121–125.
  24. Kim MS, Small CJ, Stanley SA, et al. The central melanocortin system affects the hypothalamo-pituitary thyroid axis and may mediate the effect of leptin. J Clin Invest. 2000; 105(7): 1005–1011.
  25. Nillni EA. Regulation of prohormone convertases in hypothalamic neurons: implications for prothyrotropin-releasing hormone and proopiomelanocortin. Endocrinology. 2007; 148(9): 4191–4200.
  26. Radwanska P, Kosior-Korzecka U. Effect of leptin on thyroid-stimulating hormone secretion and nitric oxide release from pituitary cells of ewe lambs in vitro. J Physiol Pharmacol. 2014; 65(1): 145–151.
  27. Mantzoros CS, Ozata M, Negrao AB, et al. Synchronicity of frequently sampled thyrotropin (TSH) and leptin concentrations in healthy adults and leptin-deficient subjects: evidence for possible partial TSH regulation by leptin in humans. J Clin Endocrinol Metab. 2001; 86(7): 3284–3291.
  28. Fernández-Real JM, López-Bermejo A, Casamitjana R, et al. Novel interactions of adiponectin with the endocrine system and inflammatory parameters. J Clin Endocrinol Metab. 2003; 88(6): 2714–2718.
  29. Soltys BJ, Kang D, Gupta RS. Localization of P32 protein (gC1q-R) in mitochondria and at specific extramitochondrial locations in normal tissues. Histochem Cell Biol. 2000; 114(3): 245–255.
  30. Zhou Y, Yang Y, Zhou T, et al. Adiponectin and Thyroid Cancer: Insight into the Association between Adiponectin and Obesity. Aging Dis. 2021; 12(2): 597–613.
  31. Walczak K, Siemińska L. The relationships between selected serum adipokines and thyroid function in patients with obesity. Endokrynol Pol. 2022; 73(2): 353–360.
  32. Sotak Š, Schroner Z, Lazúrová I, et al. The Association Between Three Adipocytokines (Adiponectin, Resistin and Visfatin) And Thyroid Status in Patients With Type 2 Diabetes Mellitus and Autoimmune Thyroiditis. Physiol Res. 2021; 70(6): 865–874.
  33. García-Solís P, García OP, Hernández-Puga G, et al. Thyroid hormones and obesity: a known but poorly understood relationship. Endokrynol Pol. 2018; 69(3): 292–303.
  34. Lu S, Guan Q, Liu Y, et al. Role of extrathyroidal TSHR expression in adipocyte differentiation and its association with obesity. Lipids Health Dis. 2012; 11: 17.
  35. Malcher-Lopes R, Di S, Marcheselli VS, et al. Opposing crosstalk between leptin and glucocorticoids rapidly modulates synaptic excitation via endocannabinoid release. J Neurosci. 2006; 26(24): 6643–6650.
  36. Pralong FP, Roduit R, Waeber G, et al. Leptin inhibits directly glucocorticoid secretion by normal human and rat adrenal gland. Endocrinology. 1998; 139(10): 4264–4268.
  37. Ahima RS, Prabakaran D, Mantzoros C, et al. Role of leptin in the neuroendocrine response to fasting. Nature. 1996; 382(6588): 250–252.
  38. Faulkner JL, Belin de Chantemèle EJ. Leptin and Aldosterone. Vitam Horm. 2019; 109: 265–284.
  39. Li P, Sun F, Cao HM, et al. Expression of adiponectin receptors in mouse adrenal glands and the adrenocortical Y-1 cell line: adiponectin regulates steroidogenesis. Biochem Biophys Res Commun. 2009; 390(4): 1208–1213.
  40. John K, Marino JS, Sanchez ER, et al. The glucocorticoid receptor: cause of or cure for obesity? Am J Physiol Endocrinol Metab. 2016; 310(4): E249–E257.
  41. Wang L, Li S, Zhao A, et al. The expression of sex steroid synthesis and inactivation enzymes in subcutaneous adipose tissue of PCOS patients. J Steroid Biochem Mol Biol. 2012; 132(1-2): 120–126.
