Skip to main content
Top
Published in: Journal of Assisted Reproduction and Genetics 3/2024

19-02-2024 | Embryo Biology

The role of CoQ10 in embryonic development

Authors: Xueke He, Hao Chen, Minjun Liao, Xiaomei Zhao, Dawei Zhang, Miao Jiang, Zhisheng Jiang

Published in: Journal of Assisted Reproduction and Genetics | Issue 3/2024

Login to get access

Abstract

Coenzyme Q10 (CoQ10) is a natural component widely present in the inner membrane of mitochondria. CoQ10 functions as a key cofactor for adenosine triphosphate (ATP) production and exhibits antioxidant properties in vivo. Mitochondria, as the energy supply center of cells, play a crucial role in germ cell maturation and embryonic development, a complicated process of cell division and cellular differentiation that transforms from a single cell (zygote) to a multicellular organism (fetus). Here, we discuss the effects of CoQ10 on oocyte maturation and the important role of CoQ10 in the growth of various organs during different stages of fetal development. These allowed us to gain a deeper understanding of the pathophysiology of embryonic development and the potential role of CoQ10 in improving fertility quality. They also provide a reference for further developing its application in clinical treatments.
Literature
3.
go back to reference Van den Veyver IB, Al-Hussaini TK. Biparental hydatidiform moles: a maternal effect mutation affecting imprinting in the offspring. Hum Reprod Update. 2006;12(3):233–42.PubMedCrossRef Van den Veyver IB, Al-Hussaini TK. Biparental hydatidiform moles: a maternal effect mutation affecting imprinting in the offspring. Hum Reprod Update. 2006;12(3):233–42.PubMedCrossRef
4.
go back to reference Zhao J, et al. Metabolic remodelling during early mouse embryo development. Nat Metab. 2021;3(10):1372–84.PubMedCrossRef Zhao J, et al. Metabolic remodelling during early mouse embryo development. Nat Metab. 2021;3(10):1372–84.PubMedCrossRef
5.
go back to reference Wang M, et al. Autophagy: a multifaceted player in the fate of sperm. Hum Reprod Update. 2022;28(2):200–31.PubMedCrossRef Wang M, et al. Autophagy: a multifaceted player in the fate of sperm. Hum Reprod Update. 2022;28(2):200–31.PubMedCrossRef
6.
go back to reference Gott AL, et al. Non-invasive measurement of pyruvate and glucose uptake and lactate production by single human preimplantation embryos. Hum Reprod. 1990;5(1):104–8.PubMedCrossRef Gott AL, et al. Non-invasive measurement of pyruvate and glucose uptake and lactate production by single human preimplantation embryos. Hum Reprod. 1990;5(1):104–8.PubMedCrossRef
7.
9.
go back to reference Wu YD Yang, and GY Chen. Targeted disruption of Rab1a causes early embryonic lethality. Int J Mol Med. 2022;49(4):46. Wu YD Yang, and GY Chen. Targeted disruption of Rab1a causes early embryonic lethality. Int J Mol Med. 2022;49(4):46.
10.
go back to reference Drovandi S, et al. Variation of the clinical spectrum and genotype-phenotype associations in coenzyme Q10 deficiency associated glomerulopathy. Kidney Int. 2022;102(3):592–603.PubMedCrossRef Drovandi S, et al. Variation of the clinical spectrum and genotype-phenotype associations in coenzyme Q10 deficiency associated glomerulopathy. Kidney Int. 2022;102(3):592–603.PubMedCrossRef
11.
go back to reference Griffiths KK, et al. Inefficient thermogenic mitochondrial respiration due to futile proton leak in a mouse model of fragile X syndrome. FASEB J. 2020;34(6):7404–26.PubMedCrossRef Griffiths KK, et al. Inefficient thermogenic mitochondrial respiration due to futile proton leak in a mouse model of fragile X syndrome. FASEB J. 2020;34(6):7404–26.PubMedCrossRef
12.
go back to reference Ogasahara S, et al. Treatment of Kearns-Sayre syndrome with coenzyme Q10. Neurology. 1986;36(1):45–53.PubMedCrossRef Ogasahara S, et al. Treatment of Kearns-Sayre syndrome with coenzyme Q10. Neurology. 1986;36(1):45–53.PubMedCrossRef
13.
go back to reference Pallotti F et al. The roles of coenzyme Q in disease: direct and indirect involvement in cellular functions. Int J Mol Sci. 2021;23(1):128. Pallotti F et al. The roles of coenzyme Q in disease: direct and indirect involvement in cellular functions. Int J Mol Sci. 2021;23(1):128.
14.
go back to reference Gutierrez-Mariscal FM et al. Coenzyme Q(10) Supplementation for the reduction of oxidative stress: clinical implications in the treatment of chronic diseases. Int J Mol Sci. 2020;21(21):7870. Gutierrez-Mariscal FM et al. Coenzyme Q(10) Supplementation for the reduction of oxidative stress: clinical implications in the treatment of chronic diseases. Int J Mol Sci. 2020;21(21):7870.
16.
go back to reference Lapuente-Brun E, et al. Supercomplex assembly determines electron flux in the mitochondrial electron transport chain. Science. 2013;340(6140):1567–70.ADSPubMedCrossRef Lapuente-Brun E, et al. Supercomplex assembly determines electron flux in the mitochondrial electron transport chain. Science. 2013;340(6140):1567–70.ADSPubMedCrossRef
18.
go back to reference Alcazar-Fabra M, et al. Primary coenzyme Q deficiencies: a literature review and online platform of clinical features to uncover genotype-phenotype correlations. Free Radic Biol Med. 2021;167:141–80.PubMedCrossRef Alcazar-Fabra M, et al. Primary coenzyme Q deficiencies: a literature review and online platform of clinical features to uncover genotype-phenotype correlations. Free Radic Biol Med. 2021;167:141–80.PubMedCrossRef
19.
go back to reference Zhao M, et al. L-shaped association between dietary coenzyme Q10 intake and high-sensitivity C-reactive protein in Chinese adults: a national cross-sectional study. Food Funct. 2023;14(21):9815–24.PubMedCrossRef Zhao M, et al. L-shaped association between dietary coenzyme Q10 intake and high-sensitivity C-reactive protein in Chinese adults: a national cross-sectional study. Food Funct. 2023;14(21):9815–24.PubMedCrossRef
20.
go back to reference Paredes-Fuentes AJ et al. Coenzyme Q(10) Treatment monitoring in different human biological samples. Antioxidants (Basel). 2020;9(10):979. Paredes-Fuentes AJ et al. Coenzyme Q(10) Treatment monitoring in different human biological samples. Antioxidants (Basel). 2020;9(10):979.
21.
go back to reference Griffiths KKA Wang, and RJ Levy. Assessment of open probability of the mitochondrial permeability transition pore in the setting of coenzyme Q excess. J Vis Exp. 2022(184):10.3791/63646. Griffiths KKA Wang, and RJ Levy. Assessment of open probability of the mitochondrial permeability transition pore in the setting of coenzyme Q excess. J Vis Exp. 2022(184):10.3791/63646.
22.
go back to reference Barajas M, et al. The newborn Fmr1 knockout mouse: a novel model of excess ubiquinone and closed mitochondrial permeability transition pore in the developing heart. Pediatr Res. 2021;89(3):456–63.PubMedCrossRef Barajas M, et al. The newborn Fmr1 knockout mouse: a novel model of excess ubiquinone and closed mitochondrial permeability transition pore in the developing heart. Pediatr Res. 2021;89(3):456–63.PubMedCrossRef
23.
go back to reference Chazaud C, et al. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2-MAPK pathway. Dev Cell. 2006;10(5):615–24.PubMedCrossRef Chazaud C, et al. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2-MAPK pathway. Dev Cell. 2006;10(5):615–24.PubMedCrossRef
25.
go back to reference Gardner RL, Rossant J. Investigation of the fate of 4–5 day post-coitum mouse inner cell mass cells by blastocyst injection. J Embryol Exp Morphol. 1979;52:141–52.PubMed Gardner RL, Rossant J. Investigation of the fate of 4–5 day post-coitum mouse inner cell mass cells by blastocyst injection. J Embryol Exp Morphol. 1979;52:141–52.PubMed
26.
go back to reference Lawson KA and RA Pedersen. Clonal analysis of cell fate during gastrulation and early neurulation in the mouse. Ciba Found Symp, 1992;165:3–21. Lawson KA and RA Pedersen. Clonal analysis of cell fate during gastrulation and early neurulation in the mouse. Ciba Found Symp, 1992;165:3–21.
27.
28.
go back to reference Tam PP, Tan SS. The somitogenetic potential of cells in the primitive streak and the tail bud of the organogenesis-stage mouse embryo. Development. 1992;115(3):703–15.PubMedCrossRef Tam PP, Tan SS. The somitogenetic potential of cells in the primitive streak and the tail bud of the organogenesis-stage mouse embryo. Development. 1992;115(3):703–15.PubMedCrossRef
29.
go back to reference Wardle FC. Mesoderm differentiation in vertebrate development and regenerative medicine. Semin Cell Dev Biol. 2022;127:1–2.PubMedCrossRef Wardle FC. Mesoderm differentiation in vertebrate development and regenerative medicine. Semin Cell Dev Biol. 2022;127:1–2.PubMedCrossRef
30.
go back to reference Adhikari D, et al. Oocyte mitochondria-key regulators of oocyte function and potential therapeutic targets for improving fertility. Biol Reprod. 2022;106(2):366–77.PubMedCrossRef Adhikari D, et al. Oocyte mitochondria-key regulators of oocyte function and potential therapeutic targets for improving fertility. Biol Reprod. 2022;106(2):366–77.PubMedCrossRef
31.
go back to reference Santos TA, El Shourbagy S, St John JC. Mitochondrial content reflects oocyte variability and fertilization outcome. Fertil Steril. 2006;85(3):584–91. Santos TA, El Shourbagy S, St John JC. Mitochondrial content reflects oocyte variability and fertilization outcome. Fertil Steril. 2006;85(3):584–91.
32.
go back to reference Van Blerkom J. Mitochondria in human oogenesis and preimplantation embryogenesis: engines of metabolism, ionic regulation and developmental competence. Reproduction. 2004;128(3):269–80.PubMedCrossRef Van Blerkom J. Mitochondria in human oogenesis and preimplantation embryogenesis: engines of metabolism, ionic regulation and developmental competence. Reproduction. 2004;128(3):269–80.PubMedCrossRef
33.
go back to reference Tarazona AM, et al. Mitochondrial activity, distribution and segregation in bovine oocytes and in embryos produced in vitro. Reprod Domest Anim. 2006;41(1):5–11.PubMedCrossRef Tarazona AM, et al. Mitochondrial activity, distribution and segregation in bovine oocytes and in embryos produced in vitro. Reprod Domest Anim. 2006;41(1):5–11.PubMedCrossRef
34.
go back to reference Czernik M, et al. Author correction: mitochondrial function and intracellular distribution is severely affected in in vitro cultured mouse embryos. Sci Rep. 2022;12(1):21276.ADSPubMedPubMedCentralCrossRef Czernik M, et al. Author correction: mitochondrial function and intracellular distribution is severely affected in in vitro cultured mouse embryos. Sci Rep. 2022;12(1):21276.ADSPubMedPubMedCentralCrossRef
35.
go back to reference Marchante M, et al. Deciphering reproductive aging in women using a NOD/SCID mouse model for distinct physiological ovarian phenotypes. Aging (Albany NY). 2023;15(20):10856–74.PubMed Marchante M, et al. Deciphering reproductive aging in women using a NOD/SCID mouse model for distinct physiological ovarian phenotypes. Aging (Albany NY). 2023;15(20):10856–74.PubMed
36.
go back to reference He J, et al. Theaflavin 3, 3’-digallate delays ovarian aging by improving oocyte quality and regulating granulosa cell function. Oxid Med Cell Longev. 2021;2021:7064179.PubMedPubMedCentralCrossRef He J, et al. Theaflavin 3, 3’-digallate delays ovarian aging by improving oocyte quality and regulating granulosa cell function. Oxid Med Cell Longev. 2021;2021:7064179.PubMedPubMedCentralCrossRef
37.
go back to reference Qin X, et al. TrkB agonist antibody ameliorates fertility deficits in aged and cyclophosphamide-induced premature ovarian failure model mice. Nat Commun. 2022;13(1):914.ADSPubMedPubMedCentralCrossRef Qin X, et al. TrkB agonist antibody ameliorates fertility deficits in aged and cyclophosphamide-induced premature ovarian failure model mice. Nat Commun. 2022;13(1):914.ADSPubMedPubMedCentralCrossRef
38.
39.
go back to reference Jiang Z, Shen H. Mitochondria: emerging therapeutic strategies for oocyte rescue. Reprod Sci. 2022;29(3):711–22.PubMedCrossRef Jiang Z, Shen H. Mitochondria: emerging therapeutic strategies for oocyte rescue. Reprod Sci. 2022;29(3):711–22.PubMedCrossRef
41.
go back to reference Zhang H, et al. Melatonin improves the quality of maternally aged oocytes by maintaining intercellular communication and antioxidant metabolite supply. Redox Biol. 2022;49:102215.PubMedCrossRef Zhang H, et al. Melatonin improves the quality of maternally aged oocytes by maintaining intercellular communication and antioxidant metabolite supply. Redox Biol. 2022;49:102215.PubMedCrossRef
42.
go back to reference Liu J, et al. Transcriptomic responses of porcine cumulus cells to heat exposure during oocytes in vitro maturation. Mol Reprod Dev. 2021;88(1):43–54.PubMedCrossRef Liu J, et al. Transcriptomic responses of porcine cumulus cells to heat exposure during oocytes in vitro maturation. Mol Reprod Dev. 2021;88(1):43–54.PubMedCrossRef
43.
go back to reference Hu Y, et al. Transcriptomic profiles reveal the characteristics of oocytes and cumulus cells at GV, MI, and MII in follicles before ovulation. J Ovarian Res. 2023;16(1):225.PubMedPubMedCentralCrossRef Hu Y, et al. Transcriptomic profiles reveal the characteristics of oocytes and cumulus cells at GV, MI, and MII in follicles before ovulation. J Ovarian Res. 2023;16(1):225.PubMedPubMedCentralCrossRef
44.
go back to reference Babayev E, Duncan FE. Age-associated changes in cumulus cells and follicular fluid: the local oocyte microenvironment as a determinant of gamete quality. Biol Reprod. 2022;106(2):351–65.PubMedPubMedCentralCrossRef Babayev E, Duncan FE. Age-associated changes in cumulus cells and follicular fluid: the local oocyte microenvironment as a determinant of gamete quality. Biol Reprod. 2022;106(2):351–65.PubMedPubMedCentralCrossRef
45.
go back to reference Da Luz CM, et al. Altered transcriptome in cumulus cells of infertile women with advanced endometriosis with and without endometrioma. Reprod Biomed Online. 2021;42(5):952–62.PubMedCrossRef Da Luz CM, et al. Altered transcriptome in cumulus cells of infertile women with advanced endometriosis with and without endometrioma. Reprod Biomed Online. 2021;42(5):952–62.PubMedCrossRef
46.
go back to reference Ma Y, et al. Corrigendum: arachidonic acid in follicular fluid of PCOS induces oxidative stress in a human ovarian granulosa tumor cell line (KGN) and upregulates GDF15 expression as a response. Front Endocrinol (Lausanne). 2022;13:988767.PubMedCrossRef Ma Y, et al. Corrigendum: arachidonic acid in follicular fluid of PCOS induces oxidative stress in a human ovarian granulosa tumor cell line (KGN) and upregulates GDF15 expression as a response. Front Endocrinol (Lausanne). 2022;13:988767.PubMedCrossRef
47.
go back to reference Jochems R, et al. Follicular fluid steroid hormones and in vitro embryo development in Duroc and Landrace pigs. Theriogenology. 2022;190:15–21.PubMedCrossRef Jochems R, et al. Follicular fluid steroid hormones and in vitro embryo development in Duroc and Landrace pigs. Theriogenology. 2022;190:15–21.PubMedCrossRef
48.
go back to reference Yu L, et al. Follicular fluid steroid and gonadotropic hormone levels and mitochondrial function from exosomes predict embryonic development. Front Endocrinol (Lausanne). 2022;13:1025523.PubMedCrossRef Yu L, et al. Follicular fluid steroid and gonadotropic hormone levels and mitochondrial function from exosomes predict embryonic development. Front Endocrinol (Lausanne). 2022;13:1025523.PubMedCrossRef
49.
go back to reference Krawczyk K, et al. Persistent organic pollutants affect steroidogenic and apoptotic activities in granulosa cells and reactive oxygen species concentrations in oocytes in the mouse. Reprod Fertil Dev. 2023;35(3):294–305.PubMedCrossRef Krawczyk K, et al. Persistent organic pollutants affect steroidogenic and apoptotic activities in granulosa cells and reactive oxygen species concentrations in oocytes in the mouse. Reprod Fertil Dev. 2023;35(3):294–305.PubMedCrossRef
50.
go back to reference Barcelos IP, Haas RH. CoQ10 and aging. Biology (Basel). 2019;8(2):28. Barcelos IP, Haas RH. CoQ10 and aging. Biology (Basel). 2019;8(2):28.
51.
go back to reference Yang CX, et al. CoQ10 improves meiotic maturation of pig oocytes through enhancing mitochondrial function and suppressing oxidative stress. Theriogenology. 2021;159:77–86.PubMedCrossRef Yang CX, et al. CoQ10 improves meiotic maturation of pig oocytes through enhancing mitochondrial function and suppressing oxidative stress. Theriogenology. 2021;159:77–86.PubMedCrossRef
52.
go back to reference Giannubilo SR et al. CoQ10 supplementation in patients undergoing IVF-ET: the relationship with follicular fluid content and oocyte maturity. Antioxidants (Basel). 2018;7(10):141. Giannubilo SR et al. CoQ10 supplementation in patients undergoing IVF-ET: the relationship with follicular fluid content and oocyte maturity. Antioxidants (Basel). 2018;7(10):141.
53.
go back to reference Niu YJ, et al. Ubiquinol-10 delays postovulatory oocyte aging by improving mitochondrial renewal in pigs. Aging (Albany NY). 2020;12(2):1256–71.PubMedCrossRef Niu YJ, et al. Ubiquinol-10 delays postovulatory oocyte aging by improving mitochondrial renewal in pigs. Aging (Albany NY). 2020;12(2):1256–71.PubMedCrossRef
54.
go back to reference Brown AM, McCarthy HE. The effect of CoQ10 supplementation on ART treatment and oocyte quality in older women. Hum Fertil (Camb). 2023;26(6):1544–52. Brown AM, McCarthy HE. The effect of CoQ10 supplementation on ART treatment and oocyte quality in older women. Hum Fertil (Camb). 2023;26(6):1544–52.
55.
go back to reference Yang J, et al. Human follicular fluid shows diverse metabolic profiles at different follicle developmental stages. Reprod Biol Endocrinol. 2020;18(1):74.PubMedPubMedCentralCrossRef Yang J, et al. Human follicular fluid shows diverse metabolic profiles at different follicle developmental stages. Reprod Biol Endocrinol. 2020;18(1):74.PubMedPubMedCentralCrossRef
56.
go back to reference Lee CH, et al. Coenzyme Q10 ameliorates the quality of mouse oocytes during in vitro culture. Zygote. 2022;30(2):249–57.PubMedCrossRef Lee CH, et al. Coenzyme Q10 ameliorates the quality of mouse oocytes during in vitro culture. Zygote. 2022;30(2):249–57.PubMedCrossRef
57.
go back to reference Yang L, et al. Systematic understanding of anti-aging effect of coenzyme Q10 on oocyte through a network pharmacology approach. Front Endocrinol (Lausanne). 2022;13:813772.PubMedCrossRef Yang L, et al. Systematic understanding of anti-aging effect of coenzyme Q10 on oocyte through a network pharmacology approach. Front Endocrinol (Lausanne). 2022;13:813772.PubMedCrossRef
58.
go back to reference Heydarnejad A, et al. Supplementation of maturation medium with CoQ10 enhances developmental competence of ovine oocytes through improvement of mitochondrial function. Mol Reprod Dev. 2019;86(7):812–24.PubMedCrossRef Heydarnejad A, et al. Supplementation of maturation medium with CoQ10 enhances developmental competence of ovine oocytes through improvement of mitochondrial function. Mol Reprod Dev. 2019;86(7):812–24.PubMedCrossRef
59.
go back to reference Ruiz-Conca M, et al. Apoptosis and glucocorticoid-related genes mRNA expression is modulated by coenzyme Q10 supplementation during in vitro maturation and vitrification of bovine oocytes and cumulus cells. Theriogenology. 2022;192:62–72.PubMedCrossRef Ruiz-Conca M, et al. Apoptosis and glucocorticoid-related genes mRNA expression is modulated by coenzyme Q10 supplementation during in vitro maturation and vitrification of bovine oocytes and cumulus cells. Theriogenology. 2022;192:62–72.PubMedCrossRef
60.
go back to reference Gendelman M, Roth Z. Incorporation of coenzyme Q10 into bovine oocytes improves mitochondrial features and alleviates the effects of summer thermal stress on developmental competence. Biol Reprod. 2012;87(5):118.PubMedCrossRef Gendelman M, Roth Z. Incorporation of coenzyme Q10 into bovine oocytes improves mitochondrial features and alleviates the effects of summer thermal stress on developmental competence. Biol Reprod. 2012;87(5):118.PubMedCrossRef
61.
go back to reference Trapphoff T, et al. Postovulatory aging affects dynamics of mRNA, expression and localization of maternal effect proteins, spindle integrity and pericentromeric proteins in mouse oocytes. Hum Reprod. 2016;31(1):133–49.PubMedCrossRef Trapphoff T, et al. Postovulatory aging affects dynamics of mRNA, expression and localization of maternal effect proteins, spindle integrity and pericentromeric proteins in mouse oocytes. Hum Reprod. 2016;31(1):133–49.PubMedCrossRef
62.
go back to reference Miao Y, et al. Nicotinamide mononucleotide supplementation reverses the declining quality of maternally aged oocytes. Cell Rep. 2020;32(5):107987.PubMedCrossRef Miao Y, et al. Nicotinamide mononucleotide supplementation reverses the declining quality of maternally aged oocytes. Cell Rep. 2020;32(5):107987.PubMedCrossRef
63.
go back to reference Zielinska AP et al. Meiotic kinetochores fragment into multiple lobes upon cohesin loss in aging eggs. Curr Biol. 2019;29(22):3749–3765. Zielinska AP et al. Meiotic kinetochores fragment into multiple lobes upon cohesin loss in aging eggs. Curr Biol. 2019;29(22):3749–3765.
64.
go back to reference Miao Y, et al. Postovulatory aging causes the deterioration of porcine oocytes via induction of oxidative stress. FASEB J. 2018;32(3):1328–37.PubMedCrossRef Miao Y, et al. Postovulatory aging causes the deterioration of porcine oocytes via induction of oxidative stress. FASEB J. 2018;32(3):1328–37.PubMedCrossRef
65.
go back to reference Zhang M, et al. Coenzyme Q10 ameliorates the quality of postovulatory aged oocytes by suppressing DNA damage and apoptosis. Free Radic Biol Med. 2019;143:84–94.PubMedCrossRef Zhang M, et al. Coenzyme Q10 ameliorates the quality of postovulatory aged oocytes by suppressing DNA damage and apoptosis. Free Radic Biol Med. 2019;143:84–94.PubMedCrossRef
66.
go back to reference Jeong SM, et al. SIRT4 has tumor-suppressive activity and regulates the cellular metabolic response to DNA damage by inhibiting mitochondrial glutamine metabolism. Cancer Cell. 2013;23(4):450–63.PubMedPubMedCentralCrossRef Jeong SM, et al. SIRT4 has tumor-suppressive activity and regulates the cellular metabolic response to DNA damage by inhibiting mitochondrial glutamine metabolism. Cancer Cell. 2013;23(4):450–63.PubMedPubMedCentralCrossRef
67.
68.
go back to reference Xing X, et al. Coenzyme Q10 supplement rescues postovulatory oocyte aging by regulating SIRT4 expression. Curr Mol Pharmacol. 2022;15(1):190–203.PubMed Xing X, et al. Coenzyme Q10 supplement rescues postovulatory oocyte aging by regulating SIRT4 expression. Curr Mol Pharmacol. 2022;15(1):190–203.PubMed
69.
