DOI QR코드

DOI QR Code

Nervonic Acid Inhibits Replicative Senescence of Human Wharton's Jelly-Derived Mesenchymal Stem Cells

  • Sun Jeong Kim (Cell and Gene Therapy Institute, ENCell Co. Ltd.) ;
  • Soojin Kwon (Cell and Gene Therapy Institute, ENCell Co. Ltd.) ;
  • Soobeen Chung (Cell and Gene Therapy Institute, ENCell Co. Ltd.) ;
  • Eun Joo Lee (Cell and Gene Therapy Institute, ENCell Co. Ltd.) ;
  • Sang Eon Park (Cell and Gene Therapy Institute, ENCell Co. Ltd.) ;
  • Suk-Joo Choi (Department of Obstetrics and Gynecology, Samsung Medical Center) ;
  • Soo-Young Oh (Department of Obstetrics and Gynecology, Samsung Medical Center) ;
  • Gyu Ha Ryu (Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University) ;
  • Hong Bae Jeon (Cell and Gene Therapy Institute, ENCell Co. Ltd.) ;
  • Jong Wook Chang (Cell and Gene Therapy Institute, ENCell Co. Ltd.)
  • Received : 2023.06.28
  • Accepted : 2023.08.01
  • Published : 2024.02.28

Abstract

Cellular senescence causes cell cycle arrest and promotes permanent cessation of proliferation. Since the senescence of mesenchymal stem cells (MSCs) reduces proliferation and multipotency and increases immunogenicity, aged MSCs are not suitable for cell therapy. Therefore, it is important to inhibit cellular senescence in MSCs. It has recently been reported that metabolites can control aging diseases. Therefore, we aimed to identify novel metabolites that regulate the replicative senescence in MSCs. Using a fecal metabolites library, we identified nervonic acid (NA) as a candidate metabolite for replicative senescence regulation. In replicative senescent MSCs, NA reduced senescence-associated 𝛽-galactosidase positive cells, the expression of senescence-related genes, as well as increased stemness and adipogenesis. Moreover, in non-senescent MSCs, NA treatment delayed senescence caused by sequential subculture and promoted proliferation. We confirmed, for the first time, that NA delayed and inhibited cellular senescence. Considering optimal concentration, duration, and timing of drug treatment, NA is a novel potential metabolite that can be used in the development of technologies that regulate cellular senescence.

Keywords

Acknowledgement

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2021R1F1A1064060) and supported by the Korean Fund for Regenerative Medicine funded by the Ministry of Science and ICT, and Ministry of Health and Welfare (RS-2022-00060268). This research was also supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF) funded by the Ministry of Science & ICT (RS-2023-00223069) and supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HR22C1363). This work was partly supported by the Collabo R&D between Industry, Academy, and Research Institute of MSS (S3098634), and by Samsung Medical Center (SMO1230051).

References

  1. Hernandez-Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence. Trends Cell Biol 2018;28:436-453.
  2. Kim SJ, Park SE, Jeong JB, et al. Wharton's jelly-derived mesenchymal stem cells with high aurora kinase A expression show improved proliferation, migration, and therapeutic potential. Stem Cells Int 2022;2022:4711499.
  3. Weng Z, Wang Y, Ouchi T, et al. Mesenchymal stem/stromal cell senescence: hallmarks, mechanisms, and combating strategies. Stem Cells Transl Med 2022;11:356-371.
  4. Zhou X, Hong Y, Zhang H, Li X. Mesenchymal stem cell senescence and rejuvenation: current status and challenges. Front Cell Dev Biol 2020;8:364.
  5. Liu J, Ding Y, Liu Z, Liang X. Senescence in mesenchymal stem cells: functional alterations, molecular mechanisms, and rejuvenation strategies. Front Cell Dev Biol 2020;8:258.
  6. Sharma R. Emerging interrelationship between the gut microbiome and cellular senescence in the context of aging and disease: perspectives and therapeutic opportunities. Probiotics Antimicrob Proteins 2022;14:648-663.
  7. Parker A, Romano S, Ansorge R, et al. Fecal microbiota transfer between young and aged mice reverses hallmarks of the aging gut, eye, and brain. Microbiome 2022;10:68.
  8. Zheng W, Kollmeyer J, Symolon H, et al. Ceramides and other bioactive sphingolipid backbones in health and disease: lipidomic analysis, metabolism and roles in membrane structure, dynamics, signaling and autophagy. Biochim Biophys Acta 2006;1758:1864-1884.
  9. Li Q, Chen J, Yu X, Gao JM. A mini review of nervonic acid: source, production, and biological functions. Food Chem 2019;301:125286.
  10. Yu J, Yuan T, Zhang X, Jin Q, Wei W, Wang X. Quantification of nervonic acid in human milk in the first 30 days of lactation: influence of lactation stages and comparison with infant formulae. Nutrients 2019;11:1892.
  11. Wu R, Zhong S, Ni M, et al. Effects of Malania oleifera Chun oil on the improvement of learning and memory function in mice. Evid Based Complement Alternat Med 2020;2020:8617143.
  12. Kwon S, Ki SM, Park SE, et al. Anti-apoptotic effects of human Wharton's jelly-derived mesenchymal stem cells on skeletal muscle cells mediated via secretion of XCL1. Mol Ther 2016;24:1550-1560.
  13. Palumbo P, Lombardi F, Siragusa G, Cifone MG, Cinque B, Giuliani M. Methods of isolation, characterization and expansion of human adipose-derived stem cells (ASCs): an overview. Int J Mol Sci 2018;19:1897.
  14. Choi YS, Park YB, Ha CW, et al. Different characteristics of mesenchymal stem cells isolated from different layers of full term placenta. PLoS One 2017;12:e0172642.
  15. Kim JY, Kim DH, Kim DS, et al. Galectin-3 secreted by human umbilical cord blood-derived mesenchymal stem cells reduces amyloid-beta42 neurotoxicity in vitro. FEBS Lett 2010;584:3601-3608.
  16. Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315-317.
  17. Byun HO, Lee YK, Kim JM, Yoon G. From cell senescence to age-related diseases: differential mechanisms of action of senescence-associated secretory phenotypes. BMB Rep 2015;48:549-558. Erratum in: BMB Rep 2016;49:641-650.
  18. Kamal MM, Kassem DH. Therapeutic potential of Wharton's jelly mesenchymal stem cells for diabetes: achievements and challenges. Front Cell Dev Biol 2020;8:16.
  19. Neri S, Borzi RM. Molecular mechanisms contributing to mesenchymal stromal cell aging. Biomolecules 2020;10:340.
  20. Li J, Han S, Cousin W, Conboy IM. Age-specific functional epigenetic changes in p21 and p16 in injury-activated satellite cells. Stem Cells 2015;33:951-961.
  21. Stein GH, Drullinger LF, Soulard A, Dulic V. Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. Mol Cell Biol 1999;19:2109-2117.
  22. Liao Z, Yeo HL, Wong SW, Zhao Y. Cellular senescence: mechanisms and therapeutic potential. Biomedicines 2021;9:1769.
  23. Kumari R, Jat P. Mechanisms of cellular senescence: cell cycle arrest and senescence associated secretory phenotype. Front Cell Dev Biol 2021;9:645593.
  24. Chinnadurai R, Rajan D, Ng S, et al. Immune dysfunctionality of replicative senescent mesenchymal stromal cells is corrected by IFNγ priming. Blood Adv 2017;1:628-643.
  25. Nam K, Oh S, Lee KM, Yoo SA, Shin I. CD44 regulates cell proliferation, migration, and invasion via modulation of c-Src transcription in human breast cancer cells. Cell Signal 2015;27:1882-1894.
  26. Ludwig N, Szczepanski MJ, Gluszko A, et al. CD44(+) tumor cells promote early angiogenesis in head and neck squamous cell carcinoma. Cancer Lett 2019;467:85-95.
  27. Petruk N, Tuominen S, Akerfelt M, et al. CD73 facilitates EMT progression and promotes lung metastases in triple-negative breast cancer. Sci Rep 2021;11:6035.
  28. Dijk W, Kersten S. Regulation of lipoprotein lipase by Angptl4. Trends Endocrinol Metab 2014;25:146-155.
  29. Adhikary T, Brandt DT, Kaddatz K, et al. Inverse PPARβ/δ agonists suppress oncogenic signaling to the ANGPTL4 gene and inhibit cancer cell invasion. Oncogene 2013;32:5241-5252.
  30. Li X, Chen T, Shi Q, et al. Angiopoietin-like 4 enhances metastasis and inhibits apoptosis via inducing bone morphogenetic protein 7 in colorectal cancer cells. Biochem Biophys Res Commun 2015;467:128-134.
  31. Conte M, Franceschi C, Sandri M, Salvioli S. Perilipin 2 and age-related metabolic diseases: a new perspective. Trends Endocrinol Metab 2016;27:893-903.
  32. Attie AD, Kastelein JP, Hayden MR. Pivotal role of ABCA1 in reverse cholesterol transport influencing HDL levels and susceptibility to atherosclerosis. J Lipid Res 2001;42:1717-1726.
  33. Tao H, Han Z, Han ZC, Li Z. Proangiogenic features of mesenchymal stem cells and their therapeutic applications. Stem Cells Int 2016;2016:1314709.
  34. Muppala S, Xiao R, Krukovets I, et al. Thrombospondin-4 mediates TGF-β-induced angiogenesis. Oncogene 2017;36:5189-5198.
  35. Zhang Q, Zhou M, Wu X, et al. Promoting therapeutic angiogenesis of focal cerebral ischemia using thrombospondin-4 (TSP4) gene-modified bone marrow stromal cells (BMSCs) in a rat model. J Transl Med 2019;17:111.
  36. Isenberg JS, Roberts DD. Thrombospondin-1 in maladaptive aging responses: a concept whose time has come. Am J Physiol Cell Physiol 2020;319:C45-C63.
  37. Liu Y, Chen Q. Senescent mesenchymal stem cells: disease mechanism and treatment strategy. Curr Mol Biol Rep 2020;6:173-182.
  38. Yuan SN, Wang MX, Han JL, et al. Improved colonic inflammation by nervonic acid via inhibition of NF-κB signaling pathway of DSS-induced colitis mice. Phytomedicine 2023;112:154702.
  39. Ito TK, Yokoyama M, Yoshida Y, et al. A crucial role for CDC42 in senescence-associated inflammation and atherosclerosis. PLoS One 2014;9:e102186.
  40. Lee Y, Clinton J, Yao C, Chang SH. Interleukin-17D promotes pathogenicity during infection by suppressing CD8 T cell activity. Front Immunol 2019;10:1172.