  42. Wang F, Vihma V, Soronen J, et al. 17β-Estradiol and estradiol fatty acyl esters and estrogen-converting enzyme expression in adipose tissue in obese men and women. J Clin Endocrinol Metab. 2013; 98(12): 4923–4931.
  43. Kinoshita T, Honma S, Shibata Y, et al. An innovative LC-MS/MS-based method for determining CYP 17 and CYP 19 activity in the adipose tissue of pre- and postmenopausal and ovariectomized women using 13C-labeled steroid substrates. J Clin Endocrinol Metab. 2014; 99(4): 1339–1347.
  44. Stulnig TM, Waldhäusl W. 11beta-Hydroxysteroid dehydrogenase Type 1 in obesity and Type 2 diabetes. Diabetologia. 2004; 47(1): 1–11.
  45. Sarma S, Sockalingam S, Dash S. Obesity as a multisystem disease: Trends in obesity rates and obesity-related complications. Diabetes Obes Metab. 2021; 23 Suppl 1: 3–16.
  46. Woo CY, Jang JE, Lee SE, et al. Mitochondrial Dysfunction in Adipocytes as a Primary Cause of Adipose Tissue Inflammation. Diabetes Metab J. 2019; 43(3): 247–256.
  47. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017; 542(7640): 177–185.
  48. Yanai H, Yoshida H. Beneficial Effects of Adiponectin on Glucose and Lipid Metabolism and Atherosclerotic Progression: Mechanisms and Perspectives. Int J Mol Sci. 2019; 20(5).
  49. Abella V, Scotece M, Conde J, et al. Leptin in the interplay of inflammation, metabolism and immune system disorders. Nat Rev Rheumatol. 2017; 13(2): 100–109.
  50. Kreitschmann-Andermahr I, Suarez P, Jennings R, et al. GH/IGF-I regulation in obesity--mechanisms and practical consequences in children and adults. Horm Res Paediatr. 2010; 73(3): 153–160.
  51. Gleeson HK, Lissett CA, Shalet SM. Insulin-like growth factor-I response to a single bolus of growth hormone is increased in obesity. J Clin Endocrinol Metab. 2005; 90(2): 1061–1067.
  52. van Hulsteijn LT, Pasquali R, Casanueva F, et al. Prevalence of endocrine disorders in obese patients: systematic review and meta-analysis. Eur J Endocrinol. 2020; 182(1): 11–21.
  53. Cohen PG. Obesity in men: the hypogonadal-estrogen receptor relationship and its effect on glucose homeostasis. Med Hypotheses. 2008; 70(2): 358–360.
  54. Liu Y, Ding Z. Obesity, a serious etiologic factor for male subfertility in modern society. Reproduction. 2017; 154(4): R123–R131.
  55. Baldelli R, Dieguez C, Casanueva FF. The role of leptin in reproduction: experimental and clinical aspects. Ann Med. 2002; 34(1): 5–18.
  56. Unuane D, Tournaye H, Velkeniers B, et al. Endocrine disorders & female infertility. Best Pract Res Clin Endocrinol Metab. 2011; 25(6): 861–873.
  57. Sanchez-Garrido MA, Tena-Sempere M. Metabolic dysfunction in polycystic ovary syndrome: Pathogenic role of androgen excess and potential therapeutic strategies. Mol Metab. 2020; 35: 100937.
  58. Pasquali R, Gambineri A, Pagotto U. The impact of obesity on reproduction in women with polycystic ovary syndrome. BJOG. 2006; 113(10): 1148–1159.
  59. Fassnacht M, Schlenz N, Schneider SB, et al. Beyond adrenal and ovarian androgen generation: Increased peripheral 5 alpha-reductase activity in women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2003; 88(6): 2760–2766.
  60. Baranowska-Bik A. Therapy of obesity in women with PCOS using GLP-1 analogues - benefits and limitations [Terapia otyłości u kobiet z PCOS przy zastosowaniu analogów GLP-1 - korzyści i ograniczenia]. Endokrynol Pol. 2022; 73(3): 627–643.
  61. Carmina E, Bucchieri S, Mansueto P, et al. Circulating levels of adipose products and differences in fat distribution in the ovulatory and anovulatory phenotypes of polycystic ovary syndrome. Fertil Steril. 2009; 91(4 Suppl): 1332–1335.
