Acessibilidade / Reportar erro

The sperm mitochondria: clues and challenges

Abstract

Sperm cells rely on different substrates to fulfil thei energy demand for different functions and diverse moments of their life. Species specific mechanism involve both energy substrate transport and their utilization: hexose transporters, a protein family of facilitative passive transporters of glucose and other hexose, have been identified in spermatozoa of different species and, within the species, their localization has been identified and, in some cases, linked to specific glycilitic enzyme presence. The catabolism of hexose sources for energy purposes has been studied in various species, and recent advances has been made in the knowledge of metabolic strategies of sperm cells. In particular, the importance of aerobic metabolism has been defined and described in horse, boar and even mouse spermatozoa; bull sperm cells demonstrate to have a good adaptability and capacity to switch between glycolysis and oxidative phosphorylation; finally, dog sperm cells have been demonstrated to have a great plasticity in energy metabolism management, being also able to activate the anabolic pathway of glycogen syntesis.

In conclusion, the study of energy management and mitochondrial function in spermatozoa of different specie furnishes important base knowledge to define new media for preservation as well as newbases for reproductive biotechnologies.

Keywords:
sperm metabolism; bioenergetics; hexose uptake

Introduction

Spermatozoa are highly specialized cells that are aimed at delivering the male DNA into the new generation subjects. To do so, after being produced by the testicle, matured and stored in the epididymis, they are released in the female genital tract, where they begin the long journey to the female gamete in order to reach it and act for the fertilization process.

In mammalian sperm, between ejaculation and fertilization, there can also be another important step in sperm life, that is not occurring in natural breeding animals, but usually occurs when artificial reproduction techniques are used: conservation.

Irrespective of the imminent fate of sperm cells, it should be stated that they need some energy substrate to adapt to the environment, maintaining homeostasis and movement.

This review aims at pointing out the most recent knowledge on sperm metabolism in terms of fuelling supply, utilization of substrates and metabolic strategies, and balance between anaerobic and aerobic pathways.

Most of the knowledge on different mammalian species has been presented and reviewed by outstanding research groups and colleagues during the last years (Boguenet et al., 2021Boguenet M, Bouet PE, Spiers A, Reynier P, May-Panloup P. Mitochondria: their role in spermatozoa and in male infertility. Hum Reprod Update. 2021;27(4):697-719. http://dx.doi.org/10.1093/humupd/dmab001. PMid:33555313.
http://dx.doi.org/10.1093/humupd/dmab001...
; Moraes and Meyers, 2018Moraes CR, Meyers S. The sperm mitochondrion: organelle of many functions. Anim Reprod Sci. 2018;194:71-80. http://dx.doi.org/10.1016/j.anireprosci.2018.03.024. PMid:29605167.
http://dx.doi.org/10.1016/j.anireprosci....
; Peña et al., 2022Peña FJ, Ortiz-Rodríguez JM, Gaitskell-Phillips GL, Gil MC, Ortega-Ferrusola C, Martín-Cano FE. An integrated overview on the regulation of sperm metabolism (glycolysis-Krebs cycle-oxidative phosphorylation). Anim Reprod Sci. 2022;246:106805. http://dx.doi.org/10.1016/j.anireprosci.2021.106805. PMid:34275685.
http://dx.doi.org/10.1016/j.anireprosci....
; Rodriguez-Gil, 2006Rodriguez-Gil J. Mammalian sperm energy resources management and survival during conservation in refrigeration. Reprod Domest Anim. 2006;41(Suppl. 2):11-20. http://dx.doi.org/10.1111/j.1439-0531.2006.00765.x. PMid:16984465.
http://dx.doi.org/10.1111/j.1439-0531.20...
; Rodríguez-Gil and Bonet, 2016Rodríguez-Gil JE, Bonet S. Current knowledge on boar sperm metabolism: comparison with other mammalian species. Theriogenology. 2016;85(1):4-11. http://dx.doi.org/10.1016/j.theriogenology.2015.05.005. PMid:26094247.
http://dx.doi.org/10.1016/j.theriogenolo...
; Varner et al., 2015Varner DD, Gibb Z, Aitken RJ. Stallion fertility: a focus on the spermatozoon. Equine Vet J. 2015;47(1):16-24. http://dx.doi.org/10.1111/evj.12308. PMid:24943233.
http://dx.doi.org/10.1111/evj.12308...
) and we also focused on some specific aspects of sperm metabolism (Bucci et al., 2011Bucci D, Rodríguez-Gil JE, Vallorani C, Spinaci M, Galeati G, Tamanini C. GLUTs and mammalian sperm metabolism. J Androl. 2011;32(4):348-55. http://dx.doi.org/10.2164/jandrol.110.011197. PMid:21088231.
http://dx.doi.org/10.2164/jandrol.110.01...
). We therefore invite the reader to refer also to those review papers to widen the knowledge on the theme. In this review, we will also present some unpublished data from our labs regarding the balance and equilibrium between anaerobic and oxidative metabolism.

Energy sources for sperm cells. Not only sugar

An interesting paper by Storey (2008)Storey BT. Mammalian sperm metabolism: oxygen and sugar, friend and foe. Int J Dev Biol. 2008;52(5-6):427-37. http://dx.doi.org/10.1387/ijdb.072522bs. PMid:18649255.
http://dx.doi.org/10.1387/ijdb.072522bs...
focused on the regulatory and fuelling role of sugars in mammalian sperm life and activity (Storey, 2008Storey BT. Mammalian sperm metabolism: oxygen and sugar, friend and foe. Int J Dev Biol. 2008;52(5-6):427-37. http://dx.doi.org/10.1387/ijdb.072522bs. PMid:18649255.
http://dx.doi.org/10.1387/ijdb.072522bs...
). The author thoroughly revised a large number of studies dating back to the 1940s, in which it was first studied and reported how important and precious sugar fuelling was for sperm function.

Of particular interest, among the first studies on sperm metabolism, were the researches reported in Storey’s review by Lardy and Philips and co-workers, as well as the impressive work published by Mann (for reviewing and references see (Storey, 2008Storey BT. Mammalian sperm metabolism: oxygen and sugar, friend and foe. Int J Dev Biol. 2008;52(5-6):427-37. http://dx.doi.org/10.1387/ijdb.072522bs. PMid:18649255.
http://dx.doi.org/10.1387/ijdb.072522bs...
)); those first experimental work were aimed at defining the role of fuelling sugars for maintaining the most evident sperm function, motility, as well as to maintain sperm fertilizing ability. From that point on, a large interest was given to sugars and their role in sperm preservation under liquid storage conditions. It is well known that different species may rely on hexoses for their metabolism (Fernández-Novell et al., 2004Fernández-Novell JM, Ballester J, Medrano A, Otaegui PJ, Rigau T, Guinovart JJ, Rodríguez-Gil JE. The presence of a high-Km hexokinase activity in dog, but not in boar, sperm. FEBS Lett. 2004;570(1-3):211-6. http://dx.doi.org/10.1016/j.febslet.2004.06.015. PMid:15251466.
http://dx.doi.org/10.1016/j.febslet.2004...
; Medrano et al., 2005Medrano A, Peña A, Rigau T, Rodrìguez-Gil JE. Variations in the proportion of glycolytic/non-glycolytic energy substrates modulate sperm membrane integrity and function in diluted boar samples stored at 15-17 degrees C. Reprod Domest Anim. 2005;40:448-53. http://dx.doi.org/10.1111/j.1439-0531.2005.00599.x
http://dx.doi.org/10.1111/j.1439-0531.20...
; Peña et al., 2022Peña FJ, Ortiz-Rodríguez JM, Gaitskell-Phillips GL, Gil MC, Ortega-Ferrusola C, Martín-Cano FE. An integrated overview on the regulation of sperm metabolism (glycolysis-Krebs cycle-oxidative phosphorylation). Anim Reprod Sci. 2022;246:106805. http://dx.doi.org/10.1016/j.anireprosci.2021.106805. PMid:34275685.
http://dx.doi.org/10.1016/j.anireprosci....
). Nevetheless, other mechanisms of fuelling have been demonstrated over time (Brooks and Mann, 1972Brooks DE, Mann T. Relation between the oxidation state of nicotinamide-adenine dinucleotide and the metabolism of spermatozoa. Biochem J. 1972;129(5):1023-34. http://dx.doi.org/10.1042/bj1291023. PMid:4144231.
http://dx.doi.org/10.1042/bj1291023...
, 1973Brooks D, Mann T. Pyruvate metabolism in boar spermatozoa. J Reprod Fertil. 1973;34(1):105-19. http://dx.doi.org/10.1530/jrf.0.0340105. PMid:4719804.
http://dx.doi.org/10.1530/jrf.0.0340105...
; Hutson et al., 1977Hutson SM, van Dop C, Lardy HA. Mitochondrial metabolism of pyruvate in bovine spermatozoa. J Biol Chem. 1977;252(4):1309-15. http://dx.doi.org/10.1016/S0021-9258(17)40656-9. PMid:838719.
http://dx.doi.org/10.1016/S0021-9258(17)...
; Medrano et al., 2006aMedrano A, Fernández-Novell JM, Ramió L, Alvarez J, Goldberg E, Montserrat Rivera M, Guinovart JJ, Rigau T, Rodríguez-Gil JE. Utilization of citrate and lactate through a lactate dehydrogenase and ATP-regulated pathway in boar spermatozoa. Mol Reprod Dev. 2006a;73(3):369-78. http://dx.doi.org/10.1002/mrd.20414. PMid:16362974.
http://dx.doi.org/10.1002/mrd.20414...
) in different species, thus demonstrating that the direct impact of mitochondrial metabolization of energy sources could play a major role in energy obtainment from different species.

The uptake of hexose monosaccharides is exerted by different members of a protein family of carriers and these comprises the so called GLUTs (glucose transporters) which have different specificities for the substrates hexoses (Bucci et al., 2011Bucci D, Rodríguez-Gil JE, Vallorani C, Spinaci M, Galeati G, Tamanini C. GLUTs and mammalian sperm metabolism. J Androl. 2011;32(4):348-55. http://dx.doi.org/10.2164/jandrol.110.011197. PMid:21088231.
http://dx.doi.org/10.2164/jandrol.110.01...
): CLASS I transporters, (GLUT 1, 2, 3, 4, and 14) are mainly glucose transporters (excepting for GLUT2, that transport also fructose); CLASS II transporters (Glut 5, 7, 9, 11), fructose or double affinity transporters; CLASS III transporters (GLUT 6, 8, 10, 12 and HMIT), with hight affinity for glucose and a different structure if compared with CLASS I and II ones.

These proteins have been studied in sperm from different species (Angulo et al., 1998Angulo C, Rauch MC, Droppelmann A, Reyes AM, Slebe JC, Delgado-López F, Guaiquil VH, Vera JC, Concha II. Hexose transporter expression and function in mammalian spermatozoa: cellular localization and transport of hexoses and vitamin C. J Cell Biochem. 1998;71(2):189-203. http://dx.doi.org/10.1002/(SICI)1097-4644(19981101)71:2<189::AID-JCB5>3.0.CO;2-R. PMid:9779818.
http://dx.doi.org/10.1002/(SICI)1097-464...
; Bucci et al., 2010aBucci D, Isani G, Spinaci M, Tamanini C, Mari G, Zambelli D, Galeati G. Comparative immunolocalization of GLUTs 1, 2, 3 and 5 in boar, stallion and dog spermatozoa. Reprod Domest Anim. 2010a;45(2):315-22. http://dx.doi.org/10.1111/j.1439-0531.2008.01307.x. PMid:19055550.
http://dx.doi.org/10.1111/j.1439-0531.20...
, bBucci D, Spinaci M, Vallorani C, Contri A, Carluccio A, Isani G, Tamanini C, Galeati G. Detection and localization of GLUTs 1, 2, 3 and 5 in donkey spermatozoa. Reprod Domest Anim. 2010b;45(5):e217-20. http://dx.doi.org/10.1111/j.1439-0531.2009.01544.x. PMid:19930135.
http://dx.doi.org/10.1111/j.1439-0531.20...
, 2011Bucci D, Rodríguez-Gil JE, Vallorani C, Spinaci M, Galeati G, Tamanini C. GLUTs and mammalian sperm metabolism. J Androl. 2011;32(4):348-55. http://dx.doi.org/10.2164/jandrol.110.011197. PMid:21088231.
http://dx.doi.org/10.2164/jandrol.110.01...
; Sung and Moley, 2007Sung TK, Moley KH. The expression of GLUT8, GLUT9a, and GLUT9b in the mouse testis and sperm. Reprod Sci. 2007;14(5):445-55. http://dx.doi.org/10.1177/1933719107306226. PMid:17913964.
http://dx.doi.org/10.1177/19337191073062...
) such as human, rat, and bull sperm cells. GLUTs 1, 2, 3, 4, and 5 (Angulo et al., 1998Angulo C, Rauch MC, Droppelmann A, Reyes AM, Slebe JC, Delgado-López F, Guaiquil VH, Vera JC, Concha II. Hexose transporter expression and function in mammalian spermatozoa: cellular localization and transport of hexoses and vitamin C. J Cell Biochem. 1998;71(2):189-203. http://dx.doi.org/10.1002/(SICI)1097-4644(19981101)71:2<189::AID-JCB5>3.0.CO;2-R. PMid:9779818.
http://dx.doi.org/10.1002/(SICI)1097-464...
) show species specific localization within sperm head and tail, and each GLUT shows a different distribution within the same species. The immunocytochemistry results were also validated with Western Blot analysis. Our group studied GLUT 1, 2, 3, 5 in horse, donkey, boar and dog sperm cells (Bucci et al., 2010aBucci D, Isani G, Spinaci M, Tamanini C, Mari G, Zambelli D, Galeati G. Comparative immunolocalization of GLUTs 1, 2, 3 and 5 in boar, stallion and dog spermatozoa. Reprod Domest Anim. 2010a;45(2):315-22. http://dx.doi.org/10.1111/j.1439-0531.2008.01307.x. PMid:19055550.
http://dx.doi.org/10.1111/j.1439-0531.20...
), defining, with the same experimental design as Angulo, the presence and abundance of glucose transporters (immunocytochemistry and western blotting). Finally, GLUT 8 and 9 were studied in mouse testis and mature spermatozoa (Sung and Moley, 2007Sung TK, Moley KH. The expression of GLUT8, GLUT9a, and GLUT9b in the mouse testis and sperm. Reprod Sci. 2007;14(5):445-55. http://dx.doi.org/10.1177/1933719107306226. PMid:17913964.
http://dx.doi.org/10.1177/19337191073062...
)

Interesting studies have regarded the relationship between GLUTs activity and the metabolism of sperm cells: in the early 2000s, Rigau and colleagues showed that metabolic plasticity of dog spermatozoa could be related to GLUTs localization (Fernández-Novell et al., 2004Fernández-Novell JM, Ballester J, Medrano A, Otaegui PJ, Rigau T, Guinovart JJ, Rodríguez-Gil JE. The presence of a high-Km hexokinase activity in dog, but not in boar, sperm. FEBS Lett. 2004;570(1-3):211-6. http://dx.doi.org/10.1016/j.febslet.2004.06.015. PMid:15251466.
http://dx.doi.org/10.1016/j.febslet.2004...
; Rigau et al., 2001Rigau T, Farré M, Ballester J, Mogas T, Peña A, Rodríguez-Gil JE. Effects of glucose and fructose on motility patterns of dog spermatozoa from fresh ejaculates. Theriogenology. 2001;56(5):601-15. http://dx.doi.org/10.1016/S0093-691X(01)00609-4. PMid:11665883.
http://dx.doi.org/10.1016/S0093-691X(01)...
, 2002Rigau T, Rivera M, Palomo MJ, Fernández-Novell JM, Mogas T, Ballester J, Peña A, Otaegui PJ, Guinovart JJ, Rodríguez-Gil JE. Differential effects of glucose and fructose on hexose metabolism in dog spermatozoa. Reproduction. 2002;123(4):579-91. http://dx.doi.org/10.1530/rep.0.1230579. PMid:11914120.
http://dx.doi.org/10.1530/rep.0.1230579...
); similarly, it was demonstrated that GLUT 3 co-localizes with Hexokinase I in pig spermatozoa(Medrano et al., 2006bMedrano A, García-Gil N, Ramió L, Montserrat Rivera M, Fernández-Novell JM, Ramírez A, Peña A, Dolors Briz M, Pinart E, Concha II, Bonet S, Rigau T, Rodríguez-Gil JE. Hexose-specificity of hexokinase and ADP-dependence of pyruvate kinase play important roles in the control of monosaccharide utilization in freshly diluted boar spermatozoa. Mol Reprod Dev. 2006b;73(9):1179-94. http://dx.doi.org/10.1002/mrd.20480. PMid:16804879.
http://dx.doi.org/10.1002/mrd.20480...
), and this could strictly link the activity of the transporter and the enzymes responsible for the metabolization of the transported substance.

Finally, we demonstrated that GLUT 3 and 5 in dog spermatozoa (Bucci et al., 2010aBucci D, Isani G, Spinaci M, Tamanini C, Mari G, Zambelli D, Galeati G. Comparative immunolocalization of GLUTs 1, 2, 3 and 5 in boar, stallion and dog spermatozoa. Reprod Domest Anim. 2010a;45(2):315-22. http://dx.doi.org/10.1111/j.1439-0531.2008.01307.x. PMid:19055550.
http://dx.doi.org/10.1111/j.1439-0531.20...
) undergo relocalization after incubation under capacitating conditions; again, a swift in metabolic rate of the sperm cells induces a modification of the localization of the suppliers of energy substrates.

Monocarboxilate transporters (MCTs) have been recently described in spermatozoa, specifically MCT1 has been identified in the sperm head; these transporters are responsible for transport of pyruvate/lactate and their presence could play a promising role in the production of next-generation sperm preservation extenders (Peña et al., 2022Peña FJ, Ortiz-Rodríguez JM, Gaitskell-Phillips GL, Gil MC, Ortega-Ferrusola C, Martín-Cano FE. An integrated overview on the regulation of sperm metabolism (glycolysis-Krebs cycle-oxidative phosphorylation). Anim Reprod Sci. 2022;246:106805. http://dx.doi.org/10.1016/j.anireprosci.2021.106805. PMid:34275685.
http://dx.doi.org/10.1016/j.anireprosci....
).

Different strategies for energy obtainment and mitochondria

Since a long time, it was recognised that spermatozoa from different species have different metabolic strategies to obtain energy for their metabolic activity (Bucci et al., 2011Bucci D, Rodríguez-Gil JE, Vallorani C, Spinaci M, Galeati G, Tamanini C. GLUTs and mammalian sperm metabolism. J Androl. 2011;32(4):348-55. http://dx.doi.org/10.2164/jandrol.110.011197. PMid:21088231.
http://dx.doi.org/10.2164/jandrol.110.01...
; Gibb and Aitken, 2016Gibb Z, Aitken RJ. The impact of sperm metabolism during in vitro storage : the stallion as a model. BioMed Res Int. 2016;2016:9380609. http://dx.doi.org/10.1155/2016/9380609. PMid:26881234.
http://dx.doi.org/10.1155/2016/9380609...
; Peña et al., 2022Peña FJ, Ortiz-Rodríguez JM, Gaitskell-Phillips GL, Gil MC, Ortega-Ferrusola C, Martín-Cano FE. An integrated overview on the regulation of sperm metabolism (glycolysis-Krebs cycle-oxidative phosphorylation). Anim Reprod Sci. 2022;246:106805. http://dx.doi.org/10.1016/j.anireprosci.2021.106805. PMid:34275685.
http://dx.doi.org/10.1016/j.anireprosci....
; Rodriguez-Gil, 2006Rodriguez-Gil J. Mammalian sperm energy resources management and survival during conservation in refrigeration. Reprod Domest Anim. 2006;41(Suppl. 2):11-20. http://dx.doi.org/10.1111/j.1439-0531.2006.00765.x. PMid:16984465.
http://dx.doi.org/10.1111/j.1439-0531.20...
; Rodríguez-Gil and Bonet, 2016Rodríguez-Gil JE, Bonet S. Current knowledge on boar sperm metabolism: comparison with other mammalian species. Theriogenology. 2016;85(1):4-11. http://dx.doi.org/10.1016/j.theriogenology.2015.05.005. PMid:26094247.
http://dx.doi.org/10.1016/j.theriogenolo...
; Varner et al., 2015Varner DD, Gibb Z, Aitken RJ. Stallion fertility: a focus on the spermatozoon. Equine Vet J. 2015;47(1):16-24. http://dx.doi.org/10.1111/evj.12308. PMid:24943233.
http://dx.doi.org/10.1111/evj.12308...
); this section will furnish a brief description of the different energy obtainment strategies exerted by different species to sustain sperm function.

It is noteworthy to point out that sperm cells may use preferably the anaerobic pathway (glycolysis) or the aerobic one (oxidative phosphorylation) to obtain energy (see Figure 1).

Figure 1
Representation of the anaerobic and aerobic energy obtainment pathways.

Boar spermatozoa

Boar sperm cells are recognized as a typical phenotype of slow, short-living cells (Marin et al., 2003Marin S, Chiang K, Bassilian S, Lee W-NP, Boros LG, Fernández-Novell JM, Centelles JJ, Medrano A, Rodriguez-Gil JE, Cascante M. Metabolic strategy of boar spermatozoa revealed by a metabolomic characterization. FEBS Lett. 2003;554(3):342-6. http://dx.doi.org/10.1016/S0014-5793(03)01185-2. PMid:14623091.
http://dx.doi.org/10.1016/S0014-5793(03)...
; Rodriguez-Gil, 2006Rodriguez-Gil J. Mammalian sperm energy resources management and survival during conservation in refrigeration. Reprod Domest Anim. 2006;41(Suppl. 2):11-20. http://dx.doi.org/10.1111/j.1439-0531.2006.00765.x. PMid:16984465.
http://dx.doi.org/10.1111/j.1439-0531.20...
); this fact is related to the physiology of reproduction in this species, in which the female, thus having a long lasting estrous (2-4 days) has a receptibility peak in the middle 24 hours of the estrous period; it is reported that sperm cells in the female genital tract could live no more than 18 hours (Johnson et al., 2000Johnson L, Weitze KF, Fiser P, Maxwell WM. Storage of boar semen. Anim Reprod Sci. 2000;62(1-3):143-72. http://dx.doi.org/10.1016/S0378-4320(00)00157-3. PMid:10924823.
http://dx.doi.org/10.1016/S0378-4320(00)...
).

Studies on metabolism of boar sperm cells defined these cells as primarily glycolytic, showing up to 95% of anaerobic metabolism, as revealed by mass spectrometry studies (Marin et al., 2003Marin S, Chiang K, Bassilian S, Lee W-NP, Boros LG, Fernández-Novell JM, Centelles JJ, Medrano A, Rodriguez-Gil JE, Cascante M. Metabolic strategy of boar spermatozoa revealed by a metabolomic characterization. FEBS Lett. 2003;554(3):342-6. http://dx.doi.org/10.1016/S0014-5793(03)01185-2. PMid:14623091.
http://dx.doi.org/10.1016/S0014-5793(03)...
). In addition, the presence of hexokinase and the metabolizing rate of glucose have led the researcher to sustain this dogma (Fernández-Novell et al., 2004Fernández-Novell JM, Ballester J, Medrano A, Otaegui PJ, Rigau T, Guinovart JJ, Rodríguez-Gil JE. The presence of a high-Km hexokinase activity in dog, but not in boar, sperm. FEBS Lett. 2004;570(1-3):211-6. http://dx.doi.org/10.1016/j.febslet.2004.06.015. PMid:15251466.
http://dx.doi.org/10.1016/j.febslet.2004...
; Medrano et al., 2005Medrano A, Peña A, Rigau T, Rodrìguez-Gil JE. Variations in the proportion of glycolytic/non-glycolytic energy substrates modulate sperm membrane integrity and function in diluted boar samples stored at 15-17 degrees C. Reprod Domest Anim. 2005;40:448-53. http://dx.doi.org/10.1111/j.1439-0531.2005.00599.x
http://dx.doi.org/10.1111/j.1439-0531.20...
, 2006bMedrano A, García-Gil N, Ramió L, Montserrat Rivera M, Fernández-Novell JM, Ramírez A, Peña A, Dolors Briz M, Pinart E, Concha II, Bonet S, Rigau T, Rodríguez-Gil JE. Hexose-specificity of hexokinase and ADP-dependence of pyruvate kinase play important roles in the control of monosaccharide utilization in freshly diluted boar spermatozoa. Mol Reprod Dev. 2006b;73(9):1179-94. http://dx.doi.org/10.1002/mrd.20480. PMid:16804879.
http://dx.doi.org/10.1002/mrd.20480...
). Anyway, the intervention of mitochondria in boar sperm cells metabolism cannot be discarded, as different studies have shown the presence of an active metabolism of mitochondrial substrates (Brooks and Mann, 1973Brooks D, Mann T. Pyruvate metabolism in boar spermatozoa. J Reprod Fertil. 1973;34(1):105-19. http://dx.doi.org/10.1530/jrf.0.0340105. PMid:4719804.
http://dx.doi.org/10.1530/jrf.0.0340105...
; Medrano et al., 2006aMedrano A, Fernández-Novell JM, Ramió L, Alvarez J, Goldberg E, Montserrat Rivera M, Guinovart JJ, Rigau T, Rodríguez-Gil JE. Utilization of citrate and lactate through a lactate dehydrogenase and ATP-regulated pathway in boar spermatozoa. Mol Reprod Dev. 2006a;73(3):369-78. http://dx.doi.org/10.1002/mrd.20414. PMid:16362974.
http://dx.doi.org/10.1002/mrd.20414...
); in addition, a recent study from our laboratories, carried out using different specific inhibitors of the electron transfer chain, demonstrated that boar spermatozoa have an active mitochondrial metabolism and that mitochondria preferably rely on complex I instead of complex II to oxidize substrates (Nesci et al., 2020Nesci S, Spinaci M, Galeati G, Nerozzi C, Pagliarani A, Algieri C, Tamanini C, Bucci D. Sperm function and mitochondrial activity: an insight on boar sperm metabolism. Theriogenology. 2020;144:82-8. http://dx.doi.org/10.1016/j.theriogenology.2020.01.004. PMid:31927418.
http://dx.doi.org/10.1016/j.theriogenolo...
). This finding is opening a new interest on boar sperm mitochondria and their actual role in energy supply.

