What provides energy for sperm movement

Visual assessment of sperm motility should include total sperm motility (percentage of sperm that exhibit motility of any form), progressive sperm motility (percentage of sperm that exhibit rapid, linear movement), and sperm velocity (on an arbitrary scale of 0 [immotile] to 4 [rapidly motile]). For example, a motility of 75/70 (4) indicates that 75% of sperm were motile and 70% of sperm were progressively motile, moving rapidly across the microscopic field. Rapid progressive sperm motility generally is considered to be the most credible gauge of sperm motion for predicting the fertilizing capacity of a semen sample.

Both the initial sperm motility and the longevity of progressive sperm motility should be assessed and recorded. Initial sperm motility of raw (undiluted) semen samples can be estimated as a control for testing the possible detrimental effects of semen extenders on sperm motility. Accuracy and repeatability of the sperm motility evaluation are improved markedly by diluting the semen in an appropriate extender before analysis (see Tables 12-1 and 12-2). Warmed (37°C) nonfat dry skim milk-glucose extender, or native phosphocaseinate-based extender, serves this purpose well because it supports sperm motility and does not interfere with microscopic visualization of the sperm. To standardize the sperm motility testing protocol, all semen samples should be diluted with extender before analysis. We prefer to dilute semen to a standard concentration of 25 to 30 million sperm/mL for motility assessment. One advantage of use of a standard concentration of extended semen is that it conditions the viewer to see sperm at a consistent concentration among all stallion ejaculates evaluated. At higher concentrations, sperm motility tends to be overestimated. This concentration of sperm diluted in an appropriate extender has also been shown to maximize both immediate sperm motility and the longevity of sperm motility.

The longevity of sperm motility can be determined on raw semen samples stored at room temperature (20°C to 25°C) and on samples diluted in extender (preferably to a final sperm concentration of 25 × 106 sperm/mL) and stored at room temperature or refrigerated (4°C to 6°C). The longevity of sperm motility is enhanced by dilution of semen with extender and refrigerated storage. The Society for Theriogenology guidelines for evaluating the semen of prospective breeding stallions recommend that at least 10% progressive sperm motility be maintained in raw and extended semen samples maintained in a light-shielded environment at room temperature for 6 and 24 hours, respectively. If properly processed, semen from stallions that survives cooling well will have similar to slightly lower sperm motility after 24 hours of cooling compared to that semen when evaluated fresh. The relationship between longevity of sperm motility in samples maintained at room temperature is questioned by many investigators, whereas less dissension is found concerning the value of determining the longevity of the sperm motility in cooled samples when processed ejaculates are used for breeding with cooled, transported semen.

To accurately assess longevity of sperm motility in vitro, semen should be first be properly extended. This necessitates diluting semen so that less than 20% (volume:volume) seminal plasma (preferably 5% to 10%) remains in the extended samples, as excess seminal plasma depresses sperm membrane stability, DNA quality, and motility over time. If raw semen concentration is too dilute to allow a 1:4 dilution (i.e., 1 part semen to 4 parts extender) without lowering final sperm concentration to less than 25 million sperm/mL, then excess seminal plasma must be removed after centrifugation. To maximize sperm recovery and minimize damage to sperm (Figures 13-35 to 13-38), semen is first mixed with an equal volume of extender, and 35 mL of extended semen is placed in a 50-mL conical polypropylene centrifuge tube. Then, 3.5 mL of cushion fluid (e.g., Eqcellsire Component B, IMV Technologies, Maple Grove, Minn. or Cushion fluid, Minitúb, Tiefenbach, Germany) is placed beneath the semen in the bottom of the tube with a blunt 18-gauge spinal needle with attached syringe. The loaded tube is centrifuged at 1000×g for 20 minutes, and the supernatant is aspirated down to the 7.5 mL mark on the centrifuge tube. With the spinal needle, the cushion fluid is carefully removed, and the remaining sperm pellet is mixed with sufficient fresh extender to result in a final sperm concentration of 25 to 50 million sperm/mL yet still retain 5% to 10% seminal plasma (e.g., 4 mL of centrifuged semen was left after centrifugation, so addition of 36 mL extender results in a maximum of 10% seminal plasma being present). After the desired dilution is accomplished, the semen is used to fill nontoxic vials (or bags), ensuring all air is eliminated. The vials are placed in an insulated, dark environment for incubation at room temperature and also loaded into a container such as an Equitainer (Hamilton-Thorne Inc, Beverly, MA) for cooling and storage at 4°C to 6°C. Semen samples should be warmed to 37°C for 10 to 15 minutes before assessment of sperm motility after the appropriate incubation period.