  62. González F, González F. Inflammation in Polycystic Ovary Syndrome: underpinning of insulin resistance and ovarian dysfunction. Steroids. 2012; 77(4): 300–305.
  63. Best D, Avenell A, Bhattacharya S. How effective are weight-loss interventions for improving fertility in women and men who are overweight or obese? A systematic review and meta-analysis of the evidence. Hum Reprod Update. 2017; 23(6): 681–705.
  64. Broughton DE, Moley KH. Obesity and female infertility: potential mediators of obesity's impact. Fertil Steril. 2017; 107(4): 840–847.
  65. Pasquali R, Casanueva F, Haluzik M, et al. European Society of Endocrinology Clinical Practice Guideline: Endocrine work-up in obesity. Eur J Endocrinol. 2020; 182(1): G1–G32.
  66. Zimmermann-Belsing T, Brabant G, Holst JJ, et al. Circulating leptin and thyroid dysfunction. Eur J Endocrinol. 2003; 149(4): 257–271.
  67. Janssen IMC, Homan J, Schijns W, et al. Subclinical hypothyroidism and its relation to obesity in patients before and after Roux-en-Y gastric bypass. Surg Obes Relat Dis. 2015; 11(6): 1257–1263.
  68. Marzullo P, Minocci A, Tagliaferri MA, et al. Investigations of thyroid hormones and antibodies in obesity: leptin levels are associated with thyroid autoimmunity independent of bioanthropometric, hormonal, and weight-related determinants. J Clin Endocrinol Metab. 2010; 95(8): 3965–3972.
  69. Štěpánek L, Horáková D, Štěpánek L, et al. Free triiodothyronine/free thyroxine (FT3/FT4) ratio is strongly associated with insulin resistance in euthyroid and hypothyroid adults: a cross-sectional study. Endokrynol Pol. 2021; 72(1): 8–13.
  70. Rotondi M, Cappelli C, Leporati P, et al. A hypoechoic pattern of the thyroid at ultrasound does not indicate autoimmune thyroid diseases in patients with morbid obesity. Eur J Endocrinol. 2010; 163(1): 105–109.
  71. Björntorp P, Rosmond R. Obesity and cortisol. Nutrition. 2000; 16(10): 924–936.
  72. Rask E, Walker BR, Söderberg S, et al. Tissue-specific changes in peripheral cortisol metabolism in obese women: increased adipose 11beta-hydroxysteroid dehydrogenase type 1 activity. J Clin Endocrinol Metab. 2002; 87(7): 3330–3336.
  73. Masuzaki H, Paterson J, Shinyama H, et al. A transgenic model of visceral obesity and the metabolic syndrome. Science. 2001; 294(5549): 2166–2170.
  74. Kershaw EE, Morton NM, Dhillon H, et al. Adipocyte-specific glucocorticoid inactivation protects against diet-induced obesity. Diabetes. 2005; 54(4): 1023–1031.
  75. Wamil M, Battle JH, Turban S, et al. Novel fat depot-specific mechanisms underlie resistance to visceral obesity and inflammation in 11 β-hydroxysteroid dehydrogenase type 1-deficient mice. Diabetes. 2011; 60(4): 1158–1167.
  76. Feig PU, Shah S, Hermanowski-Vosatka A, et al. Effects of an 11β-hydroxysteroid dehydrogenase type 1 inhibitor, MK-0916, in patients with type 2 diabetes mellitus and metabolic syndrome. Diabetes Obes Metab. 2011; 13(6): 498–504.
  77. Pasquali R, Vicennati V, Gambineri A. Adrenal and gonadal function in obesity. J Endocrinol Invest. 2002; 25(10): 893–898.
  78. Monzillo LU, Hamdy O, Horton ES, et al. Effect of lifestyle modification on adipokine levels in obese subjects with insulin resistance. Obes Res. 2003; 11(9): 1048–1054.
  79. Savchenko LG, Digtiar NI, Selikhova LG, et al. Liraglutide exerts an anti-inflammatory action in obese patients with type 2 diabetes. Rom J Intern Med. 2019; 57(3): 233–240.
