Natural Biomolecules in Sperm Production: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Stefan S Du Plessis.

Poor semen quality and abnormal sperm structure or function by and large constitute a predominant presentation of male infertility and are among the major reasons why couples seek the services of assisted reproduction. Natural biomolecules, such as polyphenols or flavonoids have garnered substantial attention from the scientific community as well as public media. In the meantime, it has been suggested that administration of natural biomolecules has been shown to have a positive impact male infertility. A number of in vivo as well as in vitro studies have reported that active components isolated from natural resources could enhance blood circulation in the male reproductive system and support the synthesis and secretion of androgens. Animal and human reports also suggest that natural biomolecules may play important roles in the enhancement of semen quality, including sperm concentration, motility, vitality, and structural integrity. 

  • natural biomolecules
  • polyphenols
  • flavonoids
  • spermatozoa
  • male fertility
  • spermatogenesis
Please wait, diff process is still running!

References

  1. Risuleo, G.; La Mesa, C. Resveratrol: Biological Activities and Potential Use in Health and Disease. In Nutraceuticals in Veterinary Medicine, 1st ed.; Gupta, R., Srivastava, A., Lall, R., Eds.; Springer: Cham, Germany, 2019; pp. 215–226.
  2. Gambini, J.; Inglés, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol: In Vitro and In Vivo Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxid. Med. Cell. Longev. 2015, 2015, 837042.
  3. Mongioì, L.M.; Perelli, S.; Condorelli, R.A.; Barbagallo, F.; Crafa, A.; Cannarella, R.; La Vignera, S.; Calogero, A.E. The Role of Resveratrol in Human Male Fertility. Molecules 2021, 26, 2495.
  4. Mohar, D.S.; Malik, S. The Sirtuin System: The Holy Grail of Resveratrol? J. Clin. Exp. Cardiolog. 2012, 3, 216.
  5. Pasquariello, R.; Verdile, N.; Brevini, T.A.L.; Gandolfi, F.; Boiti, C.; Zerani, M.; Maranesi, M. The Role of Resveratrol in Mammalian Reproduction. Molecules 2020, 25, 4554.
  6. Illiano, E.; Trama, F.; Zucchi, A.; Iannitti, R.G.; Fioretti, B.; Costantini, E. Resveratrol-Based Multivitamin Supplement Increases Sperm Concentration and Motility in Idiopathic Male Infertility: A Pilot Clinical Study. J. Clin. Med. 2020, 9, 4017.
  7. Juan, M.E.; González-Pons, E.; Munuera, T.; Ballester, J.; Rodríguez-Gil, J.E.; Planas, J.M. trans-Resveratrol, a natural antioxidant from grapes, increases sperm output in healthy rats. J. Nutr. 2005, 135, 757–135760.
  8. Ourique, G.M.; Finamor, I.A.; Saccol, E.M.; Riffel, A.P.; Pês, T.S.; Gutierrez, K.; Gonçalves, P.B.; Baldisserotto, B.; Pavanato, M.A.; Barreto, K.P. Resveratrol improves sperm motility, prevents lipid peroxidation and enhances antioxidant defences in the testes of hyperthyroid rats. Reprod. Toxicol. 2013, 37, 31–39.
  9. Abdelali, A.; Al-bader, M.; Kilarkaje, N. Effects of Trans -Resveratrol on hyperglycemiainduced abnormal spermatogenesis, DNA damage and alterations in poly (ADP-ribose) polymerase signaling in rat testis. Toxicol. Appl. Pharmacol. 2016, 311, 61–73.
  10. Bahmanzadeh, M.; Goodarzi, M.T.; Rezaei Farimani, A.; Fathi, N.; Alizadeh, Z. Resveratrol supplementation improves DNA integrity and sperm parameters in streptozotocin-nicotinamide-induced type 2 diabetic rats. Andrologia 2019, 51, e13313.
  11. Guo, Y.; Wang, A.; Liu, X.; Li, E. Effects of resveratrol on reducing spermatogenic dysfunction caused by high-intensity exercise. Reprod. Biol. Endocrinol. 2019, 17, 42.
  12. Shati, A.A. Resveratrol improves sperm parameter and testicular apoptosis in cisplatin-treated rats: Effects on ERK1/2, JNK, and Akt pathways. Syst. Biol. Reprod. Med. 2019, 65, 236–249.
  13. Ajayi, A.F.; Akhigbe, R.E. Codeine-induced sperm DNA damage is mediated predominantly by oxidative stress rather than apoptosis. Redox Rep. 2020, 25, 33–40.
  14. Jalili, C.; Salashoor, M.R.; Jalili, F.; Kakabarei, S.; Akrami, A.; Sohrabi, M.; Aookhsah, M.; Ghanbari, A. Therapeutic Effect of Resveratrol on Morphine-Induced Damage in Male Reproductive System of Mice by Reducing Nitric Oxide Serum Level. Int. J. Morphol. 2017, 35, 1342–1347.
  15. Jalili, C.; Roshankhah, S.; Salahshoor, M.R.; Mohammadi, M.M. Resveratrol Attenuates Malathion Induced Damage in Some Reproductive Parameters by Decreasing Oxidative Stress and Lipid Peroxidation in Male Rats. J. Family Reprod. Health 2019, 13, 70–79.
  16. Jhanji, M.; Rao, C.N.; Sajish, M. Towards resolving the enigma of the dichotomy of resveratrol: Cis- and trans-resveratrol have opposite effects on TyrRS-regulated PARP1 activation. GeroScience 2021, 43, 1171–1200.
  17. Collodel, G.; Federico, M.; Geminiani, M.; Martini, S.; Bonechi, C.; Rossi, C.; Figura, N.; Moretti, E. Effect of trans-resveratrol on induced oxidative stress in human sperm and in rat germinal cells. Reprod. Toxicol. 2011, 31, 239–246.
  18. Tvrdá, E.; Lukáč, N.; Lukáčová, J.; Hashim, F.; Massányi, P. In vitro supplementation of resveratrol to bovine spermatozoa: Effects on motility, viability and superoxide production. J. Microbiol. Biotechnol. Food Sci. 2015, 4, 336–341.
  19. Shabani Nashtaei, M.; Amidi, F.; Gilani, S.; Aleyasin, A.; Bakhshalizadeh, S.; Naji, M.; Nekoonam, S. Protective features of resveratrol on human spermatozoa cryopreservation may be mediated through 50 AMP-activated protein kinase activation. Andrology 2016, 5, 313–326.
  20. Cui, X.; Jing, X.; Wu, X.; Yan, M. Protective effect of resveratrol on spermatozoa function in male infertility induced by excess weight and obesity. Mol. Med. Rep. 2016, 14, 4659–4665.
  21. Pervaiz, S.; Holme, A.L. Resveratrol: Its biologic targets and functional activity. Antioxid. Redox. Signal. 2009, 11, 2851–2897.
  22. Alamo, A.; Condorelli, R.A.; Mongioì, L.M.; Cannarella, R.; Giacone, F.; Calabrese, V.; La Vignera, S.; Calogero, A.E. En-vironment and Male Fertility: Effects of Benzo-a-Pyrene and Resveratrol on Human Sperm Function In Vitro. J. Clin. Med. 2019, 8, 561.
  23. Tvrdá, E.; Kováčik, A.; Tušimová, E.; Massányi, P.; Lukáč, N. Resveratrol offers protection to oxidative stress induced by ferrous ascorbate in bovine spermatozoa. J. Environ. Sci. Health A Tox. Hazard. Subst. Environ. Eng. 2015, 50, 1440–1451.
  24. Li, C.Y.; Zhao, Y.H.; Hao, H.S.; Wang, H.Y.; Huang, J.M.; Yan, C.L.; Du, W.H.; Pang, Y.W.; Zhang, P.P.; Liu, Y.; et al. Resveratrol significantly improves the fertilisation capacity of bovine sex-sorted semen by inhibiting apoptosis and lipid peroxidation. Sci. Rep. 2018, 8, 7603.
  25. Mohammadzadeh, M.; Khalili, M.A.; Ramezani, V.; Hamishehkar, H.; Marvast, L.D.; Mangoli, E.; Rajabi, M.; Sales, Z.A.; Talebi, A.R. Does resveratrol affect prepared sperm parameters and chromatin quality in normozoospermic and asthenozoospermic patients before and after freezing? A lab trial study. Int. J. Reprod. Biomed. 2020, 18, 755–764.
  26. Assunção, C.M.; Mendes, V.R.A.; Brandão, F.Z.; Batista, R.I.T.P.; Souza, E.D.; Carvalho, B.C.; Quintão, C.C.R.; Raposo, N.R.B.; Camargo, L.S.A. Effects of resveratrol in bull semen extender on post-thaw sperm quality and capacity for fertilization and embryo development. Anim. Reprod. Sci. 2021, 226, 106697.
  27. Lone, F.A.; Naikoo, M.; Shah, S.M.; Darzi, S.A.; Farooq, J. Effect of Idebenone, Resveratrol and Taurine on the Sperm Quality and Lipid Peroxidation of Cryopreserved Crossbred Ram Semen. Cryo Lett. 2021, 42, 146–153.
  28. Sun, L.; Fan, X.; Zeng, Y.; Wang, L.; Zhu, Z.; Li, R.; Tian, X.; Wang, Y.; Lin, Y.; Wu, D.; et al. Resveratrol protects boar sperm in vitro via its antioxidant capacity. Zygote 2020, 28, 417–424.
  29. Branco, C.S.; Garcez, M.E.; Pasqualotto, F.F.; Erdtman, B.; Salvador, M. Resveratrol and ascorbic acid prevent DNA damage induced by cryopreservation in human semen. Cryobiology 2010, 60, 235–237.
  30. Garcez, M.E.; Branco, C.D.S.; Lara, L.V.; Pasqualotto, F.F.; Salvador, M. Effects of resveratrol supplementation on cryopreservation medium of human semen. Fertil. Steril. 2010, 94, 2118–2121.
  31. Zheng, J.; Ramirez, V.D. Inhibition of mitochondrial proton F0F1-ATPase/ATP synthase by polyphenolic phytochemicals. Br. J. Pharmacol. 2000, 130, 1115–1123.
  32. Emerling, B.M.; Weinberg, F.; Snyder, C.; Burgess, Z.; Mutlu, G.M.; Viollet, B.; Budinger, G.R.S.; Chandel, N.S. Hypoxic activation of AMPK is dependent on mitochondrial ROS but independent of an increase in AMP/ATP ratio. Free Radic. Biol. Med. 2009, 46, 1386–1391.
  33. Shabani Nashtaei, M.; Nekoonam, S.; Naji, M.; Bakhshalizadeh, S.; Amidi, F. Cryoprotective effect of resveratrol on DNA damage and crucial human sperm messenger RNAs, possibly through 5′ AMP-activated protein kinase activation. Cell Tissue Bank. 2018, 19, 87–95.
  34. Kim, J.K.; Park, S.U. Quercetin and its role in biological functions: An updated review. EXCLI J. 2018, 17, 856–863.
  35. Ulusoy, H.G.; Sanlier, N. A minireview of quercetin: From its metabolism to possible mechanisms of its biological activities. Crit. Rev. Food Sci. Nutr. 2020, 60, 3290–3303.
  36. Xu, D.; Hu, M.J.; Wang, Y.Q.; Cui, Y.L. Antioxidant Activities of Quercetin and Its Complexes for Medicinal Application. Molecules 2019, 24, 1123.
  37. Ranawat, P.; Pathak, C.M.; Khanduja, K.L. A new perspective on the quercetin paradox in male reproductive dysfunction. Phytother. Res. 2013, 27, 802–810.
  38. Taepongsorat, L.; Tangpraprutgul, P.; Kitana, N.; Malaivijitnond, S. Stimulating effects of quercetin on sperm quality and reproductive organs in adult male rats. Asian J. Androl. 2008, 10, 249–258.
  39. Yelumalai, S.; Giribabu, N.; Karim, K.; Omar, S.Z.; Salleh, N.B. In vivo administration of quercetin ameliorates sperm oxidative stress, inflammation, preserves sperm morphology and functions in streptozotocin-nicotinamide induced adult male diabetic rats. Arch. Med. Sci. 2019, 15, 240–249.
  40. Naseer, Z.; Ahmad, E.; Aksoy, M.; Epikmen, E.T. Impact of quercetin supplementation on testicular functions in summer heat-stressed rabbits. World Rabbit Sci. 2020, 28, 19–27.
  41. Jahan, S.; Rehman, S.; Ullah, H.; Munawar, A.; Ain, Q.U.; Iqbal, T. Ameliorative effect of quercetin against arsenic-induced sperm DNA damage and daily sperm production in adult male rats. Drug Chem. Toxicol. 2016, 39, 290–296.
  42. Ben Abdallah, F.; Fetoui, H.; Zribi, N.; Fakhfakh, F.; Keskes, L. Quercetin attenuates lambda cyhalothrin-induced reproductive toxicity in male rats. Environ. Toxicol. 2013, 28, 673–680.
  43. Bu, T.L.; Jia, Y.D.; Lin, J.X.; Mi, Y.L.; Zhang, C.Q. Alleviative effect of quercetin on germ cells intoxicated by 3-methyl-4-nitrophenol from diesel exhaust particles. J. Zhejiang Univ. Sci. B 2012, 13, 318–326.
  44. Ranawat, P. Quercetin Exerts Deleterious Effects on the Reproductive Potential of Male Mice. Biophys. J. 2013, 104, 151a–152a.
  45. Aravindakshan, M.; Chauhan, P.S.; Sundaram, K. Studies on germinal effects of quercetin, a naturally occurring flavonoid. Mutat. Res. 1985, 144, 99–106.
  46. Boots, A.W.; Li, H.; Schins, R.P.; Duffin, R.; Heemskerk, J.W.; Bast, A.; Haenen, G.R. The quercetin paradox. Toxicol. Appl. Pharmacol. 2007, 222, 89–96.
  47. Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of cell death: The calcium-apoptosis link. Nat. Rev. Mol. Cell. Biol. 2003, 4, 552–565.
  48. Diao, R.; Gan, H.; Tian, F.; Cai, X.; Zhen, W.; Song, X.; Duan, Y.G. In vitro antioxidation effect of Quercetin on sperm function from the infertile patients with leukocytospermia. Am. J. Reprod. Immunol. 2019, 82, e13155.
  49. Tvrdá, E.; Tušimová, E.; Kováčik, A.; Paál, D.; Libová, L.; Lukáč, N. Protective Effects of Quercetin on Selected Oxidative Biomarkers in Bovine Spermatozoa Subjected to Ferrous Ascorbate. Reprod. Domest. Anim. 2016, 51, 524–537.
  50. Tvrdá, E.; Debacker, M.; Ďuračka, M.; Kováč, J.; Bučko, O. Quercetin and Naringenin Provide Functional and Antioxidant Protection to Stored Boar Semen. Animals 2020, 10, 1930.
  51. Kim, T.H.; Yuh, I.S.; Park, I.C.; Cheong, H.T.; Kim, J.T.; Park, C.K.; Yang, B.K. Effects of Quercetin and Genistein on Boar Sperm Characteristics and Porcine IVF Embyo Developments. J. Emb. Trans. 2014, 29, 141–148.
  52. Winn, E.; Whitaker, B.D. Quercetin supplementation to the thawing and incubation media of boar sperm improves post-thaw sperm characteristics and the in vitro production of pig embryos. Reprod. Biol. 2020, 20, 315–320.
  53. Seifi-Jamadi, A.; Kohram, H.; Shahneh, A.Z.; Ansari, M.; Macías-García, B. Quercetin Ameliorate Motility in Frozen-Thawed Turkmen Stallions Sperm. J. Equine Vet. Sci. 2016, 45, 73–77.
  54. Rakha, B.A.; Qurrat-Ul-Ain Ansari, M.S.; Akhter, S.; Akhter, A.; Awan, M.A.; Santiago-Moreno, J. Effect of Quercetin on Oxidative Stress, Mitochondrial Activity, and Quality of Indian Red Jungle Fowl (Gallus gallus murghi) Sperm. Biopreserv. Biobank. 2020, 18, 311–320.
  55. Ardeshirnia, R.; Zandi, M.; Sanjabi, M.R. The effect of quercetin on fertility of frozen-thawed ram epididymal spermatozoa. S. Afr. J. Anim. Sci. 2017, 47, 237–244.
  56. El-Khawagah, A.R.M.; Kandiel, M.M.M.; Samir, H. Effect of Quercetin Supplementation in Extender on Sperm Kinematics, Extracellular Enzymes Release, and Oxidative Stress of Egyptian Buffalo Bulls Frozen-Thawed Semen. Front. Vet. Sci. 2020, 7, 604460.
  57. Askari Jahromi, M.; Movahedin, M.; Mazaheri, Z.; Amanlu, M.; Mowla, S.J.; Batooli, H. Evaluating the effects of Escanbil (Calligonum) extract on the expression level of Catsper gene variants and sperm motility in aging male mice. Iran. J. Reprod. Med. 2014, 12, 459–466.
  58. Williams, K.M.; Ford, W.C. Effects of Ca-ATPase inhibitors on the intracellular calcium activity and motility of human spermatozoa. Int. J. Androl. 2003, 26, 366–375.
  59. Avdatek, F.; Yeni, D.; Inanç, M.E.; Çil, B.; Tuncer, B.P.; Türkmen, R.; Tasdemir, U. Supplementation of quercetin for advanced DNA integrity in bull semen cryopreservation. Andrologia 2018, 50, e12975.
  60. Bansal, A.K.; Bilaspuri, G.S. Impacts of oxidative stress and antioxidants on semen functions. Vet. Med. Int. 2010, 50, e12975.
  61. Tvrdá, E.; Lukáč, N.; Lukáčová, J.; Jambor, T.; Hashim, F.; Massányi, P. Dose-and time-dependent in vitro effects of quercetin on bovine spermatozoa activity and superoxide production. Folia Vet. 2014, 58, 224–231.
  62. Gibb, Z.; Butler, T.J.; Morris, L.H.A.; Maxwell, W.M.C.; Grupen, C.G. Quercetin improves the postthaw characteristics of cryopreserved sex-sorted and non-sorted stallion sperm. Theriogenology 2013, 79, 1001–1009.
  63. Moretti, E.; Mazzi, L.; Terzuoli, G.; Bonechi, C.; Iacoponi, F.; Martini, S.; Rossi, C.; Collodel, G. Effect of quercetin, rutin, naringenin and epicatechin on lipid peroxidation induced in human sperm. Reprod. Toxicol. 2012, 34, 651–657.
  64. Ahmadi, S.; Bashiri, R.; Ghadiri-Anari, A.; Nadjarzadeh, A. Antioxidant supplements and semen parameters: An evidence based review. Int. J. Reprod. Biomed. 2016, 14, 729–736.
  65. Seifi-Jamadi, A.; Ahmad, E.; Ansari, M.; Kohram, H. Antioxidant effect of quercetin in an extender containing DMA or glycerol on freezing capacity of goat semen. Cryobiology 2017, 75, 15–20.
  66. Silva, L.F.M.C.; Araujo, E.A.B.; Oliveira, S.N.; Dalanezi, F.M.; Andrade, L.R.P.; Hartwig, F.P.; Pinto, B.M.; Guasti, P.; Crespilho, A.M.; Freitas, C.P.; et al. Quercetin reduces oxidative stress after cryopreservation of stallion spermatozoa. J. Equine Vet. Sci. 2016, 43, 77–78.
  67. Khanduja, K.L.; Verma, A.; Bhardwaj, A. Impairment of human sperm motility and viability by quercetin is independent of lipid peroxidation. Andrologia 2001, 33, 277–281.
  68. Silva, E.C.B.; Arruda, L.C.P.; Silva, S.V.; Souza, H.M.; Guerra, M.M.P. High resveratrol or quercetin concentrations reduce the oscillation index of frozen goat semen. Arq. Bras. Med. Vet. Zootec. 2016, 68, 5.
  69. Borges, M.S.; Born, J.L.B.; Conti, L.M.; Segabinazzi, L.G.; Nichi, M.; Kawai, G.K.V.; Leite, R.F.; Peixoto, K.C., Jr.; Dell’Aqua, J.A., Jr.; Papa, F.O.; et al. Paradoxical Effect of Quercetin Antioxidant on Goat Sperm Parameters after Cryopreservation. Cryo Lett. 2020, 41, 128–134.
  70. Liang, X.; Xia, Z.; Yan, J.; Wang, Y.; Xue, S.; Zhang, X. Quercetin inhibits human sperm functions by reducing sperm [Ca2+]i and tyrosine phosphorylation. Pak. J. Pharm. Sci. 2016, 29, 2391–2396.
  71. Li, M.W.; Yudin, A.I.; Van de Voort, C.A.; Sabeur, K.; Primakoff, P.; Overstreet, J.W. Inhibition of monkey sperm hyaluronidase activity and heterologous cumulus penetration by flavonoids. Biol. Reprod. 1997, 56, 1383–1389.
  72. Przybylska, S. Lycopene—A bioactive carotenoid offering multiple health benefits: A review. Int. J. Food Sci. Technol. 2020, 55, 11–32.
  73. Arballo, J.; Amengual, J.; Erdman, J.W., Jr. Lycopene: A Critical Review of Digestion, Absorption, Metabolism, and Excretion. Antioxidants 2021, 10, 342.
  74. Goyal, A.; Delves, G.H.; Chopra, M.; Lwaleed, B.A.; Cooper, A.J. Can lycopene be delivered into semen via prostasomes? In vitro incorporation and retention studies. Int. J. Androl. 2006, 29, 528–533.
  75. Mirahmadi, M.; Azimi-Hashemi, S.; Saburi, E.; Kamali, H.; Pishbin, M.; Hadizadeh, F. Potential inhibitory effect of lycopene on prostate cancer. Biomed. Pharmacother. 2020, 219, 110459.
  76. Durairajanayagam, D.; Agarwal, A.; Ong, C.; Prashast, P. Lycopene and male infertility. Asian J. Androl. 2014, 16, 420–425.
  77. Palan, P.; Naz, R. Changes in various antioxidant levels in human seminal plasma related to immunoinfertility. Arch. Androl. 1996, 36, 139–143.
  78. Goyal, A.; Chopra, M.; Lwaleed, B.A.; Birch, B.; Cooper, A.J. The effects of dietary lycopene supplementation on human seminal plasma. BJU Int. 2007, 99, 1456–1460.
  79. Williams, E.A.; Parker, M.; Robinson, A.; Pitt, S.; Pacey, A.A. A randomized placebo-controlled trial to investigate the effect of lactolycopene on semen quality in healthy males. Eur. J. Nutr. 2020, 59, 825–833.
  80. Yamamoto, Y.; Aizawa, K.; Mieno, M.; Karamatsu, M.; Hirano, Y.; Furui, K.; Miyashita, T.; Yamazaki, K.; Inakuma, T.; Sato, I.; et al. The effects of tomato juice on male infertility. Asia Pac. J. Clin. Nutr. 2017, 26, 65–71.
  81. Nouri, M.; Amani, R.; Nasr-Esfahani, M.; Tarrahi, M.J. The effects of lycopene supplement on the spermatogram and seminal oxidative stress in infertile men: A randomized, double-blind, placebo-controlled clinical trial. Phytother. Res. 2019, 33, 3203–3211.
  82. Mohanty, N.K.; Kumar, S.; Jha, A.K.; Arora, R.P. Management of idiopathic oligoasthenospermia with lycopene. Indian J. Urol. 2001, 18, 57.
  83. Gupta, N.P.; Kumar, R. Lycopene therapy in idiopathic male infertility-a preliminary report. Int. Urol. Nephrol. 2002, 34, 369–372.
  84. Filipcikova, R.; Oborna, I.; Brezinova, J.; Novotny, J.; Wojewodka, G.; De Sanctis, J.B. Lycopene improves the distorted ratio between AA/DHA in the seminal plasma of infertile males and increases the likelihood of successful pregnancy. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 2015, 159, 77–82.