70.
go back to reference McReynolds S et al. Impact of maternal aging on the molecular signature of human cumulus cells. Fertil Steril. 2012;98(6):1574–80. McReynolds S et al. Impact of maternal aging on the molecular signature of human cumulus cells. Fertil Steril. 2012;98(6):1574–80.
71.
go back to reference Ben-Meir A et al. Co-enzyme Q10 supplementation rescues cumulus cells dysfunction in a maternal aging model. Antioxidants (Basel). 2019;8(3):58. Ben-Meir A et al. Co-enzyme Q10 supplementation rescues cumulus cells dysfunction in a maternal aging model. Antioxidants (Basel). 2019;8(3):58.
72.
go back to reference Agarwal A, et al. Male oxidative stress infertility (MOSI): proposed terminology and clinical practice guidelines for management of idiopathic male infertility. World J Mens Health. 2019;37(3):296–312.PubMedPubMedCentralCrossRef Agarwal A, et al. Male oxidative stress infertility (MOSI): proposed terminology and clinical practice guidelines for management of idiopathic male infertility. World J Mens Health. 2019;37(3):296–312.PubMedPubMedCentralCrossRef
74.
go back to reference Tourzani DA et al. Caput ligation renders immature mouse sperm motile and capable to undergo cAMP-dependent phosphorylation. Int J Mol Sci. 2021;22(19):10241. Tourzani DA et al. Caput ligation renders immature mouse sperm motile and capable to undergo cAMP-dependent phosphorylation. Int J Mol Sci. 2021;22(19):10241.
75.
go back to reference Moustakli E et al. Sperm mitochondrial content and mitochondrial DNA to nuclear DNA ratio are associated with body mass index and progressive motility. Biomedicines. 2023;11(11):3014. Moustakli E et al. Sperm mitochondrial content and mitochondrial DNA to nuclear DNA ratio are associated with body mass index and progressive motility. Biomedicines. 2023;11(11):3014.
76.
go back to reference Park YJ, et al. Low sperm motility is determined by abnormal protein modification during epididymal maturation. World J Mens Health. 2022;40(3):526–35.PubMedPubMedCentralCrossRef Park YJ, et al. Low sperm motility is determined by abnormal protein modification during epididymal maturation. World J Mens Health. 2022;40(3):526–35.PubMedPubMedCentralCrossRef
77.
go back to reference Park YJ, Pang MG. Mitochondrial functionality in male fertility: from spermatogenesis to fertilization. Antioxidants (Basel). 2021;10(1):98. Park YJ, Pang MG. Mitochondrial functionality in male fertility: from spermatogenesis to fertilization. Antioxidants (Basel). 2021;10(1):98.
78.
go back to reference Chen X, et al. Identification of differentially expressed proteins between bull X and Y spermatozoa. J Proteomics. 2012;77:59–67.PubMedCrossRef Chen X, et al. Identification of differentially expressed proteins between bull X and Y spermatozoa. J Proteomics. 2012;77:59–67.PubMedCrossRef
79.
go back to reference Sengupta P, et al. Oxidative stress and idiopathic male infertility. Adv Exp Med Biol. 2022;1358:181–204.PubMedCrossRef Sengupta P, et al. Oxidative stress and idiopathic male infertility. Adv Exp Med Biol. 2022;1358:181–204.PubMedCrossRef
81.
go back to reference Liu KS, et al. Effect and mechanisms of reproductive tract infection on oxidative stress parameters, sperm DNA fragmentation, and semen quality in infertile males. Reprod Biol Endocrinol. 2021;19(1):97.PubMedPubMedCentralCrossRef Liu KS, et al. Effect and mechanisms of reproductive tract infection on oxidative stress parameters, sperm DNA fragmentation, and semen quality in infertile males. Reprod Biol Endocrinol. 2021;19(1):97.PubMedPubMedCentralCrossRef
82.
go back to reference Oseguera-Lopez I et al. Perfluorooctane sulfonate (PFOS) and perfluorohexane sulfonate (PFHxS) alters protein phosphorylation, increase ROS levels and DNA fragmentation during in vitro capacitation of boar spermatozoa. Animals (Basel). 2020;10(10):1934. Oseguera-Lopez I et al. Perfluorooctane sulfonate (PFOS) and perfluorohexane sulfonate (PFHxS) alters protein phosphorylation, increase ROS levels and DNA fragmentation during in vitro capacitation of boar spermatozoa. Animals (Basel). 2020;10(10):1934.
83.
go back to reference Li KP, Yang XS, Wu T. The effect of antioxidants on sperm quality parameters and pregnancy rates for idiopathic male infertility: a network meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne). 2022;13:810242.PubMedCrossRef Li KP, Yang XS, Wu T. The effect of antioxidants on sperm quality parameters and pregnancy rates for idiopathic male infertility: a network meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne). 2022;13:810242.PubMedCrossRef
84.
go back to reference Alahmar AT, et al. Coenzyme Q10 improves sperm parameters, oxidative stress markers and sperm DNA fragmentation in infertile patients with idiopathic oligoasthenozoospermia. World J Mens Health. 2021;39(2):346–51.PubMedCrossRef Alahmar AT, et al. Coenzyme Q10 improves sperm parameters, oxidative stress markers and sperm DNA fragmentation in infertile patients with idiopathic oligoasthenozoospermia. World J Mens Health. 2021;39(2):346–51.PubMedCrossRef
85.
go back to reference Balercia G, et al. Coenzyme Q10 treatment in infertile men with idiopathic asthenozoospermia: a placebo-controlled, double-blind randomized trial. Fertil Steril. 2009;91(5):1785–92.PubMedCrossRef Balercia G, et al. Coenzyme Q10 treatment in infertile men with idiopathic asthenozoospermia: a placebo-controlled, double-blind randomized trial. Fertil Steril. 2009;91(5):1785–92.PubMedCrossRef
86.
go back to reference Safarinejad MR. Efficacy of coenzyme Q10 on semen parameters, sperm function and reproductive hormones in infertile men. J Urol. 2009;182(1):237–48.PubMedCrossRef Safarinejad MR. Efficacy of coenzyme Q10 on semen parameters, sperm function and reproductive hormones in infertile men. J Urol. 2009;182(1):237–48.PubMedCrossRef
87.
go back to reference Bellusci M, et al. Distal phalangeal erythema in an infant with biallelic PDSS1 mutations: expanding the phenotype of primary coenzyme Q(10) deficiency. JIMD Rep. 2021;62(1):3–5.PubMedPubMedCentralCrossRef Bellusci M, et al. Distal phalangeal erythema in an infant with biallelic PDSS1 mutations: expanding the phenotype of primary coenzyme Q(10) deficiency. JIMD Rep. 2021;62(1):3–5.PubMedPubMedCentralCrossRef
89.
go back to reference Laugwitz L, et al. Human COQ4 deficiency: delineating the clinical, metabolic and neuroimaging phenotypes. J Med Genet. 2022;59(9):878–87.PubMedCrossRef Laugwitz L, et al. Human COQ4 deficiency: delineating the clinical, metabolic and neuroimaging phenotypes. J Med Genet. 2022;59(9):878–87.PubMedCrossRef
90.
go back to reference Wang N, et al. A family segregating lethal primary coenzyme Q10 deficiency due to two novel COQ6 variants. Front Genet. 2021;12: 811833.PubMedCrossRef Wang N, et al. A family segregating lethal primary coenzyme Q10 deficiency due to two novel COQ6 variants. Front Genet. 2021;12: 811833.PubMedCrossRef
91.
go back to reference Olgac A, et al. A rare case of primary coenzyme Q10 deficiency due to COQ9 mutation. J Pediatr Endocrinol Metab. 2020;33(1):165–70.PubMedCrossRef Olgac A, et al. A rare case of primary coenzyme Q10 deficiency due to COQ9 mutation. J Pediatr Endocrinol Metab. 2020;33(1):165–70.PubMedCrossRef
92.
go back to reference Chambers BE, NE Weaver, Wingert RA. The "3Ds" of growing kidney organoids: advances in nephron development, disease modeling, and drug screening. Cells. 2023;12(4):549. Chambers BE, NE Weaver, Wingert RA. The "3Ds" of growing kidney organoids: advances in nephron development, disease modeling, and drug screening. Cells. 2023;12(4):549.
93.
go back to reference Zhai SB, et al. Early-onset COQ8B (ADCK4) glomerulopathy in a child with isolated proteinuria: a case report and literature review. BMC Nephrol. 2020;21(1):406.PubMedPubMedCentralCrossRef Zhai SB, et al. Early-onset COQ8B (ADCK4) glomerulopathy in a child with isolated proteinuria: a case report and literature review. BMC Nephrol. 2020;21(1):406.PubMedPubMedCentralCrossRef
94.
go back to reference Stanczyk M, et al. CoQ10-related sustained remission of proteinuria in a child with COQ6 glomerulopathy-a case report. Pediatr Nephrol. 2018;33(12):2383–7.PubMedPubMedCentralCrossRef Stanczyk M, et al. CoQ10-related sustained remission of proteinuria in a child with COQ6 glomerulopathy-a case report. Pediatr Nephrol. 2018;33(12):2383–7.PubMedPubMedCentralCrossRef
95.