  80. Kopp HP, Krzyzanowska K, Möhlig M, et al. Effects of marked weight loss on plasma levels of adiponectin, markers of chronic subclinical inflammation and insulin resistance in morbidly obese women. Int J Obes (Lond). 2005; 29(7): 766–771.
  81. Gajda SN, Kuryłowicz A, Żach M, et al. Diagnosis and treatment of thyroid disorders in obese patients - what do we know? Endokrynol Pol. 2019; 70(3): 271–276.
  82. Kozłowska L, Rosołowska-Huszcz D. Leptin, thyrotropin, and thyroid hormones in obese/overweight women before and after two levels of energy deficit. Endocrine. 2004; 24(2): 147–153.
  83. Radetti G, Longhi S, Baiocchi M, et al. Changes in lifestyle improve body composition, thyroid function, and structure in obese children. J Endocrinol Invest. 2012; 35(3): 281–285.
  84. Agnihothri RV, Courville AB, Linderman JD, et al. Moderate weight loss is sufficient to affect thyroid hormone homeostasis and inhibit its peripheral conversion. Thyroid. 2014; 24(1): 19–26.
  85. Rask E, Simonyte K, Lönn L, et al. Cortisol metabolism after weight loss: associations with 11 β-HSD type 1 and markers of obesity in women. Clin Endocrinol (Oxf). 2013; 78(5): 700–705.
  86. London E, Castonguay TW. Diet and the role of 11beta-hydroxysteroid dehydrogenase-1 on obesity. J Nutr Biochem. 2009; 20(7): 485–493.
  87. Gu Y, Zhou G, Zhou F, et al. Life Modifications and PCOS: Old Story But New Tales. Front Endocrinol (Lausanne). 2022; 13: 808898.
  88. Pasquali R. Contemporary approaches to the management of polycystic ovary syndrome. Ther Adv Endocrinol Metab. 2018; 9(4): 123–134.
  89. Crosignani PG, Colombo M, Vegetti W, et al. Overweight and obese anovulatory patients with polycystic ovaries: parallel improvements in anthropometric indices, ovarian physiology and fertility rate induced by diet. Hum Reprod. 2003; 18(9): 1928–1932.
  90. Pasquali R, Gambineri A, Cavazza C, et al. Heterogeneity in the responsiveness to long-term lifestyle intervention and predictability in obese women with polycystic ovary syndrome. Eur J Endocrinol. 2011; 164(1): 53–60.
  91. Lotti F, Marchiani S, Corona G, et al. Metabolic Syndrome and Reproduction. Int J Mol Sci. 2021; 22(4).
  92. Fildes A, Charlton J, Rudisill C, et al. Probability of an Obese Person Attaining Normal Body Weight: Cohort Study Using Electronic Health Records. Am J Public Health. 2015; 105(9): e54–e59.
  93. Canadian Adult Obesity Clinical Practice Guidelines. https://obesitycanada.ca/guidelines/.
  94. Bąk-Sosnowska M, Białkowska M, Bogdański P, et al. Zalecenia kliniczne dotyczące postępowania u chorych na otyłość 2022 — stanowisko Polskiego Towarzystwa Leczenia Otyłości. Med Prakt. 2022(Wyd Spec): 1–87.
  95. Cariou B. Harnessing the incretin system beyond glucose control: potential cardiovascular benefits of GLP-1 receptor agonists in type 2 diabetes. Diabetes Metab. 2012; 38(4): 298–308.
  96. Lyu X, Yan K, Wang X, et al. A novel anti-obesity mechanism for liraglutide by improving adipose tissue leptin resistance in high-fat diet-fed obese mice. Endocr J. 2022; 69(10): 1233–1244.
  97. Li D, Xu X, Zhang Y, et al. Liraglutide treatment causes upregulation of adiponectin and downregulation of resistin in Chinese type 2 diabetes. Diabetes Res Clin Pract. 2015; 110(2): 224–228.
  98. Jensterle M, Janez A, Fliers E, et al. The role of glucagon-like peptide-1 in reproduction: from physiology to therapeutic perspective. Hum Reprod Update. 2019; 25(4): 504–517.
  99. Jensterle M, Podbregar A, Goricar K, et al. Effects of liraglutide on obesity-associated functional hypogonadism in men. Endocr Connect. 2019; 8(3): 195–202.
  100. Giagulli VA, Carbone MD, Ramunni MI, et al. Adding liraglutide to lifestyle changes, metformin and testosterone therapy boosts erectile function in diabetic obese men with overt hypogonadism. Andrology. 2015; 3(6): 1094–1103.
  101. Niafar M, Pourafkari L, Porhomayon J, et al. A systematic review of GLP-1 agonists on the metabolic syndrome in women with polycystic ovaries. Arch Gynecol Obstet. 2016; 293(3): 509–515.
  102. Heppner KM, Baquero AF, Bennett CM, et al. GLP-1R Signaling Directly Activates Arcuate Nucleus Kisspeptin Action in Brain Slices but Does not Rescue Luteinizing Hormone Inhibition in Ovariectomized Mice During Negative Energy Balance. eNeuro. 2017; 4(1).
  103. Izzi-Engbeaya C, Jones S, Crustna Y, et al. Effects of Glucagon-like Peptide-1 on the Reproductive Axis in Healthy Men. J Clin Endocrinol Metab. 2020; 105(4): 1119–1125.
  104. Hu W, Song R, Cheng R, et al. Use of GLP-1 Receptor Agonists and Occurrence of Thyroid Disorders: a Meta-Analysis of Randomized Controlled Trials. Front Endocrinol (Lausanne). 2022; 13: 927859.
  105. Ye J, Xu J, Wen W, et al. Effect of Liraglutide on Serum TSH Levels in Patients with NAFLD and its Underlying Mechanisms. Int J Clin Pract. 2022; 2022: 1786559.
  106. Eisenberg D, Shikora SA, Kothari SN, et al. 2022 American Society for Metabolic and Bariatric Surgery (ASMBS) and International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO): Indications for Metabolic and Bariatric Surgery. Surg Obes Relat Dis. 2022; 18(12): 1345–1356.
  107. Hafida S, Mirshahi T, Nikolajczyk BS. The impact of bariatric surgery on inflammation: quenching the fire of obesity? Curr Opin Endocrinol Diabetes Obes. 2016; 23(5): 373–378.
  108. Camastra S, Vitali A, Anselmino M, et al. Muscle and adipose tissue morphology, insulin sensitivity and beta-cell function in diabetic and nondiabetic obese patients: effects of bariatric surgery. Sci Rep. 2017; 7(1): 9007.
  109. Guan B, Chen Y, Yang J, et al. Effect of Bariatric Surgery on Thyroid Function in Obese Patients: a Systematic Review and Meta-Analysis. Obes Surg. 2017; 27(12): 3292–3305.
  110. Handisurya A, Riedl M, Vila G, et al. Serum vaspin concentrations in relation to insulin sensitivity following RYGB-induced weight loss. Obes Surg. 2010; 20(2): 198–203.
  111. Lee Y, Dang JT, Switzer N, et al. Impact of Bariatric Surgery on Male Sex Hormones and Sperm Quality: a Systematic Review and Meta-Analysis. Obes Surg. 2019; 29(1): 334–346.
  112. Emami MR, Safabakhsh M, Khorshidi M, et al. Effect of bariatric surgery on endogenous sex hormones and sex hormone-binding globulin levels: a systematic review and meta-analysis. Surg Obes Relat Dis. 2021; 17(9): 1621–1636.
  113. Escobar-Morreale HF, Santacruz E, Luque-Ramírez M, et al. Prevalence of 'obesity-associated gonadal dysfunction' in severely obese men and women and its resolution after bariatric surgery: a systematic review and meta-analysis. Hum Reprod Update. 2017; 23(4): 390–408.
  114. Juiz-Valiña P, Pena-Bello L, Cordido M, et al. Altered GH-IGF-1 Axis in Severe Obese Subjects is Reversed after Bariatric Surgery-Induced Weight Loss and Related with Low-Grade Chronic Inflammation. J Clin Med. 2020; 9(8).

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl