Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2024

Open Access 01-12-2024 | Research

Effect of melatonin on developmental competence, mitochondrial distribution, and intensity of fresh and vitrified/thawed in vitro matured buffalo oocytes

Authors: Omaima Mohamed Kandil, Samar Mahfouz Abd El Rahman, Rania S. Ali, Esraa Aly Ismail, Nehad M. Ibrahim

Published in: Reproductive Biology and Endocrinology | Issue 1/2024

Login to get access

Abstract

Background: In livestock breeding, oocyte cryopreservation is crucial for preserving and transferring superior genetic traits. This study was conducted to examine the additional effect of melatonin to maturation and vitrification media on the in vitro developmental capacity, mitochondrial distribution, and intensity of buffalo oocytes. The study involved obtaining ovaries from a slaughterhouse and conducting two phases. In the first phase, high-quality oocytes were incubated in a maturation medium with or without 10−9M melatonin for 22 h (at 38.5°C in 5% CO2). Matured oocytes were fertilized in vitro and cultured in SOF media for seven days. In the second phase, vitrified in vitro matured oocytes were stored in vitrified media (basic media (BM) containing a combination of cryoprotectants (20% Ethyl Glycol and 20% Dimethyl sulfoxide), with or without melatonin, and then stored in liquid nitrogen. Normal vitrified/thawed oocytes were fertilized in vitro and cultured as described. Finally, the matured oocytes from the fresh and vitrified/thawed groups, both with and without melatonin, were stained using DAPI and Mitotracker red to detect their viability (nuclear maturation), mitochondrial intensity, and distribution using a confocal microscope. The study found that adding 10−9M melatonin to the maturation media significantly increased maturation (85.47%), fertilization rate (84.21%)cleavage (89.58%), and transferable embryo (48.83%) rates compared to the group without melatonin (69.85%,79.88%, 75.55%, and 37.25% respectively). Besides that, the addition of melatonin to the vitrification media improved the recovery rate of normal oocytes (83.75%), as well as the cleavage (61.80%) and transferable embryo (27.00%) rates when compared to the vitrified TCM group (67.46%, 51.40%, and 17.00%, respectively). The diffuse mitochondrial distribution was higher in fresh with melatonin (TCM + Mel) (80%) and vitrified with melatonin (VS2 + Mel groups) (76.70%), Furthermore, within the same group, while the mitochondrial intensity was higher in the TCM + Mel group (1698.60) than other group. In conclusion, Melatonin supplementation improves the developmental competence and mitochondrial distribution in buffalo oocytes in both cases(in vitro maturation and vitrification).
Appendix
Available only for authorised users
Literature
1.
go back to reference Baruselli PS, de Carvalho JGS, Elliff FM, da Silva JCB, Chello D, de Carvalho NAT. Embryo transfer in buffalo (Bubalus bubalis). Theriogenology. 2020;150:221–8.PubMedCrossRef Baruselli PS, de Carvalho JGS, Elliff FM, da Silva JCB, Chello D, de Carvalho NAT. Embryo transfer in buffalo (Bubalus bubalis). Theriogenology. 2020;150:221–8.PubMedCrossRef
2.
go back to reference Currin L, Baldassarre H, de Macedo MP, Glanzner WG, Gutierrez K, Lazaris K, et al. Factors affecting the efficiency of in vitro embryo production in prepubertal mediterranean water buffalo. Animals. 2022;12(24):3549.PubMedPubMedCentralCrossRef Currin L, Baldassarre H, de Macedo MP, Glanzner WG, Gutierrez K, Lazaris K, et al. Factors affecting the efficiency of in vitro embryo production in prepubertal mediterranean water buffalo. Animals. 2022;12(24):3549.PubMedPubMedCentralCrossRef
