Ameliorative effects of propolis upon reproductive toxicity in males

Article information

Clin Exp Reprod Med. 2023;50(1):12-18
Publication date (electronic) : 2023 February 23
doi : https://doi.org/10.5653/cerm.2022.05785
Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
Corresponding author: Saleem Ali Banihani Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan Tel: +962-798867702 Fax: +962-2-7095070 E-mail: sabanihani@just.edu.jo
Received 2022 October 31; Revised 2023 January 2; Accepted 2023 January 14.

Abstract

Propolis is a sticky natural product produced by honeybees. Research studies have discussed the effectiveness of propolis, directly or indirectly, for ameliorating reproductive toxicity in males; however, this research has not yet been reviewed. The current paper presents an integrative summary of all research studies in Scopus and PubMed that investigated the effects of propolis on semen quality, and hence on male fertility, in conditions of reproductive toxicity. The consensus indicates that propolis ameliorates reproductive toxicity and enhances semen quality in vivo in test animals. These effects may be attributable to the ability of propolis to reduce testicular oxidative damage, enhance testicular antioxidant defense mechanisms, increase nitric oxide production, reduce testicular apoptotic injury, and boost testosterone production. However, to generalize these effects in humans would require further research.

Introduction

Propolis, also called bee glue, is a resinous dark-brown mixture produced by honeybees, generally by mixing beeswax and bee saliva with exudates collected from sap flows, tree buds, or other botanic sources [1,2]. Since 1945, particularly in the Scopus database, approximately 7000 published research articles on the impact of propolis on human health have been published, forming a scientific consensus that propolis has potential pharmacological properties. The number of publications on the health effects of propolis has increased dramatically in the last decade. As a result, propolis has been recognized as having antioxidant [3-6], anticancer [7-10], anti-inflammatory [11-13], and antimicrobial and antiviral activities [4,5,14,15]. It has also been recognized as an effective natural product in various diseases and disorders such as cardiovascular diseases [16,17], skin injury and skin care [18,19], diabetes [20-22], neuronal degenerative diseases [23-25], and hypertension [26-28].

Amid the recent increase in research on the ability of propolis to ameliorate specific diseases studies have also investigated the effectiveness of propolis in counteracting reproductive toxicity in males. To date, the PubMed database contains 26 articles that associate propolis and male reproductive toxicity. The current paper reviews these studies and presents an integrative summary of this specific research context. To achieve this collective summary, we gathered studies from Scopus and PubMed that are published in English and that investigate the effects of propolis on male reproductive toxicity. To extract all related studies, we used the keywords “propolis” in combination with “sperm” and “semen.” In addition, to incorporate an explanatory summary of how propolis may function in conditions of reproductive toxicity, relevant mechanistic studies were discussed and reviewed.

Effect of propolis on semen quality in induced reproductive toxicity conditions

Table 1 shows the direct research studies that investigate and discuss the ameliorative effect of propolis against various reproductive toxicants in males [29-38]. These studies were conducted in the last two decades, during which attention toward natural medicine has dramatically increased worldwide [39], and the majority were performed in the Middle East (Egypt and Turkey). One reason for this trend may be that many cohorts in the Middle East are relying on natural remedies [40,41].

Direct research studies on propolis and its reported effects on semen quality parameters in different conditions of reproductive toxicity induced in males

Almost all included studies that discussed the ameliorative effect of propolis against reproductive toxicants were conducted on animals. The one exception, an in vitro study, was conducted on ejaculated human semen (Table 1). Half of the reproductive toxicants in these studies were chosen to be anticancer agents (cisplatin, paclitaxel, mitomycin C, doxorubicin, and streptozotocin), three were toxic metals (aluminum, cadmium, and copper); one was an anti-fungal agent (triphenyltin), and one was a cancerous agent (benzo[a]pyrene). In in vivo system studies, the duration of exposure to these toxicants ranged from 17 to 84 days, while the propolis dose ranged from 200 mg/kg of body weight (BW), equivalent to approximately 33.3 mg/kg of human BW, to 400 mg/kg of BW, equivalent to 66.7 mg/kg of human BW [42].

Together, almost all included studies revealed protective effects against induced reproductive toxicity (Table 1). In the animals tested, propolis improved semen quality measures such as sperm motility (six studies), sperm quantity (six studies), and normal forms of sperm (four studies). Among the published studies, only one, conducted in India on male mice, revealed a blunted effect of propolis on semen quality measures against mitomycin C-induced reproductive toxicity.

Collectively, these studies form a consensus that propolis may enhance semen quality in vivo when specific reproductive toxicity conditions are induced. However, such effects cannot be generalized to humans without further research.

Mechanisms of action of propolis in ameliorating reproductive toxicity

Gonadal hormones in males, especially testosterone and luteinizing hormone, are considered the primary indicators of sensitivity to exogenous reproductive toxicants. As a result, reducing induced reproductive toxicity by supporting production of these hormones, particularly testosterone, is an important way to measure the bioactivity of a given natural or synthetic substance, due to the hormones’ role in controlling spermatogenesis in males. Briefly, testosterone, which is a steroid hormone chemically derived from cholesterol, is produced in Leydig cells in response to stimulation by luteinizing hormone [43,44], a peptide hormone released from the pituitary gland. It diffuses into Sertoli cells in the seminiferous tubules in the testis [45]. Then testosterone binds to androgen receptors in the cytoplasm and nucleus of Sertoli cells, and triggers the cellular responses needed to support spermatogenesis [45].

Several studies have tested the ability of propolis to counteract a reduction in testosterone induced by exogenous toxicants [33-35,46]. Certain bioactive compounds in propolis, such as the flavonoid chrysin, were found to boost testosterone production in the testes [47]. At the molecular level, propolis was found to enhance steroidogenesis in the testes of diabetic rats by up-regulating the mRNA as well as protein levels of steroidogenic acute regulatory protein (StAR), cytochrome P450 A1 (CYP11A1; an enzyme that converts cholesterol to pregnenolone), cytochrome P450 17A1 (CYP17A1), 3β-hydroxysteroid dehydrogenase (3β-HSD), and 17β-hydroxysteroid dehydrogenase (17β-HSD) [30]. Therefore, the amelioration in semen quality parameters under conditions of reproductive toxicity may be due to the increased levels of testosterone.

Propolis extracts have been identified as having a strong antioxidant activity [48-50]. In vitro investigations revealed that the antioxidant power of propolis is attributable to the existence of many bioactive compounds, particularly phenolic compounds such as flavonoids (e.g., tectochrysin, chrysin, pinocembrin, galangin, apigenin, genkwanin, and kaempferol [51]), stilbenes, flavan-3-ols (catechins), and hydroxybenzoic acids [52]. Additional important phenolic compounds in propolis are cinnamic acid and its derivatives (e.g., hydroxycinnamic acids, ferulic acid, caffeic acid, isoferulic acid, and p-coumaric acid), as well as gallic acid, which have been identified in Polish propolis [53,54].

One known route by which exogenous toxicants induce reproductive toxicity in males is by reducing the activity of testicular antioxidant defense mechanisms. Testicular glutathione (GSH) as well as the testicular antioxidant enzymes glutathione peroxidase, catalase, and superoxide dismutase are often used to measure reproductive toxicity. As Table 2 shows, propolis was able to counteract the induced reduction in antioxidant defense mechanisms by enhancing GSH and antioxidant enzymes in the testis [29-38,46,55]. Given that a reduction in antioxidant defense mechanisms is associated with a decrease in semen quality [56,57], it is possible to infer that the favorable antioxidant effects of propolis may support its ability to improve semen quality when reproductive toxicity is induced.

Mechanistic studies on the effect of propolis in conditions of induced reproductive toxicity utilizing different toxicants

Malonyl dialdehyde (MDA), which is a stable end product created from peroxidation of essential (i.e., polyunsaturated) fatty acids in the cell [58], is considered a marker of increased cellular oxidation reactions beyond the capacity of antioxidant defense mechanisms, a state termed oxidative stress [59-62]. Among 12 mechanistic studies, five have revealed a reduction in testicular MDA when propolis is present amid induced toxicity conditions (Table 2). This scientific accord confirms the ability of propolis to protect against induced testicular oxidative damage by exogenous chemical toxicants.

One measure of reproductive toxicity is apoptosis in testicular cells. It is mediated by caspases, proteolytic enzymes belonging to the cysteine protease family, which provoke cell death by cleaving specific proteins in the nucleus and cytoplasm. As shown in Table 2, propolis treatment was found to reduce copper- [36], cadmium- [35], and doxorubicin-induced apoptotic cells in the testes [37], specifically by reducing increased levels of caspases, in particular caspase-3, in induced reproductive toxicity conditions [37]. In addition, propolis treatment prevented the immunoreactivity of hypoxia-inducible factor 1-alpha, and its expression in the testes of the propolis-treated group and the control group was found to be similar, compared to the cadmium-induced group [35].

A second useful marker of reproductive toxicity is the activity of testicular lactate dehydrogenase. In germ cells, lactate dehydrogenase, particularly type C, is required for energy (adenosine triphosphate) production [63]. In effect, a decline in the availability of lactate for germ cell development was found to contribute to the impairment of male reproductive function [63,64]. Propolis administration has been shown to enhance the activity of lactate dehydrogenase in conditions of induced reproductive toxicity, such as streptozotocin- and benzo[a]pyrene-induced toxicity [38,30].

Nitric oxide is a gaseous signaling molecule synthesized naturally in the body by nitric oxide synthases [47,65,66]. It acts as a vasodilator, particularly for blood vessels and inner muscles, which consequently increases circulation in these tissues [67]. Nitric oxide has been found to have a crucial role in male reproduction [68,69]. For example, it boosts the blood flow in the testes, thereby enhancing the activity of their physiological processes [70]. One important example is boosting testosterone synthesis by Leydig cells and subsequently activating spermatogenesis in Sertoli cells [71-73]. In male rats, propolis has been found to increase nitric oxide production in testicular tissue [55], which may support spermatogenesis.

In summary, there is evidence that propolis ameliorates reproductive toxicity and improves semen quality in vivo in test animals, but not in humans. Mechanistically, the protective effects of propolis may be due to reducing testicular oxidative damage, enhancing testicular antioxidant defense mechanisms, enhancing production of nitric oxide, reducing testicular apoptosis and cell death, and improving testosterone production.

Notes

Conflict of interest

No potential conflict of interest relevant to this article was reported.

