DOI QR코드

DOI QR Code

TRPV1 activation induces cell death of TM3 mouse Leydig cells

  • Kim, Eun-Jin (Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University) ;
  • Dang, Long Cao (Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University) ;
  • Nyiramana, Marie Merci (Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University) ;
  • Siregar, Adrian S. (Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University) ;
  • Woo, Min-Seok (Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University) ;
  • Kim, Chang-Woon (Department of Obstetrics and Gynecology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine) ;
  • Kang, Dawon (Department of Physiology, College of Medicine and Institute of Health Sciences, Gyeongsang National University)
  • Received : 2021.09.17
  • Accepted : 2021.09.22
  • Published : 2021.09.30

Abstract

The role of transient receptor potential vanilloid receptor-1 (TRPV1) has been primarily investigated in pain sensory neurons. Relatively, little research has been performed in testicular cells. TRPV1 is abundantly expressed in Leydig cells of young adult mice. This study was conducted to determine the role of the TRPV1 channel in Leydig cells. TRPV1 modulators and testosterone were treated to the mouse Leydig cell line TM3 cells for 24 h. Capsaicin, a TRPV1 activator, dose-dependently induced cell death, whereas capsazepine, a TRPV1 inhibitor, inhibited capsaicin-induced cell death. Testosterone treatment reduced capsaicin-induced cell death. High concentrations of testosterone decreased TRPV1 mRNA and protein expression levels. However, TRPV1 modulators did not affect testosterone production. These results showed that capsaicin induced cell death of Leydig cells and that testosterone reduced capsaicin-induced cell death. Our findings suggest that testosterone may regulate the survival of Leydig cells in young adult mice by decreasing the expression level of TRPV1.

Keywords

Acknowledgement

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education(2021R1I1A3044128) and the Ministry of Science and ICT (2015R1A5A2008833).