4.
go back to reference Lopes AS, Lane MTJ. Oxygen consumption and ROS production are increased at the time of fertilization and cell cleavage in bovine zygotes. Hum Reprod. 2010;25:2762–73.PubMedCrossRef Lopes AS, Lane MTJ. Oxygen consumption and ROS production are increased at the time of fertilization and cell cleavage in bovine zygotes. Hum Reprod. 2010;25:2762–73.PubMedCrossRef
5.
go back to reference Mukherjee A, Malik H, Saha AP, Dubey A, Singhal DK, Boateng S, et al. Resveratrol treatment during goat oocytes maturation enhances developmental competence of parthenogenetic and hand-made cloned blastocysts by modulating intracellular glutathione level and embryonic gene expression. J Assist Reprod Genet. 2014;31(2):229–39. Available from: http://link.springer.com/10.1007/s10815-013-0116-9.PubMedCrossRef Mukherjee A, Malik H, Saha AP, Dubey A, Singhal DK, Boateng S, et al. Resveratrol treatment during goat oocytes maturation enhances developmental competence of parthenogenetic and hand-made cloned blastocysts by modulating intracellular glutathione level and embryonic gene expression. J Assist Reprod Genet. 2014;31(2):229–39. Available from: http://​link.​springer.​com/​10.​1007/​s10815-013-0116-9.PubMedCrossRef
10.
go back to reference Kandil OM, Takla MS, Noseir WMB, Hattab SA. Effects of vitamin C and melatonin on the viability and mitochondrial distribution of in vitro matured dromedary camel oocytes. Egypt J Chem. 2022;65(3):395–405. Kandil OM, Takla MS, Noseir WMB, Hattab SA. Effects of vitamin C and melatonin on the viability and mitochondrial distribution of in vitro matured dromedary camel oocytes. Egypt J Chem. 2022;65(3):395–405.
13.
go back to reference El-Sanea AM, Abdoon ASS, Kandil OM, El-Toukhy NE, El-maaty AMA, Ahmed HH. Effect of oxygen tension and antioxidants on the developmental competence of buffalo oocytes cultured in vitro. Int J One Heal. 2021;14(1):78–84. El-Sanea AM, Abdoon ASS, Kandil OM, El-Toukhy NE, El-maaty AMA, Ahmed HH. Effect of oxygen tension and antioxidants on the developmental competence of buffalo oocytes cultured in vitro. Int J One Heal. 2021;14(1):78–84.
15.
go back to reference Ribeiro JC, Carrageta DF, Oliveira PF, Bernardino RL, Alves MG. Aquaporins and animal gamete cryopreservation: advances and future challenges. Animals. 2022;12(3):359.PubMedPubMedCentralCrossRef Ribeiro JC, Carrageta DF, Oliveira PF, Bernardino RL, Alves MG. Aquaporins and animal gamete cryopreservation: advances and future challenges. Animals. 2022;12(3):359.PubMedPubMedCentralCrossRef
16.
go back to reference Ismail EA, Elsayed MAI, Hemeida NA, Abdoon ASS, Kandil OM. Effect of different cryoprotectant agents on mitochondrial distribution and developmental competence in buffalo oocyte (Bubalus Bubalis). Egypt J Chem. 2022;65(1):539–53. Ismail EA, Elsayed MAI, Hemeida NA, Abdoon ASS, Kandil OM. Effect of different cryoprotectant agents on mitochondrial distribution and developmental competence in buffalo oocyte (Bubalus Bubalis). Egypt J Chem. 2022;65(1):539–53.
17.
go back to reference Jiang Y, Shi H, Liu Y, Zhao S, Zhao H. Applications of Melatonin in Female Reproduction in the Context of Oxidative Stress. Oxid Med Cell Longev. 2021;2021:6668365.PubMedPubMedCentralCrossRef Jiang Y, Shi H, Liu Y, Zhao S, Zhao H. Applications of Melatonin in Female Reproduction in the Context of Oxidative Stress. Oxid Med Cell Longev. 2021;2021:6668365.PubMedPubMedCentralCrossRef
18.
go back to reference Pan B, Qazi IH, Guo S, Yang J, Qin J, Lv T, et al. Melatonin improves the first cleavage of parthenogenetic embryos from vitrified–warmed mouse oocytes potentially by promoting cell cycle progression. J Anim Sci Biotechnol. 2021;12(1):1–17.CrossRef Pan B, Qazi IH, Guo S, Yang J, Qin J, Lv T, et al. Melatonin improves the first cleavage of parthenogenetic embryos from vitrified–warmed mouse oocytes potentially by promoting cell cycle progression. J Anim Sci Biotechnol. 2021;12(1):1–17.CrossRef
21.
go back to reference Gualtieri R, Kalthur G, Barbato V, Di Nardo M, Adiga SK, Talevi R. Mitochondrial dysfunction and oxidative stress caused by cryopreservation in reproductive cells. Antioxidants. 2021;10(3):1–23.CrossRef Gualtieri R, Kalthur G, Barbato V, Di Nardo M, Adiga SK, Talevi R. Mitochondrial dysfunction and oxidative stress caused by cryopreservation in reproductive cells. Antioxidants. 2021;10(3):1–23.CrossRef
28.
go back to reference Almohammed ZNH, Moghani-Ghoroghi F, Ragerdi-Kashani I, Fathi R, Tahaei LS, Naji M, et al. The effect of melatonin on mitochondrial function and autophagy in in vitro matured oocytes of aged mice. Cell J. 2020;22(1):9–16. Almohammed ZNH, Moghani-Ghoroghi F, Ragerdi-Kashani I, Fathi R, Tahaei LS, Naji M, et al. The effect of melatonin on mitochondrial function and autophagy in in vitro matured oocytes of aged mice. Cell J. 2020;22(1):9–16.
29.
go back to reference Kandil OM, Abdoon ASS, Murakami M, Otoi T, Suzuki T. New technique, using a portable co2 incubator, for the production of in vitro fertilized egyptian buffalo embryos. J Reprod Dev. 1999;45(5):315–20.CrossRef Kandil OM, Abdoon ASS, Murakami M, Otoi T, Suzuki T. New technique, using a portable co2 incubator, for the production of in vitro fertilized egyptian buffalo embryos. J Reprod Dev. 1999;45(5):315–20.CrossRef
31.
go back to reference Acuna-Castroviejo DN-NM, Reiter RJ, Escames G. Melatonin actions in the heart; more than a hormone. Melatonin Res. 2018;1:21–6.CrossRef Acuna-Castroviejo DN-NM, Reiter RJ, Escames G. Melatonin actions in the heart; more than a hormone. Melatonin Res. 2018;1:21–6.CrossRef
33.
go back to reference Manchester LC, Coto-Montes A, Boga JA, Andersen LP, Zhou Z, Galano A, et al. Melatonin: an ancient molecule that makes oxygen metabolically tolerable. J Pineal Res. 2015;59:403–19.PubMedCrossRef Manchester LC, Coto-Montes A, Boga JA, Andersen LP, Zhou Z, Galano A, et al. Melatonin: an ancient molecule that makes oxygen metabolically tolerable. J Pineal Res. 2015;59:403–19.PubMedCrossRef
36.
go back to reference Nagina G, Asima A, Nemat U, Shamim A. Effect of melatonin on maturation capacity and fertilization of Nili-Ravi buffalo (Bubalus bubalis) oocytes. Open Vet J. 2016;6(2):128–34.PubMedPubMedCentralCrossRef Nagina G, Asima A, Nemat U, Shamim A. Effect of melatonin on maturation capacity and fertilization of Nili-Ravi buffalo (Bubalus bubalis) oocytes. Open Vet J. 2016;6(2):128–34.PubMedPubMedCentralCrossRef
40.
go back to reference Pang Y, Zhao S, Sun Y, Jiang X, Hao H, Du W, et al. Protective effects of melatonin on the in vitro developmental competence of bovine oocytes. Anim Sci J. 2018;89(4):648–60.PubMedCrossRef Pang Y, Zhao S, Sun Y, Jiang X, Hao H, Du W, et al. Protective effects of melatonin on the in vitro developmental competence of bovine oocytes. Anim Sci J. 2018;89(4):648–60.PubMedCrossRef
41.
go back to reference Wu, Pan, Qazi, Yang, Guo, Yang, et al. Melatonin improves in vitro development of vitrified-warmed mouse germinal vesicle oocytes potentially via modulation of spindle assembly checkpoint-related genes. cells. 2019;8(9):1009. Available from: https://www.mdpi.com/2073-4409/8/9/1009. Wu, Pan, Qazi, Yang, Guo, Yang, et al. Melatonin improves in vitro development of vitrified-warmed mouse germinal vesicle oocytes potentially via modulation of spindle assembly checkpoint-related genes. cells. 2019;8(9):1009. Available from: https://​www.​mdpi.​com/​2073-4409/​8/​9/​1009.