References

1. Hwang S, Hwang S, Jo M, Lee CG, Rhee KJ. Oral administration of Korean propolis extract ameliorates DSS-induced colitis in BALB/c mice. Int J Med Sci 2020;17:1984–91.
2. Sforcin JM. Biological properties and therapeutic applications of propolis. Phytother Res 2016;30:894–905.
3. Bae IA, Ha JW, Choi JY, Boo YC. Antioxidant effects of Korean propolis in HaCaT keratinocytes exposed to particulate matter 10. Antioxidants (Basel) 2022;11:781.
4. Freitas AS, Cunha A, Oliveira R, Almeida-Aguiar C. Propolis antibacterial and antioxidant synergisms with gentamicin and honey. J Appl Microbiol 2022;132:2733–45.
5. Nichitoi MM, Josceanu AM, Isopescu RD, Isopencu GO, Geana EI, Ciucure CT, et al. Polyphenolics profile effects upon the antioxidant and antimicrobial activity of propolis extracts. Sci Rep 2021;11:20113.
6. Kasiotis KM, Anastasiadou P, Papadopoulos A, Machera K. Revisiting Greek propolis: chromatographic analysis and antioxidant activity study. PLoS One 2017;12e0170077.
7. Forma E, Brys M. Anticancer activity of propolis and its compounds. Nutrients 2021;13:2594.
8. Chan GC, Cheung KW, Sze DM. The immunomodulatory and anticancer properties of propolis. Clin Rev Allergy Immunol 2013;44:262–73.
9. Begnini KR, Moura de Leon PM, Thurow H, Schultze E, Campos VF, Martins Rodrigues F, et al. Brazilian red propolis induces apoptosis-like cell death and decreases migration potential in bladder cancer cells. Evid Based Complement Alternat Med 2014;2014:639856.
10. Desamero MJ, Kakuta S, Tang Y, Chambers JK, Uchida K, Estacio MA, et al. Tumor-suppressing potential of stingless bee propolis in in vitro and in vivo models of differentiated-type gastric adenocarcinoma. Sci Rep 2019;9:19635.
11. Inui S, Hosoya T, Yoshizumi K, Sato H, Kumazawa S. Phytochemical and anti-inflammatory properties of Senegalese propolis and isolated compounds. Fitoterapia 2021;151:104861.
12. Nani BD, Sardi JC, Lazarini JG, Silva DR, Massariolli AP, Cunha TM, et al. Anti-inflammatory and anti-Candida effects of brazilian organic propolis, a promising source of bioactive molecules and functional food. J Agric Food Chem 2020;68:2861–71.
13. Franchin M, Freires IA, Lazarini JG, Nani BD, da Cunha MG, Colon DF, et al. The use of Brazilian propolis for discovery and development of novel anti-inflammatory drugs. Eur J Med Chem 2018;153:49–55.
14. Gekker G, Hu S, Spivak M, Lokensgard JR, Peterson PK. Anti-HIV-1 activity of propolis in CD4(+) lymphocyte and microglial cell cultures. J Ethnopharmacol 2005;102:158–63.
15. Schnitzler P, Neuner A, Nolkemper S, Zundel C, Nowack H, Sensch KH, et al. Antiviral activity and mode of action of propolis extracts and selected compounds. Phytother Res 2010;24 Suppl 1:S20–8.
16. Braik A, Lahouel M, Merabet R, Djebar MR, Morin D. Myocardial protection by propolis during prolonged hypothermic preservation. Cryobiology 2019;88:29–37.
17. Daleprane JB, Abdalla DS. Emerging roles of propolis: antioxidant, cardioprotective, and antiangiogenic actions. Evid Based Complement Alternat Med 2013;2013:175135.
18. Kurek-Gorecka A, Gorecki M, Rzepecka-Stojko A, Balwierz R, Stojko J. Bee products in dermatology and skin care. Molecules 2020;25:556.
19. McLoone P, Tabys D, Fyfe L. Honey combination therapies for skin and wound infections: a systematic review of the literature. Clin Cosmet Investig Dermatol 2020;13:875–88.
20. Kitamura H. Effects of propolis extract and propolis-derived compounds on obesity and diabetes: knowledge from cellular and animal models. Molecules 2019;24:4394.
21. Picolotto A, Pergher D, Pereira GP, Machado KG, da Silva Barud H, Roesch-Ely M, et al. Bacterial cellulose membrane associated with red propolis as phytomodulator: improved healing effects in experimental models of diabetes mellitus. Biomed Pharmacother 2019;112:108640.
22. Tsuda T, Kumazawa S. Propolis: chemical constituents, plant origin, and possible role in the prevention and treatment of obesity and diabetes. J Agric Food Chem 2021;69:15484–94.
23. Ali AM, Kunugi H. Apitherapy for Parkinson’s disease: a focus on the effects of propolis and royal jelly. Oxid Med Cell Longev 2020;2020:1727142.
24. Shahinozzaman M, Taira N, Ishii T, Halim MA, Hossain MA, Tawata S. Anti-inflammatory, anti-diabetic, and anti-Alzheimer’s effects of prenylated flavonoids from Okinawa propolis: an investigation by experimental and computational studies. Molecules 2018;23:2479.
25. Goncalves VC, Pinheiro DJ, de la Rosa T, de Almeida AG, Scorza FA, Scorza CA. Propolis as a potential disease-modifying strategy in Parkinson’s disease: cardioprotective and neuroprotective effects in the 6-OHDA rat model. Nutrients 2020;12:1551.
26. Braakhuis A. Evidence on the health benefits of supplemental propolis. Nutrients 2019;11:2705.
27. Zhou H, Wang H, Shi N, Wu F. Potential protective effects of the water-soluble Chinese propolis on hypertension induced by high-salt intake. Clin Transl Sci 2020;13:907–15.
28. Teles F, da Silva TM, da Cruz Junior FP, Honorato VH, de Oliveira Costa H, Barbosa AP, et al. Brazilian red propolis attenuates hypertension and renal damage in 5/6 renal ablation model. PLoS One 2015;10e0116535.
29. Tatli Seven P, Seven I, Karakus S, Iflazoglu Mutlu S, Ozer Kaya S, Arkali G, et al. The in-vivo assessment of Turkish propolis and its nano form on testicular damage induced by cisplatin. J Integr Med 2021;19:451–9.
30. Nna VU, Bakar AB, Ahmad A, Umar UZ, Suleiman JB, Zakaria Z, et al. Malaysian propolis and metformin mitigate subfertility in streptozotocin-induced diabetic male rats by targeting steroidogenesis, testicular lactate transport, spermatogenesis and mating behaviour. Andrology 2020;8:731–46.
31. Abd-Elrazek AM, El-Dash HA, Said NI. The role of propolis against paclitaxel-induced oligospermia, sperm abnormality, oxidative stress and DNA damage in testes of male rats. Andrologia 2020;52e13394.
32. Kumari S, Nayak G, Lukose ST, Kalthur SG, Bhat N, Hegde AR, et al. Indian propolis ameliorates the mitomycin C-induced testicular toxicity by reducing DNA damage and elevating the antioxidant activity. Biomed Pharmacother 2017;95:252–63.
33. Yousef MI, Kamel KI, Hassan MS, El-Morsy AM. Protective role of propolis against reproductive toxicity of triphenyltin in male rabbits. Food Chem Toxicol 2010;48:1846–52.
34. Yousef MI, Salama AF. Propolis protection from reproductive toxicity caused by aluminium chloride in male rats. Food Chem Toxicol 2009;47:1168–75.
35. Cilenk KT, Ozturk I, Sonmez MF. Ameliorative effect of propolis on the cadmium-induced reproductive toxicity in male albino rats. Exp Mol Pathol 2016;101:207–13.
36. Seven I, Tatli Seven P, Gul Baykalir B, Parlak Ak T, Ozer Kaya S, Yaman M. Bee glue (propolis) improves reproductive organs, sperm quality and histological changes and antioxidant parameters of testis tissues in rats exposed to excess copper. Andrologia 2020;52e13540.
37. Rizk SM, Zaki HF, Mina MA. Propolis attenuates doxorubicin-induced testicular toxicity in rats. Food Chem Toxicol 2014;67:176–86.
38. Russo A, Troncoso N, Sanchez F, Garbarino JA, Vanella A. Propolis protects human spermatozoa from DNA damage caused by benzo[a]pyrene and exogenous reactive oxygen species. Life Sci 2006;78:1401–6.
39. Lu DY, Lu TR, Yarla NS, Lu Y, Che JY, Ding J, et al. Natural drug cancer treatments, strategies from herbal medicine to chemical or biological drugs. Stud Nat Prod Chem 2020;66:91–115.
40. Abu-Odeh AM, Talib WH. Middle East medicinal plants in the treatment of diabetes: a review. Molecules 2021;26:742.
41. Abarikwu SO, Onuah CL, Singh SK. Plants in the management of male infertility. Andrologia 2020;52e13509.
42. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J 2008;22:659–61.
43. Banihani SA. Testosterone in males as enhanced by onion (Allium Cepa L.). Biomolecules 2019;9:75.
44. Yuefeng Y, Zhiqi L, Yi C, Keyu Z, Heng W, Yuying W, et al. Testosterone deficiency promotes hypercholesteremia and attenuates cholesterol liver uptake via AR/PCSK9/LDLR pathways. Int J Endocrinol 2022;2022:7989751.
45. Smith LB, Walker WH. The regulation of spermatogenesis by androgens. Semin Cell Dev Biol 2014;30:2–13.
46. Hashem AS. Defensive impact of propolis against CCl4 actuated rats’ testicular damage. J Adv Vet Anim Res 2021;8:70–7.
47. Darwish HA, Arab HH, Abdelsalam RM. Chrysin alleviates testicular dysfunction in adjuvant arthritic rats via suppression of inflammation and apoptosis: comparison with celecoxib. Toxicol Appl Pharmacol 2014;279:129–40.
48. Russo A, Longo R, Vanella A. Antioxidant activity of propolis: role of caffeic acid phenethyl ester and galangin. Fitoterapia 2002;73 Suppl 1:S21–9.
49. Kocot J, Kielczykowska M, Luchowska-Kocot D, Kurzepa J, Musik I. Antioxidant potential of propolis, bee pollen, and royal jelly: possible medical application. Oxid Med Cell Longev 2018;2018:7074209.