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/B9780323064828000223

OSMOTIC, IONIC AND NITROGENOUS-WASTE BALANCE | Osmosensing

D. Kültz, in Encyclopedia of Fish Physiology, 2011

Role of Osmosensing for Fish Sperm Motility

Fish sperm motility is controlled tightly by osmolality and the concentration of cations. Therefore, fish sperm represents an excellent model for studying general mechanisms of cellular osmosensing in fish. Depending on habitat salinity and osmoregulatory strategy, sperm motility is either activated or inhibited by increased osmolality. In marine teleosts, sperm motility increases when released into a plasmahyperosmotic environment (SW), whereas in FW teleosts it increases upon release into a plasmahypoosmotic environment (FW). In striped bass sperm motility is promoted by Ca2+-free solutions and suppressed when [Ca2+] exceeds 10 mM. The effect of [Ca2+] is transient and irreversible; once sperm motility has been activated, it cannot be suppressed by high [Ca2+] (80 mM) and reactivation by Ca2+-free medium after suppression is also not possible. Verapamil, a Ca2+ channel inhibitor, and cAMP had no effect on sperm motility in striped bass suggesting that Ca2+ acts through other pathways. These data reinforce the notion that calcium is of central importance for osmosensing (see above). Because of the rapid time course of initiation of sperm motility, the study of sensory and signaling mechanisms underlying this event in marine versus FW fishes may provide critical clues about mechanisms of osmosensing in fish. In addition to calcium, intracellular [K+] has also been shown to be critical for the regulation of sperm motility in FW (zebrafish) and marine (pufferfish) species. Thus, intracellular inorganic cation concentrations are critical signals for osmosensory signal transduction in fish sperm.

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/B9780123745538002136

Hypoxia

Rudolf S.S. Wu, in Fish Physiology, 2009

2.3.3 Quality of Sperm and Eggs

Sperm motility is a reliable predictor for sperm quality and fertilization success (Au et al., 2002). After exposure to hypoxia for 12 weeks, sperm motility (measured by their curvilinear velocity VCL, straight‐line velocity VSL, and angular path velocity VAP) was significantly decreased in male carp (Table 3.2), indicating that sperm quality was impaired. All of the normoxic and hypoxic male carp could be induced to spawn using carp pituitary extract; however, the percentage of spawning success in the hypoxic male carp was drastically reduced from 71.4% to 8.3%, clearly demonstrating that the sperm quality produced by males was impaired by hypoxia (Wu et al., 2003).

Table 3.2. Sperm motility of carp after exposure to normoxia (7.0 mg O2/L, ∼81% saturation) and hypoxia (1.0 mg O2/L, ∼12% saturation) for 12 weeks

7.0 mg O2/L1.0 mg O2/LVCL77.42 ± 29.1346.25 ± 10.83*VSL38.83 ± 21.0110.65 ± 3.89*VAP47.69 ± 5.3821.12 ± 11.41*

Mean ± SD; n = 6. The velocity is expressed as micrometers per second.

VCL, mean curvilinear velocity; VSL, mean straight‐line velocity; VAP, angular path velocity (Wu et al., 2003).

*Values significantly different from the control (t‐test: *, p < 0.05).

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/S1546509808000034

Systems Toxicologic Pathology

Dianne M. Creasy, Robert E. Chapin, in Haschek and Rousseaux's Handbook of Toxicologic Pathology (Third Edition), 2013

Sperm Motility

Sperm motility is a functional measurement of the sperm themselves. Sperm are sampled either from the cauda or the vas deferens, and assessed for motility either manually or by CASA. In rats, motility is generally high and fairly consistent between animals: the normal range for percent motile varies between different laboratories, but control values should be in the 85–96% range for rodents, dogs, and monkeys. There is an extensive literature on the methodologic requirements for measuring motility and obtaining acceptable values (a minimum acceptable value for sperm motility in control groups is generally considered as 70%). It is recommended that the process of making these measures be assigned to a group within the research organization with the interest and skill to focus on obtaining the best and appropriately representative data from each sample.

Sperm motility can be affected by disturbances in testicular spermatogenesis or by effects on the epididymis. One way to consider these endpoints in the laboratory is that the testis provides the “hardware” (the substrate) for motility. The epididymis modifies that substrate and provides the motility-conferring “software” factors for the sperm. Thus, in a laboratory study in rodents, if normal numbers of well-formed sperm were immotile in the absence of testis pathology, one might reasonably suspect a treatment effect on the epididymis.

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/B9780124157590000595

Is the Classic Spermiogram Still Informative? How Did It Develop and Where Is It Going?

William V. Holt, in Reproductomics, 2018

The Biological Relevance of Sperm Motility

Sperm motility alone, expressed using simple parameters such as average sperm velocity or estimates of the proportion of motile spermatozoa in a sample, is a poor predictor of conception rate, whether estimated in humans or agricultural animals. Experimental approaches to determine the predictive value of sperm motility have to take account of the potential confounding influence of inconsistent numbers of inseminated spermatozoa, where there are positive correlations between sperm numbers and fertility outcomes. Natural experiments involving reproductive skews and sperm competition can, however, provide useful indications about the value of sperm motility and other parameters. Critically important clues about the relevance of sperm motility have come from a series of papers that focused on mouse reproduction (e.g., the t-haplotype mouse [80]) and transmission-ratio distortion (TRD). This is an effect whereby genetic mosaicism resulting from meiotic recombination during spermatogenesis leads to the development of genetically distinct sperm subpopulations within a single ejaculate that are either functionally advantaged or disadvantaged with respect to flagellar activity. While spermatozoa with normal flagellar activity are able to cross the uterotubal junction, enter the oviduct, and reach the oocyte, those spermatozoa with abnormal flagellar function are unable to do so. Mechanistically, TRD in the case of the t-haplotype occurs because the cell signaling cascades that control flagellar function and motility operate incorrectly. The protein kinases controlling flagellar function in these mice are overexpressed and cause abnormal sperm motility; however, those spermatozoa carrying the t-haplotype also possess a t-complex responder gene (Tcr) that corrects the overexpression of the sperm motility kinase gene (smok) and restores normal flagellar action [81]. During meiosis, the Tcr cosegregates with the Y-chromosome and causes 95% skewing of the offspring sex ratio in favor of males by promoting unbalanced fertilization success. This extreme example, which results in non-Mendelian inheritance, is paralleled by non-Mendelian transmission of retinoblastoma in humans. Girardet et al. [82] proposed that TRD and sex-ratio distortion among the offspring of males affected with sporadic bilateral retinoblastoma could be explained by the existence of a defectively imprinted gene located on the human X chromosome that produces a subpopulation of spermatozoa that show defective motility.

These data can be viewed as natural sperm competition experiments that provide an intriguing insight into possible mechanisms of sperm selection. They indicate how genetic traits, not overtly reproductive in nature, can be influenced directly by their association with protein kinase-regulated signaling cascades that affect sperm motility. Reproductive skews detected in heterospermic artificial insemination experiments [39,41,83], where spermatozoa from two or more males are mixed in equal proportions, may be partly attributable to similar mechanisms. This suggestion is supported by observations that when porcine sperm populations are activated by bicarbonate [84], a stimulator of adenylate cyclase and hence protein kinase A, heterogeneous responses are seen both within single semen samples and between individual boars [85,86] (Fig. 1.1). Subpopulations of boar spermatozoa respond to bicarbonate in different ways; some are quiescent in the absence of bicarbonate but are rapidly stimulated to maximal progressive motility, some are refractory to stimulation, and others lie somewhere in between.