References

  1. Al-Agha OM and Axiotis CA. 2007. An in-depth look at Leydig cell tumor of the testis. Arch. Pathol. Lab. Med. 131:311-317. https://doi.org/10.5858/2007-131-311-AILALC
  2. Bai X, Zhang X, Zhou Q. 2018. Effect of testosterone on TRPV1 expression in a model of orofacial myositis pain in the rat. J. Mol. Neurosci. 64:93-101. https://doi.org/10.1007/s12031-017-1009-7
  3. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D. 1997. The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 389:816-824. https://doi.org/10.1038/39807
  4. Chen SC, Chang TJ, Wu FS. 2004. Competitive inhibition of the capsaicin receptor-mediated current by dehydroepiandrosterone in rat dorsal root ganglion neurons. J. Pharmacol. Exp. Ther. 311:529-536. https://doi.org/10.1124/jpet.104.069096
  5. Defo Deeh PB, Watcho P, Wankeu-Nya M, Ngadjui E, Usman UZ. 2019. The methanolic extract of Guibourtia tessmannii (caesalpiniaceae) and selenium modulate cytosolic calcium accumulation, apoptosis and oxidative stress in R2C tumour Leydig cells: involvement of TRPV1 channels. Andrologia 51:e13216.
  6. De Toni L, Garolla A, Menegazzo M, Magagna S, Di Nisio A, Sabovic I, Rocca MS, Scattolini V, Filippi A, Foresta C. 2016. Heat sensing receptor TRPV1 is a mediator of thermotaxis in human spermatozoa. PLoS One 11:e0167622. https://doi.org/10.1371/journal.pone.0167622
  7. Jackson SJ, Andrews N, Ball D, Bellantuono I, Gray J, Hachoumi L, Holmes A, Latcham J, Petrie A, Potter P, Rice A, Ritchie A, Stewart M, Strepka C, Yeoman M, Chapman K. 2017. Does age matter? The impact of rodent age on study outcomes. Lab. Anim. 51:160-169. https://doi.org/10.1177/0023677216653984
  8. Martins D, Tavares I, Morgado C. 2014. "Hotheaded": the role OF TRPV1 in brain functions. Neuropharmacology 85:151-157. https://doi.org/10.1016/j.neuropharm.2014.05.034
  9. Mizrak SC, Gadella BM, Erdost H, Ozer A, van Pelt AM, van Dissel-Emiliani FM. 2008. Spermatogonial stem cell sensitivity to capsaicin: an in vitro study. Reprod. Biol. Endocrinol. 6:52. https://doi.org/10.1186/1477-7827-6-52
  10. Mizrak SC and van Dissel-Emiliani FM. 2008. Transient receptor potential vanilloid receptor-1 confers heat resistance to male germ cells. Fertil. Steril. 90:1290-1293. https://doi.org/10.1016/j.fertnstert.2007.10.081
  11. Mendez-Resendiz KA, Enciso-Pablo O, Gonzalez-Ramirez R, Juarez-Contreras R, Rosenbaum T, Morales-Lazaro SL. 2020. Steroids and TRP channels: a close relationship. Int. J. Mol. Sci. 21:3819. https://doi.org/10.3390/ijms21113819
  12. Nyiramana MM, Cho SB, Kim EJ, Kim MJ, Ryu JH, Nam HJ, Kim NG, Park SH, Choi YJ, Kang SS, Jung M, Shin MK, Han J, Jang IS, Kang D. 2020. Sea hare hydrolysate-induced reduction of human non-small cell lung cancer cell growth through regulation of macrophage polarization and non-apoptotic regulated cell death pathways. Cancers (Basel) 12:726. https://doi.org/10.3390/cancers12030726
  13. Pilutin A, Misiakiewicz-Has K, Kolasa A, Baranowska-Bosiacka I, Marchlewicz M, Wiszniewska B. 2014. The immunoexpression of androgen receptor, estrogen receptors alpha and beta, vanilloid type 1 receptor and cytochrome p450 aromatase in rats testis chronically treated with letrozole, an aromatase inhibitor. Folia Histochem Cytobiol 52:206-217. https://doi.org/10.5603/FHC.2014.0024
  14. Pronsato L, Boland R, Milanesi L. 2012. Testosterone exerts antiapoptotic effects against H2O2 in C2C12 skeletal muscle cells through the apoptotic intrinsic pathway. J. Endocrinol. 212:371-381. https://doi.org/10.1530/JOE-11-0234
  15. Ramirez-Barrantes R, Carvajal-Zamorano K, Rodriguez B, Cordova C, Lozano C, Simon F, Diaz P, Munoz P, Marchant I, Latorre R, Castillo K, Olivero P. 2020. TRPV1-estradiol stereospecific relationship underlies cell survival in oxidative cell death. Front. Physiol. 11:444. https://doi.org/10.3389/fphys.2020.00444
  16. Riera CE, Huising MO, Follett P, Leblanc M, Halloran J, Van Andel R, de Magalhaes Filho CD, Merkwirth C, Dillin A. 2014. TRPV1 pain receptors regulate longevity and metabolism by neuropeptide signaling. Cell 157:1023-1036. https://doi.org/10.1016/j.cell.2014.03.051
  17. Santiago J, Silva JV, Fardilha M. 2019. First insights on the presence of the unfolded protein response in human spermatozoa. Int. J. Mol. Sci. 20:5518. https://doi.org/10.3390/ijms20215518
  18. Siregar AS, Nyiramana MM, Kim EJ, Shin EJ, Kim CW, Lee DK, Hong SG, Han J, Kang D. 2019. TRPV1 is associated with testicular apoptosis in mice. J. Anim. Reprod. Biotechnol. 34:311-317. https://doi.org/10.12750/JARB.34.4.311
  19. Smith LB and Walker WH. 2014. The regulation of spermatogenesis by androgens. Semin. Cell Dev. Biol. 30:2-13. https://doi.org/10.1016/j.semcdb.2014.02.012
  20. Szallasi A, Cortright DN, Blum CA, Eid SR. 2007. The vanilloid receptor TRPV1: 10 years from channel cloning to antagonist proof-of-concept. Nat. Rev. Drug Discov. 6:357-372. https://doi.org/10.1038/nrd2280
  21. Williams PA, Kobilnyk HE, McMillan EA, Strochlic TI. 2019. MAPKAP kinase 2-mediated phosphorylation of HspA1L protects male germ cells from heat stress-induced apoptosis. Cell Stress Chaperones 24:1127-1136. https://doi.org/10.1007/s12192-019-01035-6
  22. Yang JH, Siregar AS, Kim EJ, Nyiramana MM, Shin EJ, Han J, Sohn JT, Kim JW, Kang D. 2019. Involvement of TREK-1 channel in cell viability of H9c2 rat cardiomyoblasts affected by bupivacaine and lipid emulsion. Cells 8:454. https://doi.org/10.3390/cells8050454