The Role of Polyamines in Metabolic Syndrome

Jump To References Section

Authors

  • Department of Biological Sciences, BITS Pilani, K K Birla, Goa Campus, Zuarinagar, Goa 403726 ,IN
  • Department of Biological Sciences, BITS Pilani, K K Birla, Goa Campus, Zuarinagar, Goa 403726 ,IN
  • Department of Biological Sciences, BITS Pilani, K K Birla, Goa Campus, Zuarinagar, Goa 403726 ,IN
  • Department of Biological Sciences, BITS Pilani, K K Birla, Goa Campus, Zuarinagar, Goa 403726 ,IN

DOI:

https://doi.org/10.18311/jer/2022/28962

Keywords:

Metabolic Syndrome (MetS), Obesity, Polyamines, Type 2 Diabetes Mellitus (T2D)
Metabolic disorder

Abstract

Metabolic syndrome has become a major health hazard worldwide, due to an increased consumption of fast food along with other unhealthy eating habits, decrease in physical activities and major lifestyle changes as a result of modernization. Metabolic syndrome is a non-communicable disease resulting in complications like type 2 diabetes, coronary diseases, stroke etc. The polyamines putrescine, spermidine, and spermine are naturally occurring cationic molecules essential for various cellular functions. Dysregulated polyamine circulation and metabolism have been linked to several conditions including cancer, type 2 diabetes mellitus, adipogenesis, altered lipid or glucose metabolism, chronic inflammation, etc. Several studies regarding the role of polyamines in metabolic syndrome in various models have been conducted, and the outcomes suggest a positive correlation in the prognosis of metabolic syndrome. This review provides a comprehensive discussion of metabolic syndrome, its prevalence in India and worldwide, factors causing the syndrome, and the role of polyamines in the pathogenesis of various complications of metabolic syndrome.

Downloads

Download data is not yet available.

Metrics

Metrics Loading ...

Downloads

Published

2022-08-12

How to Cite

Ajith, A., Chandra, K., Mate, N., & Banerjee, A. (2022). The Role of Polyamines in Metabolic Syndrome. Journal of Endocrinology and Reproduction, 26(2), 67–74. https://doi.org/10.18311/jer/2022/28962

Issue

Section

Review Article

 

References

Saklayen MG. The global epidemic of the metabolic syndrome. Current Hypertension Reports. 2018; 20(2):12. https://doi.org/10.1007/s11906-018-0812-z. PMid:29480368. PMCid: PMC5866840

Gupta A, Gupta V. Metabolic syndrome: what are the risks for humans? BioScience Trends. 2010; 4(5):204–12.

Grundy SM. Metabolic syndrome pandemic. Arteriosclerosis, Thrombosis, and Vascular Biology. 2008; 28(4):629–36. https://doi.org/10.1161/ATVBAHA.107.151092. PMid:18174459

Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA. Diagnosis and management of the metabolic syndrome. Circulation. 2005; 112(17):2735–52. https://doi.org/10.1161/CIRCULATIONAHA.105.169405. PMid:16157765

Samson SL, Garber AJ. Metabolic syndrome. Endocrinology and Metabolism Clinics of North America. 2014; 43(1):1–23. https://doi.org/10.1016/j.ecl.2013.09.009. PMid:24582089

Kolovou GD, Anagnostopoulou KK, Salpea KD, Mikhailidis DP. The prevalence of metabolic syndrome in various populations. The American Journal of the Medical Sciences. 2007; 333(6):362–71. https://doi.org/10.1097/MAJ.0b013e318065c3a1. PMid:17570989

Health effects of overweight and obesity in 195 countries over 25 years. New England Journal of Medicine. 2017; 377(1):13–27. https://doi.org/10.1056/NEJMoa1614362. PMid:28604169. PMCid:PMC5477817

Centers for disease control and prevention. National Diabetes Statistics Report website.

Palaniappan LP, Wong EC, Shin JJ, Fortmann SP, Lauderdale DS. Asian americans have greater prevalence of metabolic syndrome despite lower body mass index. International Journal of Obesity. 2011; 35(3):393–400. https://doi.org/10.1038/ijo.2010.152. PMid:20680014. PMCid:PMC2989340

Hirode G, Wong RJ. Trends in the prevalence of metabolic syndrome in the United States, 2011-2016. Journal of the American Medical Association. 2020; 323(24):2526. https://doi.org/10.1001/jama.2020.4501. PMid:32573660. PMCid:PMC7312413

Krishnamoorthy Y, Rajaa S, Murali S, Rehman T, Sahoo J, Kar SS. Prevalence of metabolic syndrome among adult population in India: A systematic review and meta-analysis. PLOS ONE. 2020; 15(10). https://doi.org/10.1371/journal.pone.0240971. PMid:33075086. PMCid:PMC7571716

Delavari A, Forouzanfar MH, Alikhani S, et al. First nationwide study of the prevalence of the metabolic syndrome and optimal cutoff points of waist circumference in the Middle East. Diabetes Care. 2009; 32(6):1092–7. https://doi.org/10.2337/dc08-1800. PMid:19279302. PMCid:PMC2681035

Wang Y, Mi J, Shan X, et al. Is China facing an obesity epidemic and the consequences? The trends in obesity and chronic disease in China. International Journal of Obesity. 2007; 31(1):177–88. https://doi.org/10.1038/sj.ijo.0803354. PMid:16652128

Daniele G, Mendoza GR, Winnier D, et al. The inflammatory status score including IL-6, TNF-κ, osteopontin, fractalkine, MCP-1 and adiponectin underlies whole-body insulin resistance and hyperglycemia in type 2 diabetes mellitus. Acta Diabetologica. 2014; 51(1):123–31. https://doi.org/10.1007/s00592-013-0543-1. PMid:24370923

Chandra A, Neeland IJ, Berry JD, et al. The relationship of body mass and fat distribution with incident hypertension. Journal of the American College of Cardiology 2014; 64(10):997–1002. https://doi.org/10.1016/j.jacc.2014.05.057. PMid:25190234

Zorena K, Jachimowicz-Duda O, W?? P. The cut-off value for interleukin 34 as an additional potential inflammatory biomarker for the prediction of the risk of diabetic complications. Biomarkers. 2016; 21(3):276–82. https://doi.org/10.3109/13547 50X.2016.1138321. PMid:26849008

Körner A, Kratzsch J, Gausche R, et al. New predictors of the metabolic syndrome in children —Role of Adipocytokines. Pediatric Research. 2007; 61(6):640–5. https://doi.org/10.1203/01.pdr.0000262638.48304.ef. PMid:17426657

Lago F, Gómez R, Gómez-Reino JJ, et al. Adipokines as novel modulators of lipid metabolism. Trends in Biochemical Sciences. 2009; 34(10):500–10. https://doi.org/10.1016/j.tibs.2009.06.008. PMid:19729309

Farooq R, Amin S, Hayat Bhat M, et al. Type 2 diabetes and metabolic syndrome—adipokine levels and effect of drugs. Gynecological Endocrinology. 2017; 33(1):75–8. https://doi.org/10.1080/09513590.2016.1207165. PMid:27705028

Misra A, Soares MJ, Mohan V, et al. Body fat, metabolic syndrome and hyperglycemia in South Asians. Journal of Diabetes and its Complications. 2018; 32(11):1068–75. https://doi.org/10.1016/j.jdiacomp.2018.08.001. PMid:30115487

International Diabetes Federation: IDF Diabetes Atlas (2017). 8th ed; 2017.