What provides energy for sperm movement

Fig. 1.1. Bicarbonate-induced motility stimulation in boar spermatozoa. Washed spermatozoa from three different boars were incubated at 38°C in a bicarbonate-free HEPES-buffered Tyrode’s-based medium. After 10 min incubation, 15 mM bicarbonate/5% CO2 was added to one-half of the sample, while the other half received 15 mM NaCl (as control treatments). Sperm samples were video-recorded and their motion analyzed using a Hobson Sperm Tracker (Hobson Vision Systems, the United Kingdom). Left-hand panels show that in the absence of bicarbonate, most spermatozoa (points represent individual spermatozoa) moved slowly and nonlinearly (low linearity and average path velocity), but 2 min after bicarbonate addition (right-hand panels), most spermatozoa exhibited significantly increased linearity and velocity. However, while the velocities of most spermatozoa from boar 1 cluster above 50 μm s− 1, many of those from boar 2 remain below this threshold. Boar 3 showed the most dramatic bicarbonate response (for more detailed protocols, see [86,128]). Such differential stimulation may be significant in sperm selection mechanisms.

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/B9780128125717000022

Echinoderms, Part B

Hussein Hamzeh, ... U. Benjamin Kaupp, in Methods in Cell Biology, 2019

4.1 Stroboscopic imaging

Sperm motility and navigation in a chemical gradient have been studied using caged cGMP and caged resact in combination with stroboscopic techniques allowing visualization of the rapidly beating flagellum (Fig. 10A). Motility in 2D has been mostly studied on sperm swimming in shallow recording chambers. Interaction of sperm with the glass surface of the chamber can generate Ca2 + signals that alter motility or interfere with swimming. Therefore, we used the surfactant Pluronic F-127 (0.5%) that prevents sperm sticking to the glass surface.

What provides energy for sperm movement

Fig. 10. Motility of A. punctulata sperm. (A) Superposition of several images of a sperm cell while swimming on a straight (left) or curved path (right). Pictures were acquired using dark-field microscopy and aligned to the long axis of the ellipsoid-shaped head (dotted line). (B) Drawing of the experimental setup used to generate chemical gradients upon uncaging. Photolyzing light emanating from a UV-LED is collected by a liquid light guide and then collimated and focused onto the sample using two UV-transparent lenses. Light enters the microscope via the epifluorescence port. For calibration, a thin fluorescein sample is used. The fluorescence is filtered using a long-pass filter and monitored with a camera for adjustments. (C) Swimming path of sea urchin sperm in a shallow observation chamber (red; left). The [Ca2 +]i was imaged using stroboscopic cyan light and the fluorescent Ca2 + indicator Fluo-4. A chemoattractant gradient is generated by release of resact from its caged derivative (UV profile shown in blue shades). During navigation, the path curvature oscillates, and sperm swim on drifting circles up the chemical gradient. The variation of the green fluorescence reflects oscillations of [Ca2 +]i (green). Comparison of changes in [Ca2 +]i (green; right top) and curvature of the swimming path (white). Comparison of the time derivative of [Ca2 +]i (yellow; right bottom) with the curvature of the swimming path (white). (D) Swimming path of a sperm cell swimming in a chemical gradient established by photolysis of caged chemoattractant. The concentration gradient is symmetric by rotation around the axis shown by a vertical gray line. Time is indicated in false colors along the path. Sperm display two stereotypical behaviors: smooth steady bending of the helical axis and abrupt turns (red arrowheads) that result from swimming up and down the gradient, respectively.

Panels (A) and (C) are adapted from Kaupp, U.B., & L. Alvarez. (2016). Sperm as microswimmers—Navigation and sensing at the physical limit. The European Physical Journal Special Topics, 225, 2119–2139. Panel (D) is adapted from Jikeli, J.F., Alvarez, L., Friedrich, B.M., Wilson, L.G., Pascal, Colin, R., et al. (2015). Sperm navigation along helical paths in 3D chemoattractant landscapes. Nature Communications, 6, 7985.

The sperm flagellum beats about 50 times per second. Imaging such rapidly moving objects requires very brief exposure times that can be accomplished by short light pulses produced by LED light sources (stroboscopic illumination). Using this technique, it has been possible to decipher the steering mechanism of sea urchin sperm (Fig. 10A). Specifically, the beating flagellum adopts different waveforms allowing for straight swimming or swimming along an arc. Sperm use different flagellar waveforms for navigation in a gradient.