42.
go back to reference Tian X, Wang F, Zhang L, He C, Ji P, Wang J, et al. Beneficial effects of melatonin on the in vitro maturation of sheep oocytes and its relation to melatonin receptors. Int J Mol Sci. 2017;18(4):834.PubMedPubMedCentralCrossRef Tian X, Wang F, Zhang L, He C, Ji P, Wang J, et al. Beneficial effects of melatonin on the in vitro maturation of sheep oocytes and its relation to melatonin receptors. Int J Mol Sci. 2017;18(4):834.PubMedPubMedCentralCrossRef
43.
go back to reference Saeedabadi S, Abazari-Kia AH, Rajabi H, Parivar K, Salehi M. Melatonin improves the developmental competence of goat Oocytes. Int J Fertil Steril. 2018;12(2):157–63.PubMedPubMedCentral Saeedabadi S, Abazari-Kia AH, Rajabi H, Parivar K, Salehi M. Melatonin improves the developmental competence of goat Oocytes. Int J Fertil Steril. 2018;12(2):157–63.PubMedPubMedCentral
44.
go back to reference Nishihara T, Hashimoto S, Ito K, Nakaoka Y, Matsumoto K, Hosoi Y, et al. Oral melatonin supplementation improves oocyte and embryo quality in women undergoing in vitro fertilization-embryo transfer. Gynecol Endocrinol. 2014;30(5):359–62.PubMedCrossRef Nishihara T, Hashimoto S, Ito K, Nakaoka Y, Matsumoto K, Hosoi Y, et al. Oral melatonin supplementation improves oocyte and embryo quality in women undergoing in vitro fertilization-embryo transfer. Gynecol Endocrinol. 2014;30(5):359–62.PubMedCrossRef
45.
go back to reference Tsantarliotou MP, Attanasio L, De Rosa A, Boccia L, Pellerano G, Gasparrini B. The effect of melatonin on bovine in vitro embryo development. Ital J Anim Sci. 2007;6(SUPPL. 1):488–9.CrossRef Tsantarliotou MP, Attanasio L, De Rosa A, Boccia L, Pellerano G, Gasparrini B. The effect of melatonin on bovine in vitro embryo development. Ital J Anim Sci. 2007;6(SUPPL. 1):488–9.CrossRef
48.
go back to reference Yang M, Tao J, Chai M, Wu H, Wang J, Li G, et al. Melatonin improves the quality of inferior bovine oocytes and promoted their subsequent IVF embryo development: Mechanisms and results. Molecules. 2017;22(12):2059.PubMedPubMedCentralCrossRef Yang M, Tao J, Chai M, Wu H, Wang J, Li G, et al. Melatonin improves the quality of inferior bovine oocytes and promoted their subsequent IVF embryo development: Mechanisms and results. Molecules. 2017;22(12):2059.PubMedPubMedCentralCrossRef
49.
go back to reference El-Raey M, Geshi M, Somfai T, Kaneda M, Hirako M, Abdel-Ghaffar AE, et al. Evidence of melatonin synthesis in the cumulus oocyte complexes and its role in enhancing oocyte maturation in vitro in cattle. Mol Reprod Dev. 2011;78(4):250–62.PubMedCrossRef El-Raey M, Geshi M, Somfai T, Kaneda M, Hirako M, Abdel-Ghaffar AE, et al. Evidence of melatonin synthesis in the cumulus oocyte complexes and its role in enhancing oocyte maturation in vitro in cattle. Mol Reprod Dev. 2011;78(4):250–62.PubMedCrossRef
50.
go back to reference Keshavarzi S, Salehi M, Farifteh-Nobijari F, Hosseini T, Hosseini S, Ghazifard A, et al. Melatonin modifies histone acetylation during in vitro maturation of mouse oocytes. Cell J. 2018;20(2):244–9.PubMedPubMedCentral Keshavarzi S, Salehi M, Farifteh-Nobijari F, Hosseini T, Hosseini S, Ghazifard A, et al. Melatonin modifies histone acetylation during in vitro maturation of mouse oocytes. Cell J. 2018;20(2):244–9.PubMedPubMedCentral
57.
go back to reference Kandil OM, Aboelwafa FB, Ismail EA, Kandeel SM, Ghanem N, Gamal El-Din AEK. Effects of Ascorbic Acid on Maturation Rate, Morphology, and Gene Expression of Vitrified In Vitro Matured Dromedary Camel Oocytes. World’s Vet J. 2022;12(4):418–29.CrossRef Kandil OM, Aboelwafa FB, Ismail EA, Kandeel SM, Ghanem N, Gamal El-Din AEK. Effects of Ascorbic Acid on Maturation Rate, Morphology, and Gene Expression of Vitrified In Vitro Matured Dromedary Camel Oocytes. World’s Vet J. 2022;12(4):418–29.CrossRef
59.
go back to reference Mogas T. Update on the vitrification of bovine oocytes and in vitro-produced embryos. Reprod Fertil Dev. 2019;31(1):105–17.CrossRef Mogas T. Update on the vitrification of bovine oocytes and in vitro-produced embryos. Reprod Fertil Dev. 2019;31(1):105–17.CrossRef
65.
go back to reference Parnpai R, Liang Y, Ketudat-Cairns M, Somfai T, Nagai T. Vitrification of buffalo oocytes and embryos. Theriogenology. 2016;86(1):214–20.PubMedCrossRef Parnpai R, Liang Y, Ketudat-Cairns M, Somfai T, Nagai T. Vitrification of buffalo oocytes and embryos. Theriogenology. 2016;86(1):214–20.PubMedCrossRef
66.
go back to reference Wu Z, Pan B, Qazi IH, Yang H, Guo S, Yang J, et al. Melatonin improves in vitro development of vitrified-warmed mouse germinal vesicle oocytes potentially via modulation of spindle assembly checkpoint-related genes. Cells. 2019;8(9):1009.PubMedPubMedCentralCrossRef Wu Z, Pan B, Qazi IH, Yang H, Guo S, Yang J, et al. Melatonin improves in vitro development of vitrified-warmed mouse germinal vesicle oocytes potentially via modulation of spindle assembly checkpoint-related genes. Cells. 2019;8(9):1009.PubMedPubMedCentralCrossRef
67.
go back to reference Ganji R, Nabiuni M, Faraji R. Development of mouse preantral follicle after in vitro culture in a medium containing melatonin. Cell J. 2015;16(4):546–53.PubMedPubMedCentral Ganji R, Nabiuni M, Faraji R. Development of mouse preantral follicle after in vitro culture in a medium containing melatonin. Cell J. 2015;16(4):546–53.PubMedPubMedCentral
71.
go back to reference Yu Y, Dumollard R, Rossbach A, Lai FA, Swann K. Redistribution of mitochondria leads to bursts of ATP production during spontaneous mouse oocyte maturation. J Cell Physiol. 2010;224(3):672–80.PubMedPubMedCentralCrossRef Yu Y, Dumollard R, Rossbach A, Lai FA, Swann K. Redistribution of mitochondria leads to bursts of ATP production during spontaneous mouse oocyte maturation. J Cell Physiol. 2010;224(3):672–80.PubMedPubMedCentralCrossRef
76.
go back to reference He M, Zhang T, Yang Y, Wang C. Mechanisms of oocyte maturation and related epigenetic regulation. Front Cell Dev Biol. 2021;9(March):1–18. He M, Zhang T, Yang Y, Wang C. Mechanisms of oocyte maturation and related epigenetic regulation. Front Cell Dev Biol. 2021;9(March):1–18.
78.
go back to reference Clérico G, Taminelli G, Veronesi JC, Polola J, Pagura N, Pinto C, et al. lMitochondrial function, bastocyst development and live foals born after ICSI of immature vitrified/warmed equine oocytes matured with or without melatonin. Theriogenology. 2021;160:40–9.PubMedCrossRef Clérico G, Taminelli G, Veronesi JC, Polola J, Pagura N, Pinto C, et al. lMitochondrial function, bastocyst development and live foals born after ICSI of immature vitrified/warmed equine oocytes matured with or without melatonin. Theriogenology. 2021;160:40–9.PubMedCrossRef
Metadata
Title
Effect of melatonin on developmental competence, mitochondrial distribution, and intensity of fresh and vitrified/thawed in vitro matured buffalo oocytes
Authors
Omaima Mohamed Kandil
Samar Mahfouz Abd El Rahman
Rania S. Ali
Esraa Aly Ismail
Nehad M. Ibrahim
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Reproductive Biology and Endocrinology / Issue 1/2024
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-024-01209-7

Other articles of this Issue 1/2024

Reproductive Biology and Endocrinology 1/2024 Go to the issue