50. Zheng YZ, Deng G, Liang Q, Chen DF, Guo R, Lai RC. Antioxidant activity of quercetin and its glucosides from propolis: a theoretical study. Sci Rep 2017;7:7543.
51. Kurek-Gorecka A, Rzepecka-Stojko A, Gorecki M, Stojko J, Sosada M, Swierczek-Zieba G. Structure and antioxidant activity of polyphenols derived from propolis. Molecules 2013;19:78–101.
52. Laaroussi H, Bakour M, Ousaaid D, Ferreira-Santos P, Genisheva Z, El Ghouizi A, et al. Protective effect of honey and propolis against gentamicin-induced oxidative stress and hepatorenal damages. Oxid Med Cell Longev 2021;2021:9719906.
53. Moskwa J, Naliwajko SK, Markiewicz-Zukowska R, Gromkowska-Kepka KJ, Nowakowski P, Strawa JW, et al. Chemical composition of Polish propolis and its antiproliferative effect in combination with Bacopa monnieri on glioblastoma cell lines. Sci Rep 2020;10:21127.
54. Wezgowiec J, Wieczynska A, Wieckiewicz W, Kulbacka J, Saczko J, Pachura N, et al. Polish propolis-chemical composition and biological effects in tongue cancer cells and macrophages. Molecules 2020;25:2426.
55. Selamoglu Talas Z. Propolis reduces oxidative stress in l-NAME-induced hypertension rats. Cell Biochem Funct 2014;32:150–4.
56. Asadi N, Bahmani M, Kheradmand A, Rafieian-Kopaei M. The impact of oxidative stress on testicular function and the role of antioxidants in improving it: a review. J Clin Diagn Res 2017;11:IE01–5.
57. Dutta S, Sengupta P, Slama P, Roychoudhury S. Oxidative stress, testicular inflammatory pathways, and male reproduction. Int J Mol Sci 2021;22:10043.
58. Mas-Bargues C, Escriva C, Dromant M, Borras C, Vina J. Lipid peroxidation as measured by chromatographic determination of malondialdehyde: human plasma reference values in health and disease. Arch Biochem Biophys 2021;709:108941.
59. Banihani SA. Effect of paracetamol on semen quality. Andrologia 2018;50e12874.
60. Lushchak VI. Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact 2014;224:164–75.
61. Alzoubi KH, Hasan ZA, Khabour OF, Mayyas FA, Al Yacoub ON, Banihani SA, et al. Vitamin E modifies high-fat diet-induced reduction of seizure threshold in rats: role of oxidative stress. Physiol Behav 2019;206:200–5.
62. Etemadi T, Momeni HR, Darbandi N, Abnosi MH. Silymarin modulates cadmium-induced oxidative stress in human spermatozoa. Andrologia 2022;54e14475.
63. Nna VU, Abu Bakar AB, Ahmad A, Mohamed M. Metformin mitigates impaired testicular lactate transport/utilisation and improves sexual behaviour in streptozotocin-induced diabetic rats. Arch Physiol Biochem 2021;127:51–60.
64. Li ZM, Liu N, Jiang YP, Yang JM, Zheng J, Sun M, et al. Vitexin alleviates streptozotocin-induced sexual dysfunction and fertility impairments in male mice via modulating the hypothalamus-pituitary-gonadal axis. Chem Biol Interact 2019;297:119–29.
65. Lowenstein CJ. Metabolism reprogrammed by the nitric oxide signalling molecule. Nature 2019;565:33–4.
66. Banihani SA, Khasawneh FH. Effect of lansoprazole on DNA integrity of human spermatozoa and activity of seminal creatine kinase. Andrologia 2020;52e13564.
67. Toda N, Okamura T. Modulation of renal blood flow and vascular tone by neuronal nitric oxide synthase-derived nitric oxide. J Vasc Res 2011;48:1–10.
68. Banihani SA, Shatnawi RM. Aspirin decreases human sperm motility and vitality, chelates seminal calcium, but insignificantly reduces seminal nitric oxide production. Andrologia 2020;52e13776.
69. Luo Y, Zhu Y, Basang W, Wang X, Li C, Zhou X. Roles of nitric oxide in the regulation of reproduction: a review. Front Endocrinol (Lausanne) 2021;12:752410.
70. Middendorff R, Muller D, Wichers S, Holstein AF, Davidoff MS. Evidence for production and functional activity of nitric oxide in seminiferous tubules and blood vessels of the human testis. J Clin Endocrinol Metab 1997;82:4154–61.
71. El-Migdadi F, Banihani I, Banihani SA. Clinico-hormonal correlation of oligospermic patients in the below sea level environment (Jordan Valley). Neuro Endocrinol Lett 2005;26:13–8.
72. Christin-Maitre S, Young J. Androgens and spermatogenesis. Ann Endocrinol (Paris) 2022;83:155–8.
73. Oduwole OO, Huhtaniemi IT, Misrahi M. The roles of luteinizing hormone, follicle-stimulating hormone and testosterone in spermatogenesis and folliculogenesis revisited. Int J Mol Sci 2021;22:12735.

Article information Continued

Table 1.

Direct research studies on propolis and its reported effects on semen quality parameters in different conditions of reproductive toxicity induced in males

Study origin (year) Study population Toxicant (dose) Propolis dose Duration Effects on semen quality Reference
Turkey (2021) Male Sprague-Dawley rats Cisplatin (3 mg/kg) 100 mg/kg 21 days (+) Sperm motility [29]
Malaysia (2020) Male Sprague-Dawley rats Streptozotocin (60 mg/kg) 300 mg/kg 28 days (+) Motility [30]
(+) Normal forms of sperm
(+) Sperm count
(+) Vitality
(–) Sperm DNA damage
Egypt (2020) Adult male Sprague-Dawley rats Paclitaxel (5 mg/kg) 50 mg/kg 28 days (+) Motility [31]
(+) Count
(+) Viability
(–) Sperm abnormality
(–) Sperm DNA damage
India (2017) Male mice Mitomycin C (2, 4, and 8 mg/kg body weight) 400 mg/kg 35 days (+/–) Sperm Motility [32]
(+/–) Sperm morphology
(+/–) Sperm count
(–) Sperm DNA damage
Egypt (2010) Male New Zealand white rabbits Triphenyltin (0.5 mg/kg body weight) 50 mg/kg Daily for 84 days (+) Sperm [33]
(+) Concentration
(+) Sperm motility
(+) Normal forms of sperm
Egypt (2009) Male rats Aluminum chloride (34 mg/kg body weight) 50 mg/kg Daily for 70 days (+) Sperm concentration [34]
(+) Sperm motility
(+) Vitality
(+) Normal forms of sperm
Turkey (2016) Male albino rats Cadmium (1 mg/kg/day) 50 mg/kg 17 days NA [35]
Turkey (2020) Male Sprague-Dawley rats Copper (128 mg/kg) 100 mg/kg 21 days (+) Sperm motility [36]
(+) Sperm concentration
(–) Abnormal sperm
Egypt (2014) Male Wistar albino rats Doxorubicin (6 doses at 18 mg kg) 200 mg/kg (5 day/wk) 21 days (+) Sperm count [37]
Italy (2006) Human ejaculated sperm Benzo[a]pyrene (500 µM) 6, 12, and 25 µg/mL 1 hour (–) Sperm DNA damage [38]

Table 2.