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/S0091679X1830181X

Sperm Competition in Mammals

Montserrat Gomendio, ... Eduardo R.S. Roldán, in Sperm Competition and Sexual Selection, 1998

(b) Sperm motility and sperm morphology

Good sperm motility in general is very important for active swimming along certain sections of the female tract and for penetration of physical barriers, such as the uterotubal junction and ova vestments. The percentage of motile sperm is one of the main factors lnfluenclng fertility rates (Drobnis and Overstreet 1992).

Sperm motility and sperm morphology are intricately linked because morphologically abnormal sperm swim more slowly or less efficiently (Katz et al. 1982) and are thus selected against at various levels: cervical mucus (Katz et al. 1989), uterotubal junction (Krzanowska 1974), oviduct and ova vestments (Krzanowska and Lorenc 1983; Meistrich et al. 1994). Thus, the percentage of morphologically normal sperm is also a good predictor of fertility rates in humans both in vivo and in vitro (Fig. 16.5; Liu and Baker 1994; Mortimer 1994).

What provides energy for sperm movement

Fig. 16.5. Correlation between percentage normal morphology of sperm in the insemination medium and bound to the zona pellucida (ZP).

From D. Y. Liu and H. W. G. Baker (1994) Male Factor in Human Infertility (ed.J. Tesarik), pp. 169–185, Ares-Serono Symposia Publications, Rome, with permission.Copyright © 1994

Recent evidence suggests that even some morphologically normal spermatozoa may swim inefficiently and in these cases morphologically normal sperm may be unable to fertilize. Morales et al. (1988) found that human patients suffering from infertility not only had a lower proportion of normal sperm, but also their normal sperm were less likely to be motile and swam less efficiently than normal donor sperm.

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/B9780121005436500416

Male Infertility

Paul J. Turek, in Yen & Jaffe's Reproductive Endocrinology (Seventh Edition), 2014

Oligospermia-Asthenospermia

Low sperm motility is observed in 25% of abnormal semen analyses; an isolated low sperm concentration is much less common. Low sperm motility may be due to antisperm antibodies (>50% of sperm bound) or excessive white blood cells in the ejaculate (leukocytospermia), the latter resulting in overproduction of reactive oxygen species that can damage sperm.21 Because “round cells” in the infertile ejaculate are more commonly immature germ cells (65%) rather than leukocytes,22 special leukocyte stains are recommended before treatment. Semen cultures are uninformative in asymptomatic infertile men with leukocytospermia as 83% are normally positive with multiple organisms.23 It is important to evaluate sexually transmitted diseases, penile discharge, prostatitis, or epididymitis. An expressed prostatic secretion is examined for leukocytes, and urethral cultures for Chlamydia and Mycoplasma can be considered. As a reference, the prevalence rate for PCR-based Chlamydia antigen detection is 0.3% among infertile men in general.24 Although the findings vary by both methodology and laboratory, in general, more than 50% of sperm bound with antibodies is considered significant and merits treatment.

Low sperm concentration may be due to an endocrinopathy such as prolactinoma, varicocele, or genetic causes, as outlined in Figure 24.4. Although eternally debated, there is substantial evidence to support the role of varicocele in male infertility (Table 24.3). Increasingly, genetic abnormalities should be considered in men with sperm concentrations less than 5 million/mL.25 Deletion of regions on the Y chromosome (microdeletions) occur in 6% of men with severely low sperm counts and in 15% of men with no sperm counts. In addition, 2% of men with low counts and 15% to 20% of men with no sperm counts will harbor chromosomal abnormalities detected by cytogenetic analysis (karyotype). These conditions include Klinefelter syndrome and translocations of non-sex chromosomes. Box 24.3 outlines current indications for genetic testing of infertile males. Importantly, varicocele repair for oligospermia in the setting of positive genetic findings is unlikely to improve semen quality or result in natural pregnancy.26

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/B978145572758200024X

Assisted Reproductive Technologies and Genetