Asprosin: A Novel Player in Metabolic Diseases

Mingyang Yuan, Weidong Li, Yan Zhu, Boyao Yu, Jing Wu, Mingyang Yuan, Weidong Li, Yan Zhu, Boyao Yu, Jing Wu

Abstract

Asprosin, a novel glucogenic adipokine, is encoded by two exons (exon 65 and exon 66) of the gene Fibrillin 1 (FBN1) and mainly synthesized and released by white adipose tissue during fasting. Asprosin plays a complex role in the central nervous system (CNS), peripheral tissues, and organs. It is involved in appetite, glucose metabolism, insulin resistance (IR), cell apoptosis, etc. In this review, we will summarize the newly discovered roles of asprosin in metabolic diseases including diabetes, obesity, polycystic ovarian syndrome (PCOS), and cardiovascular disease (CVD), which may contribute to future clinical diagnosis and treatment.

Keywords: CVD; PCOS; asprosin; diabetes; metabolic diseases; obesity.

Copyright © 2020 Yuan, Li, Zhu, Yu and Wu.

Figures

Figure 1
Figure 1
The possible central and peripheral effects of asprosin. In the hypothalamus, asprosin enhances the activity of AgRP neurons through activating G proteins-cAMP-PKA pathway, leading to increased appetite. In the liver, asprosin binds to OLFR734 to enhance the production and release of glucose via CREB pathway. Of note, the orexigenic and glucogenic roles of asprosin remain controversial. In the pancreatic β-cells, asprosin binds to TLR4 to promote inflammation and apoptosis of these cells and reduce insulin secretion through stimulating TLR4/JNK-mediated pathway and inhibiting cAMP levels. In MSCs, asprosin prevents these cells from oxidative stress-induced apoptosis and death by activating ERK1/2-SOD2 pathway. In the skeletal muscle, asprosin activates PKCδ/SERCA2-mediated ER stress/inflammation pathways to promote IR.
Figure 2
Figure 2
The newly discovered roles of asprosin in metabolic diseases. In diabetes, higher levels of asprosin can be observed in patients as compared to controls, and there is a positive association between serum asprosin levels and IR. Asprosin seems to show an impaired response to glucose fluctuation in both T1DM and T2DM patients. In addition, asprosin may act as a biomarker to predict T2DM onset and a novel target in the treatment of diabetes. In obesity, likewise, there are higher levels of asprosin in adult patients in comparison with controls, and asprosin may be a therapeutic target against obesity. However, the correlation between asprosin and obese children is considered as inconclusive. In PCOS, asprosin shows an inconclusive relationship to it. In CVD, asprosin may predict the severity of ACS, and it presents a possible strategy against DCM and MI.

