DOI QR코드

DOI QR Code

Protective effects of Korean Red Ginseng against sub-acute immobilization stress-induced testicular damage in experimental rats

  • Lee, Sang-Ho (Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University) ;
  • Choi, Kyung-Hwa (Department of Urology, CHA Bundang Medical Center, CHA University) ;
  • Cha, Kyu-Min (Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University) ;
  • Hwang, Seock-Yeon (Department of Biomedical Laboratory Science, College of Health and Medical Science, Daejeon University) ;
  • Park, Un-Kyu (Department of Biomedical Laboratory Science, College of Health and Medical Science, Daejeon University) ;
  • Jeong, Min-Sik (Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University) ;
  • Hong, Jae-Yup (Department of Urology, CHA Bundang Medical Center, CHA University) ;
  • Han, Chang-Kyun (Botanical Drug Laboratory, Korea Ginseng Research Institute, Korea Ginseng Corporation) ;
  • In, Gyo (Botanical Drug Laboratory, Korea Ginseng Research Institute, Korea Ginseng Corporation) ;
  • Kopalli, Spandana Rajendra (Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University) ;
  • Kim, Si-Kwan (Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University)
  • Received : 2017.07.21
  • Accepted : 2017.09.28
  • Published : 2019.01.15

Abstract

Background: Excessive stress causes varied physiological and psychological disorders including male reproductive problems. Here, we attempted to investigate the protective effects of Korean Red Ginseng (Panax ginseng Meyer; KRG) against sub-acute immobilization stress-induced testicular damage in experimental rats. Methods: Male rats (age, 4 wk; weight, 60-70 g) were divided into four groups (n = 8 in each group): normal control group, immobilization control group, immobilization group treated with 100 mg/kg of KRG daily, and immobilization group treated with 200 mg/kg of KRG daily. Normal control and immobilization control groups received vehicle only. KRG (100 mg/kg and 200 mg/kg) was mixed in the standard diet powder and fed daily for 6 mo. Parameters such as organ weight, blood chemistry, sperm kinematic values, and expression levels of testicular-related molecules were measured using commercially available kits, Western blotting, and reverse transcription polymerase chain reaction. Results: Data revealed that KRG restored the altered testis and epididymis weight in immobilization stress-induced rats significantly (p < 0.05). Further, KRG ameliorated the altered blood chemistry and sperm kinematic values when compared with the immobilization control group and attenuated the altered expression levels of spermatogenesis-related proteins (nectin-2, cAMP responsive element binding protein 1, and inhibin-${\alpha}$), sex hormone receptors (androgen receptor, luteinizing hormone receptor, and follicle-stimulating hormone receptor), and antioxidant-related enzymes (glutathione S-transferase m5, peroxiredoxin-4, and glutathione peroxidase 4) significantly in the testes of immobilization stress-induced rats. Conclusion: KRG protected immobilization stress-induced testicular damage and fertility factors in rats, thereby indicating its potential in the treatment of stress-related male sterility.

Keywords

References

  1. Cocuzza M, Sikka SC, Athayde KS, Agarwal A. Clinical relevance of oxidative stress and sperm chromatin damage in male infertility: an evidence based analysis. International Braz J Urol 2007;33:603-21. https://doi.org/10.1590/S1677-55382007000500002
  2. Brotman DJ, Golden SH, Wittstein IS. The cardiovascular toll of stress. Lancet 2007;370:1089-100. https://doi.org/10.1016/S0140-6736(07)61305-1
  3. Rosmond R, Dallman MF, Bjorntorp P. Stress-related cortisol secretion in men: relationships with abdominal obesity and endocrine, metabolic and hemodynamic abnormalities. J Clin Endocrinol Metab 1998;83:1853-9. https://doi.org/10.1210/jcem.83.6.4843
  4. Nargund VH. Effects of psychological stress on male fertility. Nature Reviews Urology 2015;12:373-82. https://doi.org/10.1038/nrurol.2015.112
  5. Kirby ED, Geraghty AC, Ubuka T, Bentley GE, Kaufer D. Stress increases putative gonadotropin inhibitory hormone and decreases luteinizing hormone in male rats. Proc Natl Acad Sci U.S.A 2009;106:11324-9. https://doi.org/10.1073/pnas.0901176106
  6. Sahin E, Gumuslu S. Alterations in brain antioxidant status, protein oxidation and lipid peroxidation in response to different stress models. Behav Brain Res 2004;155:241-8. https://doi.org/10.1016/j.bbr.2004.04.022
  7. Bitgul G, Tekmen I, Keles D, Oktay G. Protective effects of resveratrol against chronic immobilization stress on testis. ISRN Urology 2013;2013, 278720. https://doi.org/10.1155/2013/278720
  8. Samarghandian S, Farkhondeh T, Samini F, Borji A. Protective effects of carvacrol against oxidative stress induced by chronic stress in rat's brain, liver, and kidney. Biochem Res Int 2016;2016, 2645237. https://doi.org/10.1155/2016/2645237
  9. Vogler BK, Pittler MH, Ernst E. The efficacy of ginseng. A systematic review of randomised clinical trials. Eur J Clin Pharmacol 1999;55:567-75. https://doi.org/10.1007/s002280050674
  10. Zhao Z, Kim YW, Wu Y, Zhang J, Lee JH, Li X, Cho IJ, Park SM, Jung DH, Yang CH, et al. Korean Red Ginseng attenuates anxiety-like behavior during ethanol withdrawal in rats. J Ginseng Res 2014;38:256-63. https://doi.org/10.1016/j.jgr.2014.05.010
  11. Mahady GB, Gyllenhaal C, Fong HHS, Farnsworth NR. Ginseng: a review of safety and efficacy. Nutr Clin Care 2000;3:90-101. https://doi.org/10.1046/j.1523-5408.2000.00020.x
  12. Retana-Marquez S, Salazar ED, Velazquez-Moctezuma J. Effect of acute and chronic stress on masculine sexual behavior in the rat. Psychoneuroendocrinology 1996;21:39-50. https://doi.org/10.1016/0306-4530(95)00029-1
  13. Attele AS, Wu JA, Yuan CS. Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol 1999;58:1685-93. https://doi.org/10.1016/S0006-2952(99)00212-9
  14. Kopalli SR, Hwang SY, Won YJ, Kim SW, Cha KM, Han CK, Hong JY, Kim SK. Korean red ginseng extract rejuvenates testicular ineffectiveness and sperm maturation process in aged rats by regulating redox proteins and oxidative defense mechanisms. Exp Gerontol 2015;69:94-102. https://doi.org/10.1016/j.exger.2015.05.004
  15. Ramesh T, Kim SW, Sung JH, Hwang SY, Sohn SH, Yoo SK, Kim SK. Effect of fermented Panax ginseng extract (GINST) on oxidative stress and antioxidant activities in major organs of aged rats. Exp Gerontol 2012;47:77-84. https://doi.org/10.1016/j.exger.2011.10.007
  16. Won YJ, Kim BK, Shin YK, Jung SH, Yoo SK, Hwang SY, Sung JH, Kim SK. Pectinase-treated Panax ginseng extract (GINST) rescues testicular dysfunction in aged rats via redox-modulating proteins. Exp Gerontol 2014;53:57-66. https://doi.org/10.1016/j.exger.2014.02.012
  17. Kopalli SR, Cha KM, Jeong MS, Lee SH, Sung JH, Seo SK, Kim SK. Pectinasetreated Panax ginseng ameliorates hydrogen peroxide-induced oxidative stress in GC-2 sperm cells and modulates testicular gene expression in aged rats. J Ginseng Res 2016;40:185-95. https://doi.org/10.1016/j.jgr.2015.08.005
  18. Hwang SY, Kim WJ, Wee JJ, Choi JS, Kim SK. Panax ginseng improves survival and sperm quality in Guinea pigs exposed to 2,3,7,8-tetrachlorodibenzo- pdioxin. BJU International 2004;94:663-8. https://doi.org/10.1111/j.1464-410X.2004.05019.x
  19. Park HW, In G, Han ST, Lee MW, Kim SY, Kim KT, Cho BG, Han GH, Chang IM. Simultaneous determination of 30 ginsenosides in Panax ginseng preparations using ultra performance liquid chromatography. J Ginseng Res 2013;37:457-67. https://doi.org/10.5142/jgr.2013.37.457
  20. Seibel MM, Taymor ML. Emotional aspects of infertility. Fertility and Sterility 1982;37:137-45. https://doi.org/10.1016/S0015-0282(16)46029-2
  21. Nordkap L, Jensen TK, Hansen TH, Lassen AK, Bang UN, Joensen M, Blomberg Jensen NE, Skakkebæk NE, Jorgensen N. Psychological stress and testicular function: a cross-sectional study of 1,215 Danish men. Fertil Steril 2016;105:174-87. e171. https://doi.org/10.1016/j.fertnstert.2015.09.016
  22. Dong Q, Salva A, Sottas CM, Niu E, Holmes M, Hardy MP. Rapid glucocorticoid mediation of suppressed testosterone biosynthesis in male mice subjected to immobilization stress. J Androl 2004;25:973-81. https://doi.org/10.1002/j.1939-4640.2004.tb03170.x
  23. Yang WM, Park SY, Kim HM, Park EH, Park SK, Chang MS. Effects of Panax ginseng on glial cell-derived neurotrophic factor (GDNF) expression and spermatogenesis in rats. Phytother Res 2011;25:308-11. https://doi.org/10.1002/ptr.3239
  24. Janevic T, Kahn LG, Landsbergis P, Cirillo PM, Cohn BA, Liu X, Factor Litvak P. Effects of work and life stress on semen quality. Fertil Steril 2014;102:530-8. https://doi.org/10.1016/j.fertnstert.2014.04.021
  25. Clermont Y. Kinetics of spermatogenesis in mammals: seminiferous epithelium cycle and spermatogonial renewal. Physiol Rev 1972;52:198-236. https://doi.org/10.1152/physrev.1972.52.1.198
  26. Kuo SC, Teng CM, Lee JC, Ko FN, Chen SC, Wu TS. Antiplatelet components in Panax ginseng. Planta Medica 1990;56:164-7. https://doi.org/10.1055/s-2006-960916
  27. Park Y, Kwon HY, Shimi MK, Rhyu MR, Lee Y. Improved lipid profile in ovariectomized rats by red ginseng extract. Pharmazie 2011;66:450-3.
  28. Song YB, An YR, Kim SJ, Park HW, Jung JW, Kyung JS, Hwang SY, Kim YS. Lipid metabolic effect of Korean red ginseng extract in mice fed on a high-fat diet. J Sci Food Agr 2012;92:388-96. https://doi.org/10.1002/jsfa.4589
  29. Garbarino S, Magnavita N. Work stress and metabolic syndrome in police officers. A prospective study. PloS One 2015;10, e0144318. https://doi.org/10.1371/journal.pone.0144318
  30. Ortolani D, Garcia MC, Melo Thomas L, Spadari Bratfisch RC. Stress-induced endocrine response and anxiety: the effects of comfort food in rats. Stress 2014;17:211-8. https://doi.org/10.3109/10253890.2014.898059
  31. Zhu X, Hu S, Zhu L, Ding J, Zhou Y, Li G. Effects of Lycium barbarum polysaccharides on oxidative stress in hyperlipidemic mice following chronic composite psychological stress intervention. Mol Med Rep 2015;11:3445-50. https://doi.org/10.3892/mmr.2014.3128
  32. Morrison ME, Racaniello VR. Molecular cloning and expression of a murine homolog of the human poliovirus receptor gene. J Virol 1992;66:2807-13. https://doi.org/10.1128/JVI.66.5.2807-2813.1992
  33. Mueller S, Rosenquist TA, Takai Y, Bronson RA, Wimmer E. Loss of nectin-2 at Sertoli-spermatid junctions leads to male infertility and correlates with severe spermatozoan head and midpiece malformation, impaired binding to the zona pellucida, and oocyte penetration. Biol Reprod 2003;69:1330-40. https://doi.org/10.1095/biolreprod.102.014670
  34. Bouchard MJ, Dong Y, McDermott Jr BM, Lam DH, Brown KR, Shelanski M, Bellve AR, Racaniello VR. Defects in nuclear and cytoskeletal morphology and mitochondrial localization in spermatozoa of mice lacking nectin-2, a component of cell-cell adherens junctions. Mol Cell Biol 2000;20:2865-73. https://doi.org/10.1128/MCB.20.8.2865-2873.2000
  35. Hsueh AJW, Dahl KD, Vaughan J, Tucker E, Rivier J, Bardin CW, Vale W. Heterodimers and homodimers of inhibin subunits have different paracrine action in the modulation of luteinizing hormone-stimulated androgen biosynthesis. Proc Natl Acad Sci U.S.A 1987;84:5082-6. https://doi.org/10.1073/pnas.84.14.5082
  36. Cai KL, Hua GH, Ahmad S, Liang AX, Han L, Wu CJ, Yang FF, Yang LG. Action mechanism of inhibin alpha-subunit on the development of Sertoli cells and first wave of spermatogenesis in mice. PloS One 2011;6.
  37. Don J, Stelzer G. The expanding family of CREB/CREM transcription factors that are involved with spermatogenesis. Mol Cell Endocrinol 2002;187:115-24. https://doi.org/10.1016/S0303-7207(01)00696-7
  38. Mou L, Gui Y. A novel variant of androgen receptor is associated with idiopathic azoospermia. Mol Med Rep 2016;14:2915-20. https://doi.org/10.3892/mmr.2016.5587
  39. Sun LP, Song YP, Liu JJ, Liu XR, Guo AZ, Yang LG. Differential expression of luteinizing hormone receptor, androgen receptor and heat-shock protein 70 in the testis of long-distance transported mice. Genet Mol Res 2015;14:9985-93. https://doi.org/10.4238/2015.August.21.4
  40. Pakarainen T, Zhang F, Makela S, Poutanen M, Huhtaniemi I. Testosterone replacement therapy induces spermatogenesis and partially restores fertility in luteinizing hormone receptor knockout mice. Endocrinology 2005;146:596-606. https://doi.org/10.1210/en.2004-0913
  41. Tao YX, Segaloff DL. Follicle stimulating hormone receptor mutations and reproductive disorders. Prog Mol Biol Transl Sci 2009;89:115-31. https://doi.org/10.1016/S1877-1173(09)89005-4
  42. Agarwal A, Virk G, Ong C, du Plessis S. Effect of oxidative stress on male reproduction. World J Mens Health 2014;32:1-17. https://doi.org/10.5534/wjmh.2014.32.1.1
  43. Eskiocak S, Gozen AS, Yapar SB, Tavas F, Kilic AS, Eskiocak M. Glutathione and free sulphydryl content of seminal plasma in healthy medical students during and after exam stress. Hum Reprod 2005;20:2595-600. https://doi.org/10.1093/humrep/dei062
  44. Barbieri ER, Hidalgo ME, Venegas A, Smith R, Lissi EA. Varicocele-associated decrease in antioxidant defenses. J Androl 1999;20:713-7.
  45. Ursini F, Heim S, Kiess M, Maiorino M, Roveri A, Wissing J, Flohe L. Dual function of the selenoprotein PHGPx during sperm maturation. Science 1999;285:1393-6. https://doi.org/10.1126/science.285.5432.1393
  46. Iuchi Y, Okada F, Tsunoda S, Kibe N, Shirasawa N, Ikawa M, Okabe M, Ikeda Y, Fujii J. Peroxiredoxin 4 knockout results in elevated spermatogenic cell death via oxidative stress. Biochem J 2009;419:149-58. https://doi.org/10.1042/BJ20081526
  47. Bae EA, Han MJ, Shin YW, Kim DH. Inhibitory effects of Korean red ginseng and its genuine constituents ginsenosides Rg3, Rf, and Rh2 in mouse passive cutaneous anaphylaxis reaction and contact dermatitis models. Biol Pharm Bull 2006;29:1862-7. https://doi.org/10.1248/bpb.29.1862

Cited by

  1. MOTILIPERM Ameliorates Immobilization Stress-Induced Testicular Dysfunction via Inhibition of Oxidative Stress and Modulation of the Nrf2/HO-1 Pathway in SD Rats vol.21, pp.13, 2020, https://doi.org/10.3390/ijms21134750