  85. Taş, M.; Saruhan, B.G.; Kurt, D.; Yokuş, B.; Denli, M. Protective role of lycopene on aflatoxin B1 induced changes in sperm characteristics and testicular damages in rats. Kafkas Univ. Vet. Fak. Derg. 2010, 16, 597–604.
  86. Jacob, K.; Periago, M.J.; Bohm, V.; Berruezo, G.R. Influence of lycopene and vitamin C from tomato juice on biomarkers of oxidative stress and inflammation. British J. Nutr. 2008, 99, 137–146.
  87. Pirayesh Islamian, J.; Mehrali, H. Lycopene as a carotenoid provides radioprotectant and antioxidant effects by quenching radiation-induced free radical singlet oxygen: An overview. Cell J. 2015, 16, 386–391.
  88. Tvrdá, E.; Kováčik, A.; Tušimová, E.; Paál, D.; Mackovich, A.; Alimov, J.; Lukáč, N. Antioxidant efficiency of lycopene on oxidative stress—induced damage in bovine spermatozoa. J. Anim. Sci. Biotechnol. 2016, 7, 50.
  89. Ly, C.; Yockell-Lelièvre, J.; Ferraro, Z.M.; Arnason, J.T.; Ferrier, J.; Gruslin, A. The effects of dietary polyphenols on reproductive health and early development. Hum. Reprod. Update 2015, 21, 228–248.
  90. Oborna, I.; Malickova, K.; Fingerova, H.; Brezinova, J.; Horka, P.; Novotny, J.; Bryndova, H.; Filipcikova, R.; Svobodova, M. A randomized controlled trial of lycopene treatment on soluble receptor for advanced glycation end products in seminal and blood plasma of normospermic men. Am. J. Reprod. Immunol. 2011, 66, 179–184.
  91. Mangiagalli, M.G.; Cesari, V.; Cerolini, S.; Luzi, F.; Toschi, I. Effect of lycopene supplementation on semen quality and reproductive performance in rabbit. World Rabbit Sci. 2012, 20, 141–148.
  92. Dobrzyńska, M.G.M.; Gajowik, A. Amelioration of sperm count and sperm quality by lycopene supplementation in irradiated mice. Reprod. Fertil. Dev. 2020, 32, 1040–1047.
  93. Türk, G.; Ateşşahin, A.; Sönmez, M.; Yüce, A.; Ceribaşi, A.O. Lycopene protects against cyclosporine A-induced testicular toxicity in rats. Theriogenology 2007, 67, 778–785.
  94. Ateşşahin, A.; Karahan, I.; Türk, G.; Gürb, S.; Yilmaz, S.; Çeribaşi, A.O. Protective role of lycopene on cisplatin-induced changes in sperm. Reprod. Toxicol. 2006, 21, 42–47.
  95. Aly, H.A.; El-Beshbishy, H.A.; Banjar, Z.M. Mitochondrial dysfunction induced impairment of spermatogenesis in LPS-treated rats: Modulatory role of lycopene. Eur. J. Pharmacol. 2012, 677, 31–38.
  96. Tamiselvan, P.; Bharathiraja, K.; Vijayaprakash, S.; Balasubramanian, M.P. Protective role of lycopene on bisphenol A induced changes in sperm characteristics, testicular damage and oxidative stress. Int. J. Pharm. Bio. Sci. 2013, 4, 131–143.
  97. Salem, E.A.; Salem, N.A.; Maarouf, A.M.; Serefoglu, E.C.; Hellstrom, W.J. Selenium and lycopene attenuate cisplatin-induced testicular toxicity associated with oxidative stress in Wistar rats. Urology 2012, 79, 1184.e1–1184.e6.
  98. Mangiagalli, M.G.; Marelli, S.P.; Cavalchini, L.G. Effect of lycopene on fowl sperm characteristics during in vitro storage. Arch. Geflügelkd. 2007, 71, 25–29.
  99. Uysal, O.; Bucak, M.N. Effect of oxidized glutathione, bovine serum albumin, cysteine and lycopene on the quality of frozen thawed ram semen. Acta Vet. Brno 2007, 76, 383–390.
  100. Bucak, M.N.; Ataman, M.B.; Başpınar, N.; Uysal, O.; Taşpınar, M.; Bilgili, A.; Öztürk, C.; Güngör, Ş.; İnanç, M.E.; Akal, E. Lycopene and resveratrol improve post-thaw bull sperm parameters: Sperm motility, mitochondrial activity and DNA integrity. Andrologia 2015, 47, 545–552.
  101. Tvrda, E.; Mackovich, A.; Greifova, H.; Hashim, F.; Lukac, N. Antioxidant effects of lycopene on bovine sperm survival and oxidative profile following cryopreservation. Vet. Med. 2017, 62, 429–436.
  102. Zini, A.; San Gabriel, M.; Libman, J. Lycopene supplementation in vitro can protect human sperm deoxyribonucleic acid from oxidative damage. Fertil. Steril. 2010, 94, 1033–1036.
  103. Tvrdá, E.; Gosálvez, J.; Arroyo, F.; Sánchez, P.; Risco Delgado, R.J.; Sánchez, R. Dynamic assessment of human sperm DNA damage III: The effect of sperm freezing techniques. Cell Tissue Bank. 2020. online ahead of print.
  104. Rosato, M.P.; Centoducati, G.; Santacroce, M.P.; Iaffaldano, N. Effects of lycopene on in vitro quality and lipid peroxidation in refrigerated and cryopreserved turkey spermatozoa. Br. Poult. Sci. 2012, 53, 545–552.
  105. Zribi, N.; Chakroun, N.F.; Elleuch, H.; Abdallah, F.B.; Ben Hamida, A.S.; Gargouri, J.; Fakhfakh, F.; Keskes, L.A. Sperm DNA fragmentation and oxidation are independent of malondialdheyde. Reprod. Biol. Endocrinol. 2011, 9, 47.
  106. Bucak, M.N.; Tuncer, P.B.; Sariozkan, S.; Baspinar, N.; Taspinar, M.; Coyan, K.; Bilgili, A.; Akalin, P.P.; Buyukleblebici, S.; Aydos, S.; et al. Effects of antioxidants on post-thawed bovine sperm and oxidative stress parameters: Antioxidants protect DNA integrity against cryodamage. Cryobiology 2010, 61, 248–253.
  107. Coyan, K.; Baspinar, N.; Bucak, M.N.; Akalin, P.P. Effects of cysteine and ergothioneine on post-thawed Merino ram sperm and biochemical parameters. Cryobiology 2011, 63, 1–6.
  108. Paiva, S.A.; Russell, R.M. Beta-carotene and other carotenoids as antioxidants. J. Am. Coll. Nutr. 1999, 18, 426–433.
  109. Dos Santos, A.N.; de Nascimento, T.R.L.; Gondim, B.L.C.; Velo, M.M.A.C.; de Rêgo, R.I.A.; do Neto, J.R.C.; Machado, J.R.; da Silva, M.V.; de Araújo, H.W.C.; Fonseca, M.G.; et al. Catechins as Model Bioactive Compounds for Biomedical Applications. Curr. Pharm. Des. 2020, 26, 4032–4047.
  110. Roychoudhury, S.; Agarwal, A.; Virk, G.; Cho, C.L. Potential role of green tea catechins in the management of oxidative stress-associated infertility. Reprod. Biomed. Online 2017, 34, 487–498.
  111. Isemura, M. Catechin in Human Health and Disease. Molecules 2019, 24, 528.
  112. Galleano, M.; Verstraeten, S.V.; Oteiza, P.I.; Fraga, C.G. Antioxidant actions of flavonoids: Thermodynamic and kinetic analysis. Arch. Biochem. Biophys. 2010, 501, 23–30.
  113. Pirker, K.F.; Baratto, M.C.; Basosi, R.; Goodman, B.A. Influence of pH on the speciation of copper (II) in reactions with the green tea polyphenols, epigallocatechin gallate and gallic acid. J. Inorg. Biochem. 2012, 112, 10–16.
  114. Rai, A.; Gill, M.; Kinra, M.; Dsouza, L.A.; Sumalatha, S.; Raj, S.; Shetty, R.; Nandakumar, K.; Chamallamudi, M.R.; Kumar, N. Assessment of preclinical effect of (+)-catechin hydrate on sexual function: An in silico and in vivo study. Andrologia 2020, 52, e13737.
  115. Ding, J.; Wang, H.; Wu, Z.-B.; Zhao, J.; Zhang, S.; Li, W. Protection of murine spermatogenesis against ionizing radiation-induced testicular injury by a green tea polyphenol. Biol. Reprod. 2015, 114, 122333.
  116. Awoniyi, D.O.; Aboua, Y.G.; Marnewick, J.; Brooks, N. The effects of rooibos (Aspalathus linearis), green tea (Camellia sinensis) and commercial rooibos and green tea supplements on epididymal sperm in oxidative stress-induced rats. Phytother. Res. 2012, 26, 1231–1239.
  117. Zanchi, M.M.; Manfredini, V.; Dos Santos Brum, D.; Vargas, L.M.; Spiazzi, C.C.; Soares, M.B.; Izaguirry, A.P.; Santos, F.W. Green tea infusion improves cyclophosphamide-induced damage on male mice reproductive system. Toxicol. Rep. 2015, 2, 252–260.
  118. Sato, K.; Sueoka, K.; Tanigaki, R.; Tajima, H.; Nakabayashi, A.; Yoshimura, Y.; Hosoi, Y. Green tea extracts attenuate doxorubicin-induced spermatogenic disorders in conjunction with higher telomerase activity in mice. J. Assist. Reprod. Genet. 2010, 27, 501–508.
  119. Abshenas, J.; Babaei, H.; Zare, M.H.; Allahbakhshi, A.; Sharififar, F. The effects of green tea (Camellia sinensis) extract on mouse semen quality after scrotal heat stress. Vet. Res. Forum 2012, 2, 242–247.
  120. Greifova, H.; Tvrda, E.; Jambor, T.; Lukac, N. Dose- and time-dependent effects of epicatechin on bovine spermatozoa in vitro. J. Microbiol. Biotechnol. Food Sci. 2018, 2, 235–239.
  121. Tvrda, E.; Straka, P.; Galbavy, D.; Ivanic, P. Epicatechin Provides Antioxidant Protection to Bovine Spermatozoa Subjected to Induced Oxidative Stress. Molecules 2019, 24, 3226.
  122. Wittayarat, M.; Ito, A.; Kimura, T.; Namula, Z.; Luu, V.V.; Do, L.T.; Sato, Y.; Taniguchi, M.; Otoi, T. Effects of green tea polyphenol on the quality of canine semen after long-term storage at 5 °C. Reprod. Biol. 2013, 13, 251–254.
  123. Purdy, P.H.; Ericsson, S.A.; Dodson, R.E.; Sternes, K.L.; Garner, D.L. Effects of flavonoids, silibinin and catechin, on the motility of extended cooled caprine sperm. Small Rumin. Res. 2004, 55, 239–243.
  124. Silva, E.C.B.; Arruda, L.C.P.; Vieira, J.I.T.; Soares, P.C.; Guerra, M.M.P. (+)-Catechin and (-)-epigallocatechin gallate: Are these promising antioxidant therapies for frozen goat semen? Arq. Bras. Med. Vet. Zootec. 2019, 71, 521–528.
  125. Boonsorn, T.; Kongbuntad, W.; Narkkong, N.; Aengwanich, W. Effects of catechin addition to extender on sperm quality and lipid peroxidation in boar semen. Am. Eurasian J. Sustain. Agric. 2010, 7, 283–288.
  126. Watson, P.F. The effects of cold shock on sperm cell membranes. In Effects of Low Temperatures on Biological Membranes, 1st ed.; Morris, G.J., Clarke, A., Eds.; Academic Press: London, UK, 1981; pp. 189–218.
  127. De Amicis, F.; Santoro, M.; Guido, C.; Russo, A.; Aquila, S. Epigallocatechin gallate affects survival and metabolism of human sperm. Mol. Nutr. Food Res. 2012, 56, 1655–1664.
  128. Aquila, S.; Sisci, D.; Gentile, M.; Middea, E.; Siciliano, L.; Andó, S. Human ejaculated spermatozoa contain active P450 aromatase. J. Clin. Endocrinol. Metab. 2002, 87, 3385–3390.
  129. Spinaci, M.; Volpe, S.; De Ambrogi, M.; Tamanini, C.; Galeati, G. Effects of epigallocatechin-3-gallate (EGCG) on in vitro maturation and fertilization of porcine oocytes. Theriogenology 2008, 69, 877–885.
  130. Uekusa, Y.; Kamihira, M.; Nakayama, T. Dynamic behavior of tea catechins interacting with lipid membranes as determined by NMR spectroscopy. J. Agric. Food Chem. 2007, 55, 9986–9992.
  131. Nagata, H.; Takekoshi, S.; Takagi, T.; Honma, T.; Watanabe, K. Antioxidative action of flavonoids, quercetin and catechin, mediated by the activation of glutathione peroxidase. Tokai J. Exp. Clin. Med. 1999, 24, 1–11.
  132. Das, S.K.; Karmakar, S.N. Effect of green tea (camellia sinensis l.) leaf extract on reproductive system of adult male albino rats. Int. J. Physiol. Pathophysiol. Pharmacol. 2015, 7, 178–184.
  133. Jamalan, M.; Ghaffari, M.A.; Hoseinzadeh, P.; Hashemitabar, M.; Zeinali, M. Human Sperm Quality and Metal Toxicants: Protective Effects of some Flavonoids on Male Reproductive Function. Int. J. Fertil. Steril. 2016, 10, 215–523.
  134. Kennedy, J.H.; Korn, N.; Thurston, R.J. Prostaglandin levels in seminal plasma and sperm extracts of the domestic turkey, and the effects of cyclooxygenase inhibitors on sperm mobility. Reprod. Biol. Endocrinol. 2003, 1, 74.
  135. Suh, K.S.; Chon, S.; Oh, S.; Kim, S.W.; Kim, J.-W.; Kim, Y.S.; Woo, J.-T. Prooxidative effects of green tea polyphenol (−)-epigallocatethin-3-gallate on the HIT-T15 pancreatic beta cell line. Cell. Biol. Toxicol. 2010, 26, 189–199.
  136. Sang, S.; Hou, Z.; Lambert, J.D.; Yang, C.S. Redox properties of tea polyphenols and related biological activities. Antioxid. Redox Signal. 2005, 7, 1704–1714.
More