References

    1. Romere C, Duerrschmid C, Bournat J, Constable P, Jain M, Xia F, et al. . Asprosin, a fasting-induced glucogenic protein hormone. Cell. (2016) 165:566–79. 10.1016/j.cell.2016.02.063
    1. Sakai LY, Keene DR, Renard M, De Backer J. FBN1: the disease-causing gene for marfan syndrome and other genetic disorders. Gene. (2016) 591:279–91. 10.1016/j.gene.2016.07.033
    1. Lonnqvist L, Reinhardt D, Sakai L, Peltonen L. Evidence for furin-type activity-mediated C-terminal processing of profibrillin-1 and interference in the processing by certain mutations. Hum Mol Genet. (1998) 7:2039–44. 10.1093/hmg/7.13.2039
    1. Milewicz DM, Grossfield J, Cao SN, Kielty C, Covitz W, Jewett T. A mutation in FBN1 disrupts profibrillin processing and results in isolated skeletal features of the marfan syndrome. J Clin Invest. (1995) 95:2373–8. 10.1172/JCI117930
    1. Lee T, Yun S, Jeong JH, Jung TW. Asprosin impairs insulin secretion in response to glucose and viability through TLR4/JNK-mediated inflammation. Mol Cell Endocrinol. (2019) 486:96–104. 10.1016/j.mce.2019.03.001
    1. Duerrschmid C, He Y, Wang C, Li C, Bournat JC, Romere C, et al. . Asprosin is a centrally acting orexigenic hormone. Nat Med. (2017) 23:1444–53. 10.1038/nm.4432
    1. Zhang Z, Tan Y, Zhu L, Zhang B, Feng P, Gao E, et al. . Asprosin improves the survival of mesenchymal stromal cells in myocardial infarction by inhibiting apoptosis via the activated ERK1/2-SOD2 pathway. Life Sci. (2019) 231:116554. 10.1016/j.lfs.2019.116554
    1. Jung TW, Kim HC, Kim HU, Park T, Park J, Kim U, et al. . Asprosin attenuates insulin signaling pathway through PKCdelta-activated ER stress and inflammation in skeletal muscle. J Cell Physiol. (2019) 234:20888–99. 10.1002/jcp.28694
    1. Feng J, Yang Y, Yang Y, Pei H. The protective role of Asprosin against diabetes in cardiomyocytes. J Am Colle Cardiol. (2018) 72:C2 10.1016/j.jacc.2018.08.012
    1. Sohn JW. Network of hypothalamic neurons that control appetite. BMB Rep. (2015) 48:229–33. 10.5483/BMBRep.2015.48.4.272
    1. Li E, Shan H, Chen L, Long A, Zhang Y, Liu Y, et al. . OLFR734 Mediates glucose metabolism as a receptor of asprosin. Cell Metabol. (2019) 30:319–28.e8. 10.1016/j.cmet.2019.05.022
    1. von Herrath M, Pagni PP, Grove K, Christoffersson G, Tang-Christensen M, Karlsen AE, et al. . Case reports of pre-clinical replication studies in metabolism and diabetes. Cell Metabol. (2019) 29:795–802. 10.1016/j.cmet.2019.02.004
    1. Amer Diabet A. Diagnosis and classification of diabetes mellitus. Diabetes Care. (2014) 37:S81–90. 10.2337/dc14-S081
    1. Wang Y, Qu H, Xiong X, Qiu Y, Liao Y, Chen Y, et al. . Plasma asprosin concentrations are increased in individuals with glucose dysregulation and correlated with insulin resistance and first-phase insulin secretion. Med Inflam. (2018) 2018:9471583. 10.1155/2018/9471583
    1. Zhang L, Chen C, Zhou N, Fu Y, Cheng X. Circulating asprosin concentrations are increased in type 2 diabetes mellitus and independently associated with fasting glucose and triglyceride. Clin Chim Acta. (2019) 489:183–8. 10.1016/j.cca.2017.10.034
    1. Zhang X, Jiang H, Ma X, Wu H. Increased serum level and impaired response to glucose fluctuation of asprosin is associated with type 2 diabetes mellitus. J Diabetes Investig. (2019). 10.1111/jdi.13148. [Epub ahead of print].
    1. Li X, Liao M, Shen R, Zhang L, Hu H, Wu J, et al. . Plasma asprosin levels are associated with glucose metabolism, lipid, and sex hormone profiles in females with metabolic-related diseases. Med Inflam. (2018) 2018:7375294. 10.1155/2018/7375294
    1. Groener JB, Valkanou A, Kender Z, Pfeiffenberger J, Kihm L, Fleming T, et al. Asprosin response in hypoglycemia is not related to hypoglycemia unawareness but rather to insulin resistance in type 1 diabetes. PLoS ONE. (2019) 14:e0222771 10.1371/journal.pone.0222771
    1. Baykus Y, Yavuzkir S, Ustebay S, Ugur K, Deniz R, Aydin S. Asprosin in umbilical cord of newborns and maternal blood of gestational diabetes, preeclampsia, severe preeclampsia, intrauterine growth retardation and macrosemic fetus. Peptides. (2019) 120:170132. 10.1016/j.peptides.2019.170132
    1. Ko JR, Seo DY, Kim TN, Park SH, Kwak HB, Ko KS, et al. . Aerobic exercise training decreases hepatic asprosin in diabetic rats. J Clin Med. (2019) 8:666. 10.3390/jcm8050666
    1. Titchenell PM, Lazar MA, Birnbaum MJ. Unraveling the regulation of hepatic metabolism by insulin. Trends Endocrinol Metab. (2017) 28:497–505. 10.1016/j.tem.2017.03.003
    1. Unger RH. Lipid overload and overflow: metabolic trauma and the metabolic syndrome. Trends Endocrinol Metab. (2003) 14:398–403. 10.1016/j.tem.2003.09.008
    1. Wang CY, Lin TA, Liu KH, Liao CH, Liu YY, Wu VC, et al. . Serum asprosin levels and bariatric surgery outcomes in obese adults. Int J Obesity. (2019) 43:1019–25. 10.1038/s41366-018-0248-1
    1. Wang M, Yin C, Wang L, Liu Y, Li H, Li M, et al. . Serum asprosin concentrations are increased and associated with insulin resistance in children with obesity. Ann Nutr Metab. (2019) 75:205–12. 10.1159/000503808
    1. Ugur K, Aydin S. Saliva and blood asprosin hormone concentration associated with obesity. Int J Endocrinol. (2019) 2019:2521096. 10.1155/2019/2521096
    1. Ma H, Zhang G, Mou C, Fu X, Chen Y. Peripheral CB1 receptor neutral antagonist, AM6545, ameliorates hypometabolic obesity and improves adipokine secretion in monosodium glutamate induced obese mice. Front Pharmacol. (2018) 9:156. 10.3389/fphar.2018.00156
    1. Long W, Xie X, Du C, Zhao Y, Zhang C, Zhan D, et al. . Decreased circulating levels of asprosin in obese children. Horm Res Paediatr. (2019) 91:271–7. 10.1159/000500523
    1. Escobar-Morreale HF. Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment. Nat Rev Endocrinol. (2018) 14:270–84. 10.1038/nrendo.2018.24
    1. Alan M, Gurlek B, Yilmaz A, Aksit M, Aslanipour B, Gulhan I, et al. . Asprosin: a novel peptide hormone related to insulin resistance in women with polycystic ovary syndrome. Gynecol Endocrinol. (2019) 35:220–3. 10.1080/09513590.2018.1512967
    1. Chang CL, Huang SY, Hsu YC, Chin TH, Soong YK. The serum level of irisin, but not asprosin, is abnormal in polycystic ovary syndrome patients. Sci Rep. (2019) 9:6447 10.1038/s41598-019-42061-9
    1. Finegold JA, Asaria P, Francis DP. Mortality from ischaemic heart disease by country, region, and age: statistics from world health organisation and United Nations. Int J Cardiol. (2013) 168:934–45. 10.1016/j.ijcard.2012.10.046
    1. Kappetein AP, Dawkins KD, Mohr FW, Morice MC, Mack MJ, Russell ME, et al. . Current percutaneous coronary intervention and coronary artery bypass grafting practices for three-vessel and left main coronary artery disease. insights from the SYNTAX run-in phase. Eur J Cardiothorac Surg. (2006) 29:486–91. 10.1016/j.ejcts.2006.01.047
    1. Acara AC, Bolatkale M, Kiziloglu I, Ibisoglu E, Can C. A novel biochemical marker for predicting the severity of ACS with unstable angina pectoris: asprosin. Am J Emerg Med. (2018) 36:1504–5. 10.1016/j.ajem.2017.12.032
    1. Yu SP, Wei Z, Wei L. Preconditioning strategy in stem cell transplantation therapy. Transl Stroke Res. (2013) 4:76–88. 10.1007/s12975-012-0251-0

Source: PubMed

3
Abonner