Dog spermatozoa

Dog spermatozoa are retained as the opposite phenotype of boar sperm: they are fast cells, with a great surviving capacity in the female genital tract (more than 10 days). Again, this situation mirrors the reproductive physiology demands of this species, in which the bitch has a long lasting estrous (till 9 days) and sexual receptivity is not always synchronous with ovulation (up to 11 days from ejaculation to fertilization) (Foutouhi and Meyers, 2022Foutouhi A, Meyers S. Comparative oxidative metabolism in mammalian sperm. Anim Reprod Sci. 2022;247:107095. http://dx.doi.org/10.1016/j.anireprosci.2022.107095. PMid:36272255.
http://dx.doi.org/10.1016/j.anireprosci....
). Therefore, sperm cells from the dog must have the possibility to remain alive and functional for longer time, and evolute different metabolic strategies: high capacity to metabolize glucose, (Fernández-Novell et al., 2004Fernández-Novell JM, Ballester J, Medrano A, Otaegui PJ, Rigau T, Guinovart JJ, Rodríguez-Gil JE. The presence of a high-Km hexokinase activity in dog, but not in boar, sperm. FEBS Lett. 2004;570(1-3):211-6. http://dx.doi.org/10.1016/j.febslet.2004.06.015. PMid:15251466.
http://dx.doi.org/10.1016/j.febslet.2004...
), metabolic plasticity (Bucci et al., 2010aBucci D, Isani G, Spinaci M, Tamanini C, Mari G, Zambelli D, Galeati G. Comparative immunolocalization of GLUTs 1, 2, 3 and 5 in boar, stallion and dog spermatozoa. Reprod Domest Anim. 2010a;45(2):315-22. http://dx.doi.org/10.1111/j.1439-0531.2008.01307.x. PMid:19055550.
http://dx.doi.org/10.1111/j.1439-0531.20...
; Rigau et al., 2001Rigau T, Farré M, Ballester J, Mogas T, Peña A, Rodríguez-Gil JE. Effects of glucose and fructose on motility patterns of dog spermatozoa from fresh ejaculates. Theriogenology. 2001;56(5):601-15. http://dx.doi.org/10.1016/S0093-691X(01)00609-4. PMid:11665883.
http://dx.doi.org/10.1016/S0093-691X(01)...
, 2002Rigau T, Rivera M, Palomo MJ, Fernández-Novell JM, Mogas T, Ballester J, Peña A, Otaegui PJ, Guinovart JJ, Rodríguez-Gil JE. Differential effects of glucose and fructose on hexose metabolism in dog spermatozoa. Reproduction. 2002;123(4):579-91. http://dx.doi.org/10.1530/rep.0.1230579. PMid:11914120.
http://dx.doi.org/10.1530/rep.0.1230579...
), ability to activate anabolic glycogen synthesis pathways (Ballester et al., 2000Ballester J, Fernández-Novell JM, Rutllant J, García-Rocha M, Jesús Palomo M, Mogas T, Peña A, Rigau T, Guinovart JJ, Rodríguez-Gil JE. Evidence for a functional glycogen metabolism in mature mammalian spermatozoa. Mol Reprod Dev. 2000;56(2):207-19. http://dx.doi.org/10.1002/(SICI)1098-2795(200006)56:2<207::AID-MRD12>3.0.CO;2-4. PMid:10813853.
http://dx.doi.org/10.1002/(SICI)1098-279...
) and to further use glycogen for highly demanding functions (Albarracín et al., 2004Albarracín JL, Fernández-Novell JM, Ballester J, Rauch MC, Quintero-Moreno A, Peña A, Mogas T, Rigau T, Yañez A, Guinovart JJ, Slebe JC, Concha II, Rodríguez-Gil JE. Gluconeogenesis-linked glycogen metabolism is important in the achievement of in vitro capacitation of dog spermatozoa in a medium without glucose. Biol Reprod. 2004;71(5):1437-45. http://dx.doi.org/10.1095/biolreprod.104.029041. PMid:15215203.
http://dx.doi.org/10.1095/biolreprod.104...
). Meyers and colleagues (Foutouhi and Meyers, 2022Foutouhi A, Meyers S. Comparative oxidative metabolism in mammalian sperm. Anim Reprod Sci. 2022;247:107095. http://dx.doi.org/10.1016/j.anireprosci.2022.107095. PMid:36272255.
http://dx.doi.org/10.1016/j.anireprosci....
) report that canine spermatozoa demonstrated to have a high capacity to increase their oxidative metabolism when properly treated and that, in case of disruption of oxidative metabolism, they can in a certain way supply with hexose utilization.

Horse spermatozoa

Horse spermatozoa are mostly oxidative cells: it is reported that their metabolic strategy could be defined as “live fast, dye young”. Several researches from the group of Fernando Peña (Davila et al., 2015Davila MP, Muñoz PM, Tapia JA, Ferrusola CO, Silva CCB, Peña FJ. Inhibition of mitochondrial complex I leads to decreased motility and membrane integrity related to increased hydrogen peroxide and reduced ATP production, while the inhibition of glycolysis has less impact on sperm motility. PLoS One. 2015;10(9):e0138777. http://dx.doi.org/10.1371/journal.pone.0138777. PMid:26407142.
http://dx.doi.org/10.1371/journal.pone.0...
, 2016Davila MP, Muñoz PM, Bolaños JMG, Stout T A E, Gadella BM, Tapia JA, Silva CB, Ferrusola CO, Peña FJ. Mitochondrial ATP is required for the maintenance of membrane integrity in stallion spermatozoa, whereas motility requires both glycolysis and oxidative phosphorylation. Reproduction. 2016;152(6):683-94. http://dx.doi.org/10.1530/REP-16-0409. PMid:27798283.
http://dx.doi.org/10.1530/REP-16-0409...
; González-Fernández et al., 2009González-Fernández L, Ortega-Ferrusola C, Macias-Garcia B, Salido GM, Peña FJ, Tapia J. Identification of protein tyrosine phosphatases and dual-specificity phosphatases in mammalian spermatozoa and their role in sperm motility and protein tyrosine phosphorylation. Biol Reprod. 2009;80(6):1239-52. http://dx.doi.org/10.1095/biolreprod.108.073486. PMid:19211810.
http://dx.doi.org/10.1095/biolreprod.108...
; Ortiz-Rodriguez et al., 2021Ortiz-Rodriguez JM, Nerozzi C, Bucci D, Mislei B, Mari G, Tamanini C, Peña FJ, Spinaci M, Galeati G. The inhibition of spermatic cystine/glutamate antiporter xCT (SLC7A11) influences the ability of cryopreserved stallion sperm to bind to heterologous zonae pellucidae. Theriogenology. 2021;167:24-31. http://dx.doi.org/10.1016/j.theriogenology.2021.03.002. PMid:33743505.
http://dx.doi.org/10.1016/j.theriogenolo...
; Peña et al., 2022Peña FJ, Ortiz-Rodríguez JM, Gaitskell-Phillips GL, Gil MC, Ortega-Ferrusola C, Martín-Cano FE. An integrated overview on the regulation of sperm metabolism (glycolysis-Krebs cycle-oxidative phosphorylation). Anim Reprod Sci. 2022;246:106805. http://dx.doi.org/10.1016/j.anireprosci.2021.106805. PMid:34275685.
http://dx.doi.org/10.1016/j.anireprosci....
, 2015Peña FJ, Plaza Davila M, Ball B, Squires EL, Martin Muñoz P, Ortega Ferrusola C, Balao da Silva C. The impact of reproductive technologies on stallion mitochondrial function. Reprod Domest Anim. 2015;50(4):529-37. http://dx.doi.org/10.1111/rda.12551. PMid:26031351.
http://dx.doi.org/10.1111/rda.12551...
; Plaza Dávila et al., 2015Plaza Dávila M, Bucci D, Galeati G, Peña F, Mari G, Giaretta E, Tamanini C, Spinaci M. Epigallocatechin-3-Gallate (EGCG) reduces rotenone effect on stallion sperm-zona pellucida heterologous binding. Reprod Domest Anim. 2015;50(6):1011-6. http://dx.doi.org/10.1111/rda.12628. PMid:26482419.
http://dx.doi.org/10.1111/rda.12628...
) have clearly demonstrated that active and fit mitochondria are needed for horse sperm functioning in the proper way; in particular, the approach chosen in these studies, (recently applied also in our labs to better delineate the relationship between sperm motility, ROS production and mitochondrial function (Giaretta et al., 2022Giaretta E, Mislei B, Martínez-Pastor F, Nesci S, Spinaci M, Galeati G, Nerozzi C, Mari G, Tamanini C, Bucci D. Use of specific mitochondrial complex inhibitors to investigate mitochondrial involvement on horse sperm motility and ROS production. Res Vet Sci. 2022;147:12-9. http://dx.doi.org/10.1016/j.rvsc.2022.03.017. PMid:35397468.
http://dx.doi.org/10.1016/j.rvsc.2022.03...
)) was selective inhibition of electron transfer chain (ETC) of respiratory complexes to determine their role in ETC function and ROS production. Other studies, by Meyers and colleagues ( Darr et al., 2016aDarr CR, Cortopassi GA, Datta S, Varner DD, Meyers SA. Mitochondrial oxygen consumption is a unique indicator of stallion spermatozoal health and varies with cryopreservation media. Theriogenology. 2016a;86(5):1382-92. http://dx.doi.org/10.1016/j.theriogenology.2016.04.082. PMid:27242178.
http://dx.doi.org/10.1016/j.theriogenolo...
, bDarr CR, Varner DD, Teague S, Cortopassi GA, Datta S, Meyers SA. Lactate and pyruvate are major sources of energy for stallion sperm with dose effects on mitochondrial function, motility, and ROS production. Biol Reprod. 2016b;95(2):34-34. http://dx.doi.org/10.1095/biolreprod.116.140707. PMid:27335066.
http://dx.doi.org/10.1095/biolreprod.116...
; Foutouhi and Meyers, 2022Foutouhi A, Meyers S. Comparative oxidative metabolism in mammalian sperm. Anim Reprod Sci. 2022;247:107095. http://dx.doi.org/10.1016/j.anireprosci.2022.107095. PMid:36272255.
http://dx.doi.org/10.1016/j.anireprosci....
; Meyers et al., 2019Meyers S, Bulkeley E, Foutouhi A. Sperm mitochondrial regulation in motility and fertility in horses. Reprod Domest Anim. 2019;54:22-8. http://dx.doi.org/10.1111/rda.13461.
http://dx.doi.org/10.1111/rda.13461...
; Moraes and Meyers, 2018Moraes CR, Meyers S. The sperm mitochondrion: organelle of many functions. Anim Reprod Sci. 2018;194:71-80. http://dx.doi.org/10.1016/j.anireprosci.2018.03.024. PMid:29605167.
http://dx.doi.org/10.1016/j.anireprosci....
) deepened the study of the role of mitochondria in stallion semen function, using different techniques to determine Oxygen Consumption Rate (OCR) and ATP production. Finally, the group of Zamira Gibb furnished some very interesting clues on mitochondria potentiality (Gibb et al., 2014Gibb Z, Lambourne SR, Aitken RJ. The paradoxical relationship between stallion fertility and oxidative stress. Biol Reprod. 2014;91(3):77. http://dx.doi.org/10.1095/biolreprod.114.118539. PMid:25078685.
http://dx.doi.org/10.1095/biolreprod.114...
, 2015Gibb Z, Lambourne SR, Quadrelli J, Smith ND, Aitken RJ. L-carnitine and pyruvate are prosurvival factors during the storage of stallion spermatozoa at room temperature. Biol Reprod. 2015;93(4):104. http://dx.doi.org/10.1095/biolreprod.115.131326. PMid:26316064.
http://dx.doi.org/10.1095/biolreprod.115...
; Gibb and Aitken, 2016Gibb Z, Aitken RJ. The impact of sperm metabolism during in vitro storage : the stallion as a model. BioMed Res Int. 2016;2016:9380609. http://dx.doi.org/10.1155/2016/9380609. PMid:26881234.
http://dx.doi.org/10.1155/2016/9380609...
; Swegen et al., 2016Swegen A, Lambourne SR, Aitken RJ, Gibb Z. Rosiglitazone improves stallion sperm motility, ATP content, and mitochondrial function. Biol Reprod. 2016;95(5):107. http://dx.doi.org/10.1095/biolreprod.116.142687. PMid:27683266.
http://dx.doi.org/10.1095/biolreprod.116...
; Varner et al., 2015Varner DD, Gibb Z, Aitken RJ. Stallion fertility: a focus on the spermatozoon. Equine Vet J. 2015;47(1):16-24. http://dx.doi.org/10.1111/evj.12308. PMid:24943233.
http://dx.doi.org/10.1111/evj.12308...
). These intense studies have demonstrated that an impairment of the mitochondrial function is strongly deleterious for horse sperm function and that the key point to support a good functionality and survival of horse sperm cells is the presence of intact mitochondria.

Bull spermatozoa

Bull spermatozoa have been the first ones to be studied, as reported in the milestone review by Storey (Storey, 2008Storey BT. Mammalian sperm metabolism: oxygen and sugar, friend and foe. Int J Dev Biol. 2008;52(5-6):427-37. http://dx.doi.org/10.1387/ijdb.072522bs. PMid:18649255.
http://dx.doi.org/10.1387/ijdb.072522bs...
). After a relatively long period in which sperm metabolism was not the center of the studies in bull semen, new interest was focused on these features. (Bulkeley et al., 2021Bulkeley EA, Foutouhi A, Wigney K, Santistevan AC, Collins C, McNabb B, Meyers S. Effects from disruption of mitochondrial electron transport chain function on bull sperm motility. Theriogenology. 2021;176:63-72. http://dx.doi.org/10.1016/j.theriogenology.2021.09.015. PMid:34571399.
http://dx.doi.org/10.1016/j.theriogenolo...
; Chatterjee et al., 2001Chatterjee S, de Lamirande E, Gagnon C. Cryopreservation alters membrane sulfhydryl status of bull spermatozoa: protection by oxidized glutathione. Mol Reprod Dev. 2001;60(4):498-506. http://dx.doi.org/10.1002/mrd.1115. PMid:11746961.
http://dx.doi.org/10.1002/mrd.1115...
; Contri et al., 2010Contri A, Valorz C, Faustini M, Wegher L, Carluccio A. Effect of semen preparation on casa motility results in cryopreserved bull spermatozoa. Theriogenology. 2010;74(3):424-35. http://dx.doi.org/10.1016/j.theriogenology.2010.02.025. PMid:20451996.
http://dx.doi.org/10.1016/j.theriogenolo...
; Moraes et al., 2021Moraes CR, Moraes LE, Blawut B, Benej M, Papandreou I, Denko NC, Coutinho da Silva M. Effect of glucose concentration and cryopreservation on mitochondrial functions of bull spermatozoa and relationship with sire conception rate. Anim Reprod Sci. 2021;230:106779. http://dx.doi.org/10.1016/j.anireprosci.2021.106779. PMid:34048998.
http://dx.doi.org/10.1016/j.anireprosci....
; Thys et al., 2009Thys M, Vandaele L, Morrell JM, Mestach J, van Soom A, Hoogewijs M, Rodriguez-Martinez H. In vitro fertilizing capacity of frozen-thawed bull spermatozoa selected by single-layer (glycidoxypropyltrimethoxysilane) silane-coated silica colloidal centrifugation. Reprod Domest Anim. 2009;44(3):390-4. http://dx.doi.org/10.1111/j.1439-0531.2008.01081.x. PMid:18992094.
http://dx.doi.org/10.1111/j.1439-0531.20...
). Bull sperm cells are probably the most widely used in AI techniques all over the world and, generally speaking, they are easily cryopreserved; as a consequence, we register a great advance in the application of AI techniques in spite of basic research on metabolic features.

Bull spermatozoa can rely both on glycolysis and oxidative phosphorylation pathways: in normal conditions the two pathways play an integrated role as expected from somatic cells metabolism (Vishwanath and Shannon, 2000Vishwanath R, Shannon P. Storage of bovine semen in liquid and frozen state. Anim Reprod Sci. 2000;62(1-3):23-53. http://dx.doi.org/10.1016/S0378-4320(00)00153-6. PMid:10924819.
http://dx.doi.org/10.1016/S0378-4320(00)...
). In these conditions, mitochondria are “coupled” and their functionality guarantees a good function of the metabolic machinery (Bulkeley et al., 2021Bulkeley EA, Foutouhi A, Wigney K, Santistevan AC, Collins C, McNabb B, Meyers S. Effects from disruption of mitochondrial electron transport chain function on bull sperm motility. Theriogenology. 2021;176:63-72. http://dx.doi.org/10.1016/j.theriogenology.2021.09.015. PMid:34571399.
http://dx.doi.org/10.1016/j.theriogenolo...
; Moraes et al., 2021Moraes CR, Moraes LE, Blawut B, Benej M, Papandreou I, Denko NC, Coutinho da Silva M. Effect of glucose concentration and cryopreservation on mitochondrial functions of bull spermatozoa and relationship with sire conception rate. Anim Reprod Sci. 2021;230:106779. http://dx.doi.org/10.1016/j.anireprosci.2021.106779. PMid:34048998.
http://dx.doi.org/10.1016/j.anireprosci....
) and sustains motility. Anyway, after cryopreservation, bull sperm mitochondria do not work properly, probably because of cryo-injuries, as we demonstrated in recent research from our labs (Algieri et al., 2022Algieri C, Blanco-Prieto O, Llavanera M, Yeste M, Spinaci M, Mari G, Bucci D, Nesci S. Effects of cryopreservation on the mitochondrial bioenergetics of bovine sperm. Reprod Domest Anim. 2022. http://dx.doi.org/10.1111/rda.14261. PMid:36107136.
http://dx.doi.org/10.1111/rda.14261...
), in which we showed that bull frozen sperm mitochondria are uncoupled, as mitochondrial respiration does not support the ATP synthesis, in contrast with what was observed by other Authors in freshly ejaculated semen (Bulkeley et al., 2021Bulkeley EA, Foutouhi A, Wigney K, Santistevan AC, Collins C, McNabb B, Meyers S. Effects from disruption of mitochondrial electron transport chain function on bull sperm motility. Theriogenology. 2021;176:63-72. http://dx.doi.org/10.1016/j.theriogenology.2021.09.015. PMid:34571399.
http://dx.doi.org/10.1016/j.theriogenolo...
). In another research (under review) we studied the action of different ETC inhibitors on bull frozen sperm cells. The results clearly showed that these cells are more resistant than horse ones to ETC inhibition, and that only inhibition of complex III is able to significantly decrease mitochondrial membrane potential and motility (as observed also in fresh semen) (Bulkeley et al., 2021Bulkeley EA, Foutouhi A, Wigney K, Santistevan AC, Collins C, McNabb B, Meyers S. Effects from disruption of mitochondrial electron transport chain function on bull sperm motility. Theriogenology. 2021;176:63-72. http://dx.doi.org/10.1016/j.theriogenology.2021.09.015. PMid:34571399.
http://dx.doi.org/10.1016/j.theriogenolo...
). These findings together seem to contrast with the fact that frozen semen from bull have overall a good fertilizing ability; we believe that in case of oxidative phosphorylation breakdown, the glycolytic pathway is able to sustain motility and sperm cell homeostasis.

Mouse spermatozoa

Mouse sperm cells were studied under different aspects, one of these was sperm metabolism, and they were considered for a long time strictly glycolytic cells (Ford, 2006Ford WCL. Glycolysis and sperm motility: does a spoonful of sugar help the flagellum go round? Hum Reprod Update. 2006;12(3):269-74. http://dx.doi.org/10.1093/humupd/dmi053. PMid:16407453.
http://dx.doi.org/10.1093/humupd/dmi053...
; Krisfalusi et al., 2006Krisfalusi M, Miki K, Magyar PL, O’Brien DA. Multiple glycolytic enzymes are tightly bound to the fibrous sheath of mouse spermatozoa. Biol Reprod. 2006;75(2):270-8. http://dx.doi.org/10.1095/biolreprod.105.049684. PMid:16687649.
http://dx.doi.org/10.1095/biolreprod.105...
; Mukai and Okuno, 2004Mukai C, Okuno M. Glycolysis plays a major role for adenosine triphosphate supplementation in mouse sperm flagellar movement. Biol Reprod. 2004;71(2):540-7. http://dx.doi.org/10.1095/biolreprod.103.026054. PMid:15084484.
http://dx.doi.org/10.1095/biolreprod.103...
), as motility is strictly related to an active glycolytic pathway within the cell. After almost a decade from these studies, some insights were focused also in mouse sperm mitochondrial activity (Tourmente et al., 2015Tourmente M, Villar-Moya P, Rial E, Roldan ERS. Differences in ATP generation via glycolysis and oxidative phosphorylation and relationships with sperm motility in mouse species. J Biol Chem. 2015;290(33):20613-26. http://dx.doi.org/10.1074/jbc.M115.664813. PMid:26048989.
http://dx.doi.org/10.1074/jbc.M115.66481...
), thus demonstrating that mitochondria may have a role in energy production; a recent work by the same Authors (Tourmente et al., 2022Tourmente M, Sansegundo E, Rial E, Roldan ERS. Capacitation promotes a shift in energy metabolism in murine sperm. Front Cell Dev Biol. 2022;10:950979. http://dx.doi.org/10.3389/fcell.2022.950979. PMid:36081906.
http://dx.doi.org/10.3389/fcell.2022.950...
), applying novel techniques already used in bull, boar and canine spermatozoa(Foutouhi and Meyers, 2022Foutouhi A, Meyers S. Comparative oxidative metabolism in mammalian sperm. Anim Reprod Sci. 2022;247:107095. http://dx.doi.org/10.1016/j.anireprosci.2022.107095. PMid:36272255.
http://dx.doi.org/10.1016/j.anireprosci....
), demonstrated that mitochondrial ATP production plays a pivotal role in capacitation process in mouse spermatozoa, which shift their metabolism from a highly glycolytic one toward an oxidative one. As a technical note, perhaps some investigation on possible differences in sperm metabolism of the most used mouse strains could reveal the best model for translational reseach.

Concluding remarks and future perspectives

The study of sperm metabolism has undergone different moments of interest by the scientific community; anyway, this brief review underlines that knowledge on this topic is not only essential, but can represent an interesting research field. New methodologies and instruments have been developed in the last ten years, thus permitting to shift the approach toward more sensible analysis with respect to those available in the past decades. This led to new discoveries and to update some dogma that seemed to be, as per dogma definition, untouchable. Instead, the role of sperm cells mitochondria has grown in importance and the possibility to study more deeply these organelles showed that they have different roles in different species, and could show a really surprising metabolic plasticity that could be well exploited to develop new preservation strategies or to permit a better control of in vitro sperm activation (capacitation and acrosome reaction). Only in mouse sperm mitochondrial metabolism was studied under capacitating conditions (Tourmente et al., 2022Tourmente M, Sansegundo E, Rial E, Roldan ERS. Capacitation promotes a shift in energy metabolism in murine sperm. Front Cell Dev Biol. 2022;10:950979. http://dx.doi.org/10.3389/fcell.2022.950979. PMid:36081906.
http://dx.doi.org/10.3389/fcell.2022.950...
), but this approach is becoming really precious also to control capacitation in species in which in vitro fertilization IVF works well, such as bovine and porcine, and also in species in which only recently IVF protocols have been described and actuated (Felix et al., 2022Felix MR, Turner RM, Dobbie T, Hinrichs K. Successful in vitro fertilization in the horse: production of blastocysts and birth of foals after prolonged sperm incubation for capacitation. Biol Reprod. 2022;107(6):1551-64. http://dx.doi.org/10.1093/biolre/ioac172. PMid:36106756.
http://dx.doi.org/10.1093/biolre/ioac172...
).

The knowledge of basic sperm metabolism of each species, and the possibility to study at individual level the metabolic features will be of absolute interest in the future for the formulation of new extenders; current research is looking for new preservation strategies (Gibb et al., 2015Gibb Z, Lambourne SR, Quadrelli J, Smith ND, Aitken RJ. L-carnitine and pyruvate are prosurvival factors during the storage of stallion spermatozoa at room temperature. Biol Reprod. 2015;93(4):104. http://dx.doi.org/10.1095/biolreprod.115.131326. PMid:26316064.
http://dx.doi.org/10.1095/biolreprod.115...
; Rizkallah et al., 2022Rizkallah N, Chambers CG, de Graaf SP, Rickard JP. Factors affecting the survival of ram spermatozoa during liquid storage and options for improvement. Animals. 2022;12(3):244. http://dx.doi.org/10.3390/ani12030244. PMid:35158568.
http://dx.doi.org/10.3390/ani12030244...
) possibly avoiding the need to cool semen to too low temperature. This technique could have a great impact on sperm preservation and business, but new extenders should be formulated in order to get the best results in terms of sperm survival, bacterial growth control and fertility.

Acknowledgements

This research was supported by the European Commission - NextGenerationEU - ALMArie CURIE 2021 SupER - University of Bologna funded by Italian Ministry of University and Research (D.M. 737/2021)(CUP: J45F21001470005) and by EDITAL FAPERGS/CAPES 06/2018.

  • Financial support: DB was financially supported by the European Commission - NextGenerationEU - ALMArie CURIE 2021 SupER - University of Bologna funded by Italian Ministry of University and Research (D.M. 737/2021) (CUP: J45F21001470005) and by EDITAL FAPERGS/CAPES 06/2018.
  • How to cite: Bucci D, Spinaci M, Bustamante-Filho IC, Nesci S. The sperm mitochondria: clues and challenges. Anim Reprod. 2022;19(4):e20220131. https://doi.org/10.1590/1984-3143-AR2022-0131

References

  • Albarracín JL, Fernández-Novell JM, Ballester J, Rauch MC, Quintero-Moreno A, Peña A, Mogas T, Rigau T, Yañez A, Guinovart JJ, Slebe JC, Concha II, Rodríguez-Gil JE. Gluconeogenesis-linked glycogen metabolism is important in the achievement of in vitro capacitation of dog spermatozoa in a medium without glucose. Biol Reprod. 2004;71(5):1437-45. http://dx.doi.org/10.1095/biolreprod.104.029041 PMid:15215203.
    » http://dx.doi.org/10.1095/biolreprod.104.029041
  • Algieri C, Blanco-Prieto O, Llavanera M, Yeste M, Spinaci M, Mari G, Bucci D, Nesci S. Effects of cryopreservation on the mitochondrial bioenergetics of bovine sperm. Reprod Domest Anim. 2022. http://dx.doi.org/10.1111/rda.14261 PMid:36107136.
    » http://dx.doi.org/10.1111/rda.14261
  • Angulo C, Rauch MC, Droppelmann A, Reyes AM, Slebe JC, Delgado-López F, Guaiquil VH, Vera JC, Concha II. Hexose transporter expression and function in mammalian spermatozoa: cellular localization and transport of hexoses and vitamin C. J Cell Biochem. 1998;71(2):189-203. http://dx.doi.org/10.1002/(SICI)1097-4644(19981101)71:2<189::AID-JCB5>3.0.CO;2-R PMid:9779818.
    » http://dx.doi.org/10.1002/(SICI)1097-4644(19981101)71:2<189::AID-JCB5>3.0.CO;2-R
  • Ballester J, Fernández-Novell JM, Rutllant J, García-Rocha M, Jesús Palomo M, Mogas T, Peña A, Rigau T, Guinovart JJ, Rodríguez-Gil JE. Evidence for a functional glycogen metabolism in mature mammalian spermatozoa. Mol Reprod Dev. 2000;56(2):207-19. http://dx.doi.org/10.1002/(SICI)1098-2795(200006)56:2<207::AID-MRD12>3.0.CO;2-4 PMid:10813853.
    » http://dx.doi.org/10.1002/(SICI)1098-2795(200006)56:2<207::AID-MRD12>3.0.CO;2-4
  • Boguenet M, Bouet PE, Spiers A, Reynier P, May-Panloup P. Mitochondria: their role in spermatozoa and in male infertility. Hum Reprod Update. 2021;27(4):697-719. http://dx.doi.org/10.1093/humupd/dmab001 PMid:33555313.
    » http://dx.doi.org/10.1093/humupd/dmab001
  • Brooks D, Mann T. Pyruvate metabolism in boar spermatozoa. J Reprod Fertil. 1973;34(1):105-19. http://dx.doi.org/10.1530/jrf.0.0340105 PMid:4719804.
    » http://dx.doi.org/10.1530/jrf.0.0340105
  • Brooks DE, Mann T. Relation between the oxidation state of nicotinamide-adenine dinucleotide and the metabolism of spermatozoa. Biochem J. 1972;129(5):1023-34. http://dx.doi.org/10.1042/bj1291023 PMid:4144231.
    » http://dx.doi.org/10.1042/bj1291023
  • Bucci D, Isani G, Spinaci M, Tamanini C, Mari G, Zambelli D, Galeati G. Comparative immunolocalization of GLUTs 1, 2, 3 and 5 in boar, stallion and dog spermatozoa. Reprod Domest Anim. 2010a;45(2):315-22. http://dx.doi.org/10.1111/j.1439-0531.2008.01307.x PMid:19055550.
    » http://dx.doi.org/10.1111/j.1439-0531.2008.01307.x
  • Bucci D, Rodríguez-Gil JE, Vallorani C, Spinaci M, Galeati G, Tamanini C. GLUTs and mammalian sperm metabolism. J Androl. 2011;32(4):348-55. http://dx.doi.org/10.2164/jandrol.110.011197 PMid:21088231.
    » http://dx.doi.org/10.2164/jandrol.110.011197
  • Bucci D, Spinaci M, Vallorani C, Contri A, Carluccio A, Isani G, Tamanini C, Galeati G. Detection and localization of GLUTs 1, 2, 3 and 5 in donkey spermatozoa. Reprod Domest Anim. 2010b;45(5):e217-20. http://dx.doi.org/10.1111/j.1439-0531.2009.01544.x PMid:19930135.
    » http://dx.doi.org/10.1111/j.1439-0531.2009.01544.x
  • Bulkeley EA, Foutouhi A, Wigney K, Santistevan AC, Collins C, McNabb B, Meyers S. Effects from disruption of mitochondrial electron transport chain function on bull sperm motility. Theriogenology. 2021;176:63-72. http://dx.doi.org/10.1016/j.theriogenology.2021.09.015 PMid:34571399.
    » http://dx.doi.org/10.1016/j.theriogenology.2021.09.015
  • Chatterjee S, de Lamirande E, Gagnon C. Cryopreservation alters membrane sulfhydryl status of bull spermatozoa: protection by oxidized glutathione. Mol Reprod Dev. 2001;60(4):498-506. http://dx.doi.org/10.1002/mrd.1115 PMid:11746961.
    » http://dx.doi.org/10.1002/mrd.1115
  • Contri A, Valorz C, Faustini M, Wegher L, Carluccio A. Effect of semen preparation on casa motility results in cryopreserved bull spermatozoa. Theriogenology. 2010;74(3):424-35. http://dx.doi.org/10.1016/j.theriogenology.2010.02.025 PMid:20451996.
    » http://dx.doi.org/10.1016/j.theriogenology.2010.02.025
  • Darr CR, Cortopassi GA, Datta S, Varner DD, Meyers SA. Mitochondrial oxygen consumption is a unique indicator of stallion spermatozoal health and varies with cryopreservation media. Theriogenology. 2016a;86(5):1382-92. http://dx.doi.org/10.1016/j.theriogenology.2016.04.082 PMid:27242178.
    » http://dx.doi.org/10.1016/j.theriogenology.2016.04.082
  • Darr CR, Varner DD, Teague S, Cortopassi GA, Datta S, Meyers SA. Lactate and pyruvate are major sources of energy for stallion sperm with dose effects on mitochondrial function, motility, and ROS production. Biol Reprod. 2016b;95(2):34-34. http://dx.doi.org/10.1095/biolreprod.116.140707 PMid:27335066.
    » http://dx.doi.org/10.1095/biolreprod.116.140707
  • Davila MP, Muñoz PM, Bolaños JMG, Stout T A E, Gadella BM, Tapia JA, Silva CB, Ferrusola CO, Peña FJ. Mitochondrial ATP is required for the maintenance of membrane integrity in stallion spermatozoa, whereas motility requires both glycolysis and oxidative phosphorylation. Reproduction. 2016;152(6):683-94. http://dx.doi.org/10.1530/REP-16-0409 PMid:27798283.
    » http://dx.doi.org/10.1530/REP-16-0409
  • Davila MP, Muñoz PM, Tapia JA, Ferrusola CO, Silva CCB, Peña FJ. Inhibition of mitochondrial complex I leads to decreased motility and membrane integrity related to increased hydrogen peroxide and reduced ATP production, while the inhibition of glycolysis has less impact on sperm motility. PLoS One. 2015;10(9):e0138777. http://dx.doi.org/10.1371/journal.pone.0138777 PMid:26407142.
    » http://dx.doi.org/10.1371/journal.pone.0138777
  • Felix MR, Turner RM, Dobbie T, Hinrichs K. Successful in vitro fertilization in the horse: production of blastocysts and birth of foals after prolonged sperm incubation for capacitation. Biol Reprod. 2022;107(6):1551-64. http://dx.doi.org/10.1093/biolre/ioac172 PMid:36106756.
    » http://dx.doi.org/10.1093/biolre/ioac172
  • Fernández-Novell JM, Ballester J, Medrano A, Otaegui PJ, Rigau T, Guinovart JJ, Rodríguez-Gil JE. The presence of a high-Km hexokinase activity in dog, but not in boar, sperm. FEBS Lett. 2004;570(1-3):211-6. http://dx.doi.org/10.1016/j.febslet.2004.06.015 PMid:15251466.
    » http://dx.doi.org/10.1016/j.febslet.2004.06.015
  • Ford WCL. Glycolysis and sperm motility: does a spoonful of sugar help the flagellum go round? Hum Reprod Update. 2006;12(3):269-74. http://dx.doi.org/10.1093/humupd/dmi053 PMid:16407453.
    » http://dx.doi.org/10.1093/humupd/dmi053
  • Foutouhi A, Meyers S. Comparative oxidative metabolism in mammalian sperm. Anim Reprod Sci. 2022;247:107095. http://dx.doi.org/10.1016/j.anireprosci.2022.107095 PMid:36272255.
    » http://dx.doi.org/10.1016/j.anireprosci.2022.107095
  • Giaretta E, Mislei B, Martínez-Pastor F, Nesci S, Spinaci M, Galeati G, Nerozzi C, Mari G, Tamanini C, Bucci D. Use of specific mitochondrial complex inhibitors to investigate mitochondrial involvement on horse sperm motility and ROS production. Res Vet Sci. 2022;147:12-9. http://dx.doi.org/10.1016/j.rvsc.2022.03.017 PMid:35397468.
    » http://dx.doi.org/10.1016/j.rvsc.2022.03.017
  • Gibb Z, Aitken RJ. The impact of sperm metabolism during in vitro storage : the stallion as a model. BioMed Res Int. 2016;2016:9380609. http://dx.doi.org/10.1155/2016/9380609 PMid:26881234.
    » http://dx.doi.org/10.1155/2016/9380609
  • Gibb Z, Lambourne SR, Aitken RJ. The paradoxical relationship between stallion fertility and oxidative stress. Biol Reprod. 2014;91(3):77. http://dx.doi.org/10.1095/biolreprod.114.118539 PMid:25078685.
    » http://dx.doi.org/10.1095/biolreprod.114.118539
  • Gibb Z, Lambourne SR, Quadrelli J, Smith ND, Aitken RJ. L-carnitine and pyruvate are prosurvival factors during the storage of stallion spermatozoa at room temperature. Biol Reprod. 2015;93(4):104. http://dx.doi.org/10.1095/biolreprod.115.131326 PMid:26316064.
    » http://dx.doi.org/10.1095/biolreprod.115.131326
  • González-Fernández L, Ortega-Ferrusola C, Macias-Garcia B, Salido GM, Peña FJ, Tapia J. Identification of protein tyrosine phosphatases and dual-specificity phosphatases in mammalian spermatozoa and their role in sperm motility and protein tyrosine phosphorylation. Biol Reprod. 2009;80(6):1239-52. http://dx.doi.org/10.1095/biolreprod.108.073486 PMid:19211810.
    » http://dx.doi.org/10.1095/biolreprod.108.073486
  • Hutson SM, van Dop C, Lardy HA. Mitochondrial metabolism of pyruvate in bovine spermatozoa. J Biol Chem. 1977;252(4):1309-15. http://dx.doi.org/10.1016/S0021-9258(17)40656-9 PMid:838719.
    » http://dx.doi.org/10.1016/S0021-9258(17)40656-9
  • Johnson L, Weitze KF, Fiser P, Maxwell WM. Storage of boar semen. Anim Reprod Sci. 2000;62(1-3):143-72. http://dx.doi.org/10.1016/S0378-4320(00)00157-3 PMid:10924823.
    » http://dx.doi.org/10.1016/S0378-4320(00)00157-3
  • Krisfalusi M, Miki K, Magyar PL, O’Brien DA. Multiple glycolytic enzymes are tightly bound to the fibrous sheath of mouse spermatozoa. Biol Reprod. 2006;75(2):270-8. http://dx.doi.org/10.1095/biolreprod.105.049684 PMid:16687649.
    » http://dx.doi.org/10.1095/biolreprod.105.049684
  • Marin S, Chiang K, Bassilian S, Lee W-NP, Boros LG, Fernández-Novell JM, Centelles JJ, Medrano A, Rodriguez-Gil JE, Cascante M. Metabolic strategy of boar spermatozoa revealed by a metabolomic characterization. FEBS Lett. 2003;554(3):342-6. http://dx.doi.org/10.1016/S0014-5793(03)01185-2 PMid:14623091.
    » http://dx.doi.org/10.1016/S0014-5793(03)01185-2
  • Medrano A, Peña A, Rigau T, Rodrìguez-Gil JE. Variations in the proportion of glycolytic/non-glycolytic energy substrates modulate sperm membrane integrity and function in diluted boar samples stored at 15-17 degrees C. Reprod Domest Anim. 2005;40:448-53. http://dx.doi.org/10.1111/j.1439-0531.2005.00599.x
    » http://dx.doi.org/10.1111/j.1439-0531.2005.00599.x
  • Medrano A, Fernández-Novell JM, Ramió L, Alvarez J, Goldberg E, Montserrat Rivera M, Guinovart JJ, Rigau T, Rodríguez-Gil JE. Utilization of citrate and lactate through a lactate dehydrogenase and ATP-regulated pathway in boar spermatozoa. Mol Reprod Dev. 2006a;73(3):369-78. http://dx.doi.org/10.1002/mrd.20414 PMid:16362974.
    » http://dx.doi.org/10.1002/mrd.20414
  • Medrano A, García-Gil N, Ramió L, Montserrat Rivera M, Fernández-Novell JM, Ramírez A, Peña A, Dolors Briz M, Pinart E, Concha II, Bonet S, Rigau T, Rodríguez-Gil JE. Hexose-specificity of hexokinase and ADP-dependence of pyruvate kinase play important roles in the control of monosaccharide utilization in freshly diluted boar spermatozoa. Mol Reprod Dev. 2006b;73(9):1179-94. http://dx.doi.org/10.1002/mrd.20480 PMid:16804879.
    » http://dx.doi.org/10.1002/mrd.20480
  • Meyers S, Bulkeley E, Foutouhi A. Sperm mitochondrial regulation in motility and fertility in horses. Reprod Domest Anim. 2019;54:22-8. http://dx.doi.org/10.1111/rda.13461
    » http://dx.doi.org/10.1111/rda.13461
  • Moraes CR, Meyers S. The sperm mitochondrion: organelle of many functions. Anim Reprod Sci. 2018;194:71-80. http://dx.doi.org/10.1016/j.anireprosci.2018.03.024 PMid:29605167.
    » http://dx.doi.org/10.1016/j.anireprosci.2018.03.024
  • Moraes CR, Moraes LE, Blawut B, Benej M, Papandreou I, Denko NC, Coutinho da Silva M. Effect of glucose concentration and cryopreservation on mitochondrial functions of bull spermatozoa and relationship with sire conception rate. Anim Reprod Sci. 2021;230:106779. http://dx.doi.org/10.1016/j.anireprosci.2021.106779 PMid:34048998.
    » http://dx.doi.org/10.1016/j.anireprosci.2021.106779
  • Mukai C, Okuno M. Glycolysis plays a major role for adenosine triphosphate supplementation in mouse sperm flagellar movement. Biol Reprod. 2004;71(2):540-7. http://dx.doi.org/10.1095/biolreprod.103.026054 PMid:15084484.
    » http://dx.doi.org/10.1095/biolreprod.103.026054
  • Nesci S, Spinaci M, Galeati G, Nerozzi C, Pagliarani A, Algieri C, Tamanini C, Bucci D. Sperm function and mitochondrial activity: an insight on boar sperm metabolism. Theriogenology. 2020;144:82-8. http://dx.doi.org/10.1016/j.theriogenology.2020.01.004 PMid:31927418.
    » http://dx.doi.org/10.1016/j.theriogenology.2020.01.004
  • Ortiz-Rodriguez JM, Nerozzi C, Bucci D, Mislei B, Mari G, Tamanini C, Peña FJ, Spinaci M, Galeati G. The inhibition of spermatic cystine/glutamate antiporter xCT (SLC7A11) influences the ability of cryopreserved stallion sperm to bind to heterologous zonae pellucidae. Theriogenology. 2021;167:24-31. http://dx.doi.org/10.1016/j.theriogenology.2021.03.002 PMid:33743505.
    » http://dx.doi.org/10.1016/j.theriogenology.2021.03.002
  • Peña FJ, Ortiz-Rodríguez JM, Gaitskell-Phillips GL, Gil MC, Ortega-Ferrusola C, Martín-Cano FE. An integrated overview on the regulation of sperm metabolism (glycolysis-Krebs cycle-oxidative phosphorylation). Anim Reprod Sci. 2022;246:106805. http://dx.doi.org/10.1016/j.anireprosci.2021.106805 PMid:34275685.
    » http://dx.doi.org/10.1016/j.anireprosci.2021.106805
  • Peña FJ, Plaza Davila M, Ball B, Squires EL, Martin Muñoz P, Ortega Ferrusola C, Balao da Silva C. The impact of reproductive technologies on stallion mitochondrial function. Reprod Domest Anim. 2015;50(4):529-37. http://dx.doi.org/10.1111/rda.12551 PMid:26031351.
    » http://dx.doi.org/10.1111/rda.12551
  • Plaza Dávila M, Bucci D, Galeati G, Peña F, Mari G, Giaretta E, Tamanini C, Spinaci M. Epigallocatechin-3-Gallate (EGCG) reduces rotenone effect on stallion sperm-zona pellucida heterologous binding. Reprod Domest Anim. 2015;50(6):1011-6. http://dx.doi.org/10.1111/rda.12628 PMid:26482419.
    » http://dx.doi.org/10.1111/rda.12628
  • Rigau T, Farré M, Ballester J, Mogas T, Peña A, Rodríguez-Gil JE. Effects of glucose and fructose on motility patterns of dog spermatozoa from fresh ejaculates. Theriogenology. 2001;56(5):601-15. http://dx.doi.org/10.1016/S0093-691X(01)00609-4 PMid:11665883.
    » http://dx.doi.org/10.1016/S0093-691X(01)00609-4
  • Rigau T, Rivera M, Palomo MJ, Fernández-Novell JM, Mogas T, Ballester J, Peña A, Otaegui PJ, Guinovart JJ, Rodríguez-Gil JE. Differential effects of glucose and fructose on hexose metabolism in dog spermatozoa. Reproduction. 2002;123(4):579-91. http://dx.doi.org/10.1530/rep.0.1230579 PMid:11914120.
    » http://dx.doi.org/10.1530/rep.0.1230579
  • Rizkallah N, Chambers CG, de Graaf SP, Rickard JP. Factors affecting the survival of ram spermatozoa during liquid storage and options for improvement. Animals. 2022;12(3):244. http://dx.doi.org/10.3390/ani12030244 PMid:35158568.
    » http://dx.doi.org/10.3390/ani12030244
  • Rodriguez-Gil J. Mammalian sperm energy resources management and survival during conservation in refrigeration. Reprod Domest Anim. 2006;41(Suppl. 2):11-20. http://dx.doi.org/10.1111/j.1439-0531.2006.00765.x PMid:16984465.
    » http://dx.doi.org/10.1111/j.1439-0531.2006.00765.x
  • Rodríguez-Gil JE, Bonet S. Current knowledge on boar sperm metabolism: comparison with other mammalian species. Theriogenology. 2016;85(1):4-11. http://dx.doi.org/10.1016/j.theriogenology.2015.05.005 PMid:26094247.
    » http://dx.doi.org/10.1016/j.theriogenology.2015.05.005
  • Storey BT. Mammalian sperm metabolism: oxygen and sugar, friend and foe. Int J Dev Biol. 2008;52(5-6):427-37. http://dx.doi.org/10.1387/ijdb.072522bs PMid:18649255.
    » http://dx.doi.org/10.1387/ijdb.072522bs
  • Sung TK, Moley KH. The expression of GLUT8, GLUT9a, and GLUT9b in the mouse testis and sperm. Reprod Sci. 2007;14(5):445-55. http://dx.doi.org/10.1177/1933719107306226 PMid:17913964.
    » http://dx.doi.org/10.1177/1933719107306226
  • Swegen A, Lambourne SR, Aitken RJ, Gibb Z. Rosiglitazone improves stallion sperm motility, ATP content, and mitochondrial function. Biol Reprod. 2016;95(5):107. http://dx.doi.org/10.1095/biolreprod.116.142687 PMid:27683266.
    » http://dx.doi.org/10.1095/biolreprod.116.142687
  • Thys M, Vandaele L, Morrell JM, Mestach J, van Soom A, Hoogewijs M, Rodriguez-Martinez H. In vitro fertilizing capacity of frozen-thawed bull spermatozoa selected by single-layer (glycidoxypropyltrimethoxysilane) silane-coated silica colloidal centrifugation. Reprod Domest Anim. 2009;44(3):390-4. http://dx.doi.org/10.1111/j.1439-0531.2008.01081.x PMid:18992094.
    » http://dx.doi.org/10.1111/j.1439-0531.2008.01081.x
  • Tourmente M, Sansegundo E, Rial E, Roldan ERS. Capacitation promotes a shift in energy metabolism in murine sperm. Front Cell Dev Biol. 2022;10:950979. http://dx.doi.org/10.3389/fcell.2022.950979 PMid:36081906.
    » http://dx.doi.org/10.3389/fcell.2022.950979
  • Tourmente M, Villar-Moya P, Rial E, Roldan ERS. Differences in ATP generation via glycolysis and oxidative phosphorylation and relationships with sperm motility in mouse species. J Biol Chem. 2015;290(33):20613-26. http://dx.doi.org/10.1074/jbc.M115.664813 PMid:26048989.
    » http://dx.doi.org/10.1074/jbc.M115.664813
  • Varner DD, Gibb Z, Aitken RJ. Stallion fertility: a focus on the spermatozoon. Equine Vet J. 2015;47(1):16-24. http://dx.doi.org/10.1111/evj.12308 PMid:24943233.
    » http://dx.doi.org/10.1111/evj.12308
  • Vishwanath R, Shannon P. Storage of bovine semen in liquid and frozen state. Anim Reprod Sci. 2000;62(1-3):23-53. http://dx.doi.org/10.1016/S0378-4320(00)00153-6 PMid:10924819.
    » http://dx.doi.org/10.1016/S0378-4320(00)00153-6

Publication Dates

  • Publication in this collection
    13 Feb 2023
  • Date of issue
    2022

History

  • Received
    20 Dec 2022
  • Accepted
    24 Jan 2023
Colégio Brasileiro de Reprodução Animal Coronel José dias Bicalho, 1224, CEP: , 31275-050, Belo Horizonte, MG - Brasil, Tel.: 55-31-3491 7122 - Belo Horizonte - MG - Brazil
E-mail: animreprod.journal@gmail.com