DOI QR코드

DOI QR Code

Neuronatin Is Associated with an Anti-Inflammatory Role in the White Adipose Tissue

  • Ka, Hye In (Department of Biological Sciences, Sookmyung Women's University) ;
  • Han, Sora (Department of Biological Sciences, Sookmyung Women's University) ;
  • Jeong, Ae Lee (Department of Biological Sciences, Sookmyung Women's University) ;
  • Lee, Sunyi (Department of Biological Sciences, Sookmyung Women's University) ;
  • Yong, Hyo Jeong (Department of Biological Sciences, Sookmyung Women's University) ;
  • Boldbaatar, Ariundavaa (Department of Biological Sciences, Sookmyung Women's University) ;
  • Joo, Hyun Jeong (Department of Biological Sciences, Sookmyung Women's University) ;
  • Soh, Su Jung (Department of Biological Sciences, Sookmyung Women's University) ;
  • Park, Ji Young (Department of Biological Sciences, Sookmyung Women's University) ;
  • Lim, Jong-Seok (Department of Biological Sciences, Sookmyung Women's University) ;
  • Lee, Myung Sok (Department of Biological Sciences, Sookmyung Women's University) ;
  • Yang, Young (Department of Biological Sciences, Sookmyung Women's University)
  • Received : 2017.02.17
  • Accepted : 2017.03.24
  • Published : 2017.06.28

Abstract

Neuronatin (NNAT) is known to regulate ion channels during brain development and plays a role in maintaining the structure of the nervous system. A previous in silico analysis showed that Nnat was overexpressed in the adipose tissue of an obese rodent model relative to the wild type. Therefore, the aim of the present study was to investigate the function of Nnat in the adipose tissue. Because obesity is known to systemically induce low-grade inflammation, the Nnat expression level was examined in the adipose tissue obtained from C57BL/6 mice administered lipopolysaccharide (LPS). Unexpectedly, the Nnat expression level decreased in the white adipose tissue after LPS administration. To determine the role of NNAT in inflammation, 3T3-L1 cells overexpressing Nnat were treated with LPS. The level of the p65 subunit of nuclear factor-kappa B ($NF-{\kappa}B$) and the activity of $NF-{\kappa}B$ luciferase decreased following LPS treatment. These results indicate that NNAT plays an anti-inflammatory role in the adipose tissue.

Keywords

References

  1. Kahn BB, Flier JS. 2000. Obesity and insulin resistance. J. Clin. Invest. 106: 473-481. https://doi.org/10.1172/JCI10842
  2. Harvey AE, Lashinger LM, Hursting SD. 2011. The growing challenge of obesity and cancer: an inflammatory issue. Ann. NY Acad. Sci. 1229: 45-52. https://doi.org/10.1111/j.1749-6632.2011.06096.x
  3. Basen-Engquist K, Chang M. 2011. Obesity and cancer risk: recent review and evidence. Curr. Oncol. Rep. 13: 71-76. https://doi.org/10.1007/s11912-010-0139-7
  4. Greenberg AS, Obin MS. 2006. Obesity and the role of adipose tissue in inflammation and metabolism. Am. J. Clin. Nutr. 83: 461S-465S. https://doi.org/10.1093/ajcn/83.2.461S
  5. Lumeng CN, Saltiel AR. 2011. Inflammatory links between obesity and metabolic disease. J. Clin. Invest. 121: 2111-2117. https://doi.org/10.1172/JCI57132
  6. Hotamisligil GS. 2006. Inflammation and metabolic disorders. Nature 444: 860-867. https://doi.org/10.1038/nature05485
  7. Moller DE. 2000. Potential role of TNF-alpha in the pathogenesis of insulin resistance and type 2 diabetes. Trends Endocrinol. Metab. 11: 212-217. https://doi.org/10.1016/S1043-2760(00)00272-1
  8. Panee J. 2012. Monocyte chemoattractant protein 1 (MCP-1) in obesity and diabetes. Cytokine 60: 1-12. https://doi.org/10.1016/j.cyto.2012.06.018
  9. Perreault M, Marette A. 2001. Targeted disruption of inducible nitric oxide synthase protects against obesitylinked insulin resistance in muscle. Nat. Med. 7: 1138-1143. https://doi.org/10.1038/nm1001-1138
  10. Dou D, Joseph R. 1996. Cloning of human neuronatin gene and its localization to chromosome-20q 11.2-12: the deduced protein is a novel "proteolipid". Brain Res. 723: 8-22. https://doi.org/10.1016/0006-8993(96)00167-9
  11. Kagitani F, Kuroiwa Y, Wakana S, Shiroishi T, Miyoshi N, Kobayashi S, et al. 1997. Peg5/neuronatin is an imprinted gene located on sub-distal chromosome 2 in the mouse. Nucleic Acids Res. 25: 3428-3432. https://doi.org/10.1093/nar/25.17.3428
  12. Joseph R, Dou D, Tsang W. 1995. Neuronatin mRNA: alternatively spliced forms of a novel brain-specific mammalian developmental gene. Brain Res. 690: 92-98. https://doi.org/10.1016/0006-8993(95)00621-V
  13. Pitale PM, Howse W, Gorbatyuk M. 2017. Neuronatin protein in health and disease. J. Cell. Physiol. 232: 477-481. https://doi.org/10.1002/jcp.25498
  14. Vrang N, Meyre D, Froguel P, Jelsing J, Tang-Christensen M, Vatin V, et al. 2010. The imprinted gene neuronatin is regulated by metabolic status and associated with obesity. Obesity 18: 1289-1296. https://doi.org/10.1038/oby.2009.361
  15. Chu K, Tsai MJ. 2005. Neuronatin, a downstream target of BETA2/NeuroD1 in the pancreas, is involved in glucosemediated insulin secretion. Diabetes 54: 1064-1073. https://doi.org/10.2337/diabetes.54.4.1064
  16. Joe MK, Lee HJ, Suh YH, Han KL, Lim JH, Song J, et al. 2008. Crucial roles of neuronatin in insulin secretion and high glucose-induced apoptosis in pancreatic beta-cells. Cell Signal. 20: 907-915. https://doi.org/10.1016/j.cellsig.2008.01.005
  17. Suh YH, Kim WH, Moon C, Hong YH, Eun SY, Lim JH, et al. 2005. Ectopic expression of neuronatin potentiates adipogenesis through enhanced phosphorylation of cAMPresponse element-binding protein in 3T3-L1 cells. Biochem. Biophys. Res. Commun. 337: 481-489. https://doi.org/10.1016/j.bbrc.2005.09.078
  18. Margetic S, Gazzola C, Pegg GG, Hill RA. 2002. Leptin: a review of its peripheral actions and interactions. Int. J. Obes. Relat. Metab. Disord. 26: 1407-1433. https://doi.org/10.1038/sj.ijo.0802142
  19. Klok MD, Jakobsdottir S, Drent ML. 2007. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: a review. Obes. Rev. 8: 21-34. https://doi.org/10.1111/j.1467-789X.2006.00270.x
  20. Berndt J, Kovacs P, Ruschke K, Kloting N, Fasshauer M, Schon MR, et al. 2007. Fatty acid synthase gene expression in human adipose tissue: association with obesity and type 2 diabetes. Diabetologia 50: 1472-1480. https://doi.org/10.1007/s00125-007-0689-x
  21. Krogh A, Larsson B, von Heijne G, Sonnhammer EL. 2001. Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes. J. Mol. Biol. 305: 567-580. https://doi.org/10.1006/jmbi.2000.4315
  22. Tak PP, Firestein GS. 2001. NF-kappaB: a key role in inflammatory diseases. J. Clin. Invest. 107: 7-11. https://doi.org/10.1172/JCI11830
  23. Mzhavia N, Yu S, Ikeda S, Chu TT, Goldberg I, Dansky HM. 2008. Neuronatin: a new inflammation gene expressed on the aortic endothelium of diabetic mice. Diabetes 57: 2774-2783. https://doi.org/10.2337/db07-1746
  24. Weinberg MB, Utter MF. 1979. Effect of thyroid hormone on the turnover of rat liver pyruvate carboxylase and pyruvate dehydrogenase. J. Biol. Chem. 254: 9492-9499.
  25. Jitrapakdee S, St Maurice M, Rayment I, Cleland WW, Wallace JC, Attwood PV. 2008. Structure, mechanism and regulation of pyruvate carboxylase. Biochem. J. 413: 369-387. https://doi.org/10.1042/BJ20080709
  26. Jitrapakdee S, Gong Q, MacDonald MJ, Wallace JC. 1998. Regulation of rat pyruvate carboxylase gene expression by alternate promoters during development, in genetically obese rats and in insulin-secreting cells. Multiple transcripts with 5'-end heterogeneity modulate translation. J. Biol. Chem. 273: 34422-34428. https://doi.org/10.1074/jbc.273.51.34422
  27. Adib-Conquy M, Cavaillon JM. 2009. Compensatory antiinflammatory response syndrome. Thromb. Haemost. 101: 36-47. https://doi.org/10.1160/TH08-07-0421
  28. Liu TF, McCall CE. 2013. Deacetylation by SIRT1 reprograms inflammation and cancer. Genes Cancer 4: 135-147. https://doi.org/10.1177/1947601913476948
  29. Zandi E, Rothwarf DM, Delhase M, Hayakawa M, Karin M. 1997. The IkappaB kinase complex (IKK) contains two kinase subunits, IKKalpha and IKKbeta, necessary for IkappaB phosphorylation and NF-kappaB activation. Cell 91: 243-252. https://doi.org/10.1016/S0092-8674(00)80406-7
  30. Li QT, Verma IM. 2002. NF-kappa B regulation in the immune system. Nat. Rev. Immunol. 2: 725-734. https://doi.org/10.1038/nri910
  31. Lawrence T. 2009. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 1: a001651.
  32. Bonizzi G, Karin M. 2004. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 25: 280-288. https://doi.org/10.1016/j.it.2004.03.008
  33. Baker RG, Hayden MS, Ghosh S. 2011. NF-kappaB, inflammation, and metabolic disease. Cell Metab. 13: 11-22. https://doi.org/10.1016/j.cmet.2010.12.008
  34. Christian F, Smith EL, Carmody RJ. 2016. The regulation of NF-kappaB subunits by phosphorylation. Cells 5: E12. https://doi.org/10.3390/cells5010012
  35. Chen L, Fischle W, Verdin E, Greene WC. 2001. Duration of nuclear NF-kappaB action regulated by reversible acetylation. Science 293: 1653-1657. https://doi.org/10.1126/science.1062374
  36. Kiernan R, Bres V, Ng RW, Coudart MP, El Messaoudi S, Sardet C, et al. 2003. Post-activation turn-off of NF-kappa Bdependent transcription is regulated by acetylation of p65. J. Biol. Chem. 278: 2758-2766. https://doi.org/10.1074/jbc.M209572200
  37. Buerki C, Rothgiesser KM, Valovka T, Owen HR, Rehrauer H, Fey M, et al. 2008. Functional relevance of novel p300- mediated lysine 314 and 315 acetylation of RelA/p65. Nucleic Acids Res. 36: 1665-1680. https://doi.org/10.1093/nar/gkn003
  38. Rothgiesser KM, Fey M, Hottiger MO. 2010. Acetylation of p65 at lysine 314 is important for late NF-kappaBdependent gene expression. BMC Genomics 11: 22. https://doi.org/10.1186/1471-2164-11-22

Cited by

  1. Sex dependent impact of gestational stress on predisposition to eating disorders and metabolic disease vol.17, pp.None, 2017, https://doi.org/10.1016/j.molmet.2018.08.005
  2. Dynamic Changes in the Localization of Neuronatin-Positive Cells during Neurogenesis in the Embryonic Rat Brain vol.207, pp.3, 2017, https://doi.org/10.1159/000504359
  3. PPAR γ/Nnat/NF-κB Axis Involved in Promoting Effects of Adiponectin on Preadipocyte Differentiation vol.2019, pp.None, 2017, https://doi.org/10.1155/2019/5618023
  4. MiR-3064 in Epicardial Adipose-Derived Exosomes Targets Neuronatin to Regulate Adipogenic Differentiation of Epicardial Adipose Stem Cells vol.8, pp.None, 2017, https://doi.org/10.3389/fcvm.2021.709079
  5. Single-cell RNA sequencing analysis of mouse follicular somatic cells vol.105, pp.5, 2017, https://doi.org/10.1093/biolre/ioab163
  6. Strain-based and sex-biased differences in adrenal and pancreatic gene expression between KK/HlJ and C57BL/6 J mice vol.22, pp.1, 2017, https://doi.org/10.1186/s12864-021-07495-4
  7. Bisphenol A induces miR-708-5p through an ER stress-mediated mechanism altering neuronatin and neuropeptide Y expression in hypothalamic neuronal models vol.539, pp.None, 2017, https://doi.org/10.1016/j.mce.2021.111480