Mechanistic studies on the effect of propolis in conditions of induced reproductive toxicity utilizing different toxicants

Toxicant (dose) Study population Propolis dose (duration) Mechanisms of the actions of propolis in the presence of toxicant Reference
Cisplatin (3 mg/kg) Male rats 100 mg/kg (21 days) (+) Testicular GSH, CAT, GPx, and Bcl2 [29]
(–) Testicular MDA and Bax
Streptozotocin (60 mg/kg) Male rats 300 mg/kg (28 days) (+) Testicular testosterone and serum testosterone [30]
(+) mRNA of AR and LH receptor
(+) StAR, CYP11A1, CYP17A1, 3β-HSD and 17β-HSD proteins
(+) Testicular MCT2, MCT4, and mRNA of LDHc
Paclitaxel (5 mg/kg) Adult male rats 50 mg/kg/day (28 days) (–) Testicular DNA damage [31]
(+) Testicular GSH content
Mitomycin C (2, 4, and 8 mg/kg BW) Male mice 400 mg/kg (35 days) (+) Testicular testosterone [32]
(+) Inhibin B level
(–) Testicular oxidative stress
Triphenyltin (0.5 mg/kg BW) Male rabbits 50 mg/kg (84 days) (+) Plasma testosterone [33]
(+) Weights of epididymis and testes
(+) Seminal plasma glutathione S-transferase
(+) Seminal plasma phosphatases and transaminases
(–) Seminal plasma TBARS
(–) Seminal plasma LDH
Aluminum chloride (34 mg/kg BW) Male rats 50 mg/kg (70 days) (+) Plasma testosterone [34]
(+) Plasma 17-ketosteroid reductase
(+) Testicular GST and CAT
(+) Testicular GSH
Cadmium (1 mg/kg/day) Male rats 50 mg/kg (17 days) (–) Number of apoptotic cells in the testes [35]
(–) HIF-1α expression
(+/–) Serum testosterone
Copper (128 mg/kg) Male rats 100 mg/kg/day (21 days) (+) Testicular SOD, CAT, GSH [36]
(+) Johnsen's testicular score
(+) Spermatogenesis activity
(–) Testicular MDA
(–) Number of apoptotic cells in the testes
Doxorubicin (6 doses at 18 mg/kg) Male rats 200 mg/kg (5 day/wk) (21 days) (+) Testicular interleukin-4 content [37]
(+) Testicular GSH
(–) Testicular MDA
(–) Testicular myeloperoxidase and caspase-3
(–) Testicular TNFα and Fas-L
(–) Number of apoptotic cells in the testes
Benzo[a]pyrene (500 µM) Human ejaculated sperm 6, 12, and 25 µg/mL (1 hour) (–) H2O2-induced oxidation in sperm [38]
(–) TBARS formation in sperm
(–) LDH release in sperm
CCl4 (3 mL/kg/day) Adult male albino rats 200 mg/kg/day (28 days) (+) Serum testosterone [46]
(–) Testicular MDA
(+) Testicular GSH
(+) Testicular GPx, CAT, and SOD
(–) Testicular TNFα gene expression
(–) Testicular inflammation
Nω-nitro-l-arginine methyl ester (l-NAME) (40 mg/kg/day, for 15 days) Hypertensive male rats 200 mg/kg (5 days) (+) Testicular MDA [55]
(–) Testicular CAT
(+/–) Nitric oxide ([+] with only propolis))

GSH, glutathione; CAT, catalase; GPx, glutathione peroxidase; Bcl2, B-cell lymphoma 2; MDA, malonyl dialdehyde; BAX, BCL2 associated X protein; AR, androgen receptor; LH, luteinizing hormone; StAR, steroidogenesis acute regulatory protein; CYP11A1, cytochrome P450 A1; CYP17A1, cytochrome P450 17A1; HSD, hydroxysteroid dehydrogenase; MCT, monocarboxylate transporter; LDHc, lactate dehydrogenase type C; BW, body weight; TBARS, thiobarbituric acid-reactive substances; LDH, lactate dehydrogenase; GST, glutathione-S-transferase; HIF-1α, hypoxia-inducible factor 1-alpha; SOD, superoxide dismutase; TNFα, tumor necrosis factor alpha; Fas-L, Fas ligand.