Joshi SR, Anjana RM, Deepa M, et al. Prevalence of dyslipidemia in urban and rural India: The ICMR-INDIAB Study. PLoS ONE. 2014; 9(5). https://doi.org/10.1371/journal.pone.0096808. PMid:24817067. PMCid:PMC4016101

Anjana RM, Deepa M, Pradeepa R, et al. Prevalence of diabetes and prediabetes in 15 states of India: Results from the ICMRINDIAB population-based cross-sectional study. The Lancet Diabetes and Endocrinology. 2017; 5(8):585–96. https://doi. org/10.1016/S2213-8587(17)30174-2

Gupta R, Rao RS, Misra A, et al. Recent trends in epidemiology of dyslipidemias in India. Indian Heart Journal. 2017; 69(3):382– 92. https://doi.org/10.1016/j.ihj.2017.02.020. PMid:28648438. PMCid:PMC5485409

Madeo F, Eisenberg T, Pietrocola F, et al. Spermidine in health and disease. Science. 2018; 359(6374). https://doi.org/10.1126/ science.aan2788. PMid:29371440

Tabor CW, Tabor H. Methionine Adenosyltransferase (S -Adenosylmethionine Synthetase) and S -Adenosyl methionine Decarboxylase. 2006. p. 251–82. https://doi.org/10.1002/9780470123027.ch4. PMid:6364703

Wallace HM, Nuttall ME, Coleman CS. Polyamine recycling enzymes in human cancer cells; 1988. p. 331–44. https://doi.org/10.1007/978-1-4684-5637-0_29. PMid:3076329

Dudkowska M, Lai J, Gardini G, et al. Agmatine modulates the in vivo biosynthesis and interconversion of polyamines and cell proliferation. Biochimica et Biophysica Acta (BBA) – General Subjects. 2003; 1619(2):159–66. https://doi.org/10.1016/S0304-4165(02)00476-2

Ramos-Molina B, Queipo-Ortuño MI, Lambertos A, et al. Dietary and gut microbiota polyamines in obesity- and age-related diseases. Frontiers in Nutrition. 2019; 6. https://doi.org/10.3389/fnut.2019.00024. PMid:30923709. PMCid:PMC6426781

Kramer DL, Diegelman P, Jell J, et al. Polyamine acetylation modulates polyamine metabolic flux, a prelude to broader metabolic consequences. Journal of Biological Chemistry. 2008; 283(7):4241–51. https://doi.org/10.1074/jbc.M706806200. PMid:18089555

Sivashanmugam M, Jaidev J, Umashankar V, Sulochana KN. Ornithine and its role in metabolic diseases: An appraisal. Biomedicine and Pharmacotherapy. 2017; 86:185–94. https://doi.org/10.1016/j.biopha.2016.12.024. PMid:27978498

Welsh N. A role for polyamines in glucose-stimulated insulin-gene expression. Biochemical Journal. 1990; 271(2):393–7. https://doi.org/10.1042/bj2710393. PMid:2241922. PMCid:PMC1149567

Welsh N, Sjöholm A. Polyamines and insulin production in isolated mouse pancreatic islets. Biochemical Journal. 1988; 252(3):701–7. https://doi.org/10.1042/bj2520701. PMid:3138973. PMCid:PMC1149205

Mastracci TL, Robertson MA, Mirmira RG, et al. Polyamine biosynthesis is critical for growth and differentiation of the pancreas. Scientific Reports. 2015; 5(1). https://doi.org/10.1038/srep13269 PMid:26299433. PMCid:PMC4547391

Fernandez-Garcia J, Delpino-Rius A, Samarra I, et al. Type 2 Diabetes is associated with a different pattern of serum polyamines: A case-control study from the PREDIMED-Plus trial. Journal of Clinical Medicine. 2019; 8(1):71. https://doi.org/10.3390/ jcm8010071. PMid:30634588. PMCid:PMC6352090

Sharawy MH, El-Awady MS, Megahed N, et al. Attenuation of insulin resistance in rats by agmatine: role of SREBP-1c, mTOR and GLUT-2. Naunyn-Schmiedeberg’s Archives of Pharmacology. 2016; 389(1):45–56. https://doi.org/10.1007/s00210-015-1174-6. PMid:26449613

Niiranen K, Keinänen TA, Pirinen E, et al. Mice with targeted disruption of spermidine/spermine N 1 -acetyltransferase gene maintain nearly normal tissue polyamine homeostasis but show signs of insulin resistance upon aging. Journal of Cellular and Molecular Medicine. 2006; 10(4):933–45.PMid:17125596

Pirinen E, Kuulasmaa T, Pietila? M, et al. Enhanced polyamine catabolism alters homeostatic control of white adipose tissue mass, energy expenditure, and glucose metabolism. Molecular and Cellular Biology. 2007; 27(13):4953–67. https://doi.org/10.1128/ MCB.02034-06. PMid:17485446. PMCid:PMC1951486

Cerrada-Gimenez M, Tusa M, Casellas A, et al. Altered glucose-stimulated insulin secretion in a mouse line with activated polyamine catabolism. Transgenic Research. 2012; 21(4):843–53. https://doi.org/10.1007/s11248-011-9579-6. PMid:22180015

Yuan F, Zhang L, Cao Y, et al. Spermidine/spermine N1-acetyltransferase-mediated polyamine catabolism regulates beige adipocyte biogenesis. Metabolism. 2018; 85:298–304. https://doi.org/10.1016/j.metabol.2018.04.007. PMid:29715464. PMCid:PMC7269456

Codoñer-Franch P, Valls-Bellés V, Arilla-Codoñer A, Alonso-Iglesias E. Oxidant mechanisms in childhood obesity: the link between inflammation and oxidative stress. Translational Research. 2011; 158(6):369–84. https://doi.org/10.1016/j.trsl.2011.08.004. PMid:22061044

Codoñer-Franch P, Tavárez-Alonso S, Murria-Estal R, et al. Polyamines are increased in obese children and are related to markers of oxidative/nitrosative stress and angiogenesis. The Journal of Clinical Endocrinology and Metabolism. 2011; 96(9):2821–5. https://doi.org/10.1210/jc.2011-0531. PMid:21697248

Gao M, Zhao W, Li C, et al. Spermidine ameliorates non-alcoholic fatty liver disease through regulating lipid metabolism via AMPK. Biochemical and Biophysical Research Communications. 2018; 505(1):93–8. https://doi.org/10.1016/j.bbrc.2018.09.078. PMid:30241944

Sadasivan SK, Vasamsetti B, Singh J, et al. Exogenous administration of spermine improves glucose utilization and decreases bodyweight in mice. European Journal of Pharmacology. 2014; 729:94–9. https://doi.org/10.1016/j.ejphar.2014.01.073. PMid:24530553

Fernández ÁF, Bárcena C, Martínez-García GG, Tamargo-Gómez I, et al. Autophagy couteracts weight gain, lipotoxicity and pancreatic β-cell death upon hypercaloric pro-diabetic regimens. Cell Death and Disease. 2017; 8(8). https://doi.org/10.1038/ cddis.2017.373. PMid:28771229. PMCid:PMC5596561

Vuohelainen S, Pirinen E, Cerrada-Gimenez M, et al. Spermidine is indispensable in differentiation of 3T3-L1 fibroblasts to adipocytes. Journal of Cellular and Molecular Medicine. 2009; 14(6b):1683–92. https://doi.org/10.1111/j.1582-4934.2009.00808.x. PMid:19538475. PMCid:PMC3829030

Pillion DJ. Differential effects of insulin, antibodies against rat adipocyte plasma membranes, and other agents that mimic insulin action in rat adipocytes. Metabolism. 1985; 34(11):1012–9. https://doi.org/10.1016/0026-0495(85)90072-1

Ma L, Ni Y, Hu L, et al. Spermidine ameliorates high-fat diet-induced hepatic steatosis and adipose tissue inflammation in preexisting obese mice. Life Sciences. 2021; 265. https://doi.org/10.1016/j.lfs.2020.118739. PMid:33186567

Ocaña-Wilhelmi L, Cardona F, Garrido-Sanchez L, et al. Change in serum polyamine metabolome pattern after bariatric surgery in obese patients with metabolic syndrome. Surgery for Obesity and Related Diseases. 2020; 16(2):306–11. https://doi.org/10.1016/j. soard.2019.10.024. PMid:31813775