96.
go back to reference Muigg V, et al. Delayed cerebellar ataxia, a rare post-malaria neurological complication: Case report and review of the literature. Travel Med Infect Dis. 2021;44: 102177.PubMedCrossRef Muigg V, et al. Delayed cerebellar ataxia, a rare post-malaria neurological complication: Case report and review of the literature. Travel Med Infect Dis. 2021;44: 102177.PubMedCrossRef
97.
go back to reference Musumeci O, et al. Familial cerebellar ataxia with muscle coenzyme Q10 deficiency. Neurology. 2001;56(7):849–55.PubMedCrossRef Musumeci O, et al. Familial cerebellar ataxia with muscle coenzyme Q10 deficiency. Neurology. 2001;56(7):849–55.PubMedCrossRef
98.
go back to reference Monfrini E, et al. Whole-exome sequencing study of fibroblasts derived from patients with cerebellar ataxia referred to investigate CoQ10 deficiency. Neurol Genet. 2023;9(2):e200058.PubMedPubMedCentralCrossRef Monfrini E, et al. Whole-exome sequencing study of fibroblasts derived from patients with cerebellar ataxia referred to investigate CoQ10 deficiency. Neurol Genet. 2023;9(2):e200058.PubMedPubMedCentralCrossRef
99.
go back to reference Rius R, et al. Biallelic pathogenic variants in COX11 are associated with an infantile-onset mitochondrial encephalopathy. Hum Mutat. 2022;43(12):1970–8.PubMedPubMedCentralCrossRef Rius R, et al. Biallelic pathogenic variants in COX11 are associated with an infantile-onset mitochondrial encephalopathy. Hum Mutat. 2022;43(12):1970–8.PubMedPubMedCentralCrossRef
100.
102.
go back to reference Martinez PA, et al. Smad2/3-pathway ligand trap luspatercept enhances erythroid differentiation in murine beta-thalassaemia by increasing GATA-1 availability. J Cell Mol Med. 2020;24(11):6162–77.PubMedPubMedCentralCrossRef Martinez PA, et al. Smad2/3-pathway ligand trap luspatercept enhances erythroid differentiation in murine beta-thalassaemia by increasing GATA-1 availability. J Cell Mol Med. 2020;24(11):6162–77.PubMedPubMedCentralCrossRef
105.
go back to reference Robichaux DJ, et al. Mitochondrial permeability transition pore-dependent necrosis. J Mol Cell Cardiol. 2023;174:47–55.PubMedCrossRef Robichaux DJ, et al. Mitochondrial permeability transition pore-dependent necrosis. J Mol Cell Cardiol. 2023;174:47–55.PubMedCrossRef
106.
go back to reference Yan A, et al. Idebenone alleviates neuroinflammation and modulates microglial polarization in LPS-stimulated BV2 cells and MPTP-induced Parkinson’s disease mice. Front Cell Neurosci. 2018;12:529.PubMedCrossRef Yan A, et al. Idebenone alleviates neuroinflammation and modulates microglial polarization in LPS-stimulated BV2 cells and MPTP-induced Parkinson’s disease mice. Front Cell Neurosci. 2018;12:529.PubMedCrossRef
107.
go back to reference Takahashi M, Shimizu T, Shirasawa T. Reversal of slow growth and heartbeat through the restoration of mitochondrial function in clk-1-deficient mouse embryos by exogenous administration of coenzyme Q10. Exp Gerontol. 2012;47(6):425–31.PubMedCrossRef Takahashi M, Shimizu T, Shirasawa T. Reversal of slow growth and heartbeat through the restoration of mitochondrial function in clk-1-deficient mouse embryos by exogenous administration of coenzyme Q10. Exp Gerontol. 2012;47(6):425–31.PubMedCrossRef
108.
go back to reference Smith AC, et al. A family segregating lethal neonatal coenzyme Q(10) deficiency caused by mutations in COQ9. J Inherit Metab Dis. 2018;41(4):719–29.PubMedCrossRef Smith AC, et al. A family segregating lethal neonatal coenzyme Q(10) deficiency caused by mutations in COQ9. J Inherit Metab Dis. 2018;41(4):719–29.PubMedCrossRef
109.
go back to reference Danhauser K, et al. Fatal neonatal encephalopathy and lactic acidosis caused by a homozygous loss-of-function variant in COQ9. Eur J Hum Genet. 2016;24(3):450–4.PubMedCrossRef Danhauser K, et al. Fatal neonatal encephalopathy and lactic acidosis caused by a homozygous loss-of-function variant in COQ9. Eur J Hum Genet. 2016;24(3):450–4.PubMedCrossRef
110.
112.
go back to reference Budani MC, Tiboni GM. Effects of supplementation with natural antioxidants on oocytes and preimplantation embryos. Antioxidants (Basel). 2020;9(7):612. Budani MC, Tiboni GM. Effects of supplementation with natural antioxidants on oocytes and preimplantation embryos. Antioxidants (Basel). 2020;9(7):612.
Metadata
Title
The role of CoQ10 in embryonic development
Authors
Xueke He
Hao Chen
Minjun Liao
Xiaomei Zhao
Dawei Zhang
Miao Jiang
Zhisheng Jiang
Publication date
19-02-2024
Publisher
Springer US
Published in
Journal of Assisted Reproduction and Genetics / Issue 3/2024
Print ISSN: 1058-0468
Electronic ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-024-03052-6

Other articles of this Issue 3/2024

Journal of Assisted Reproduction and Genetics 3/2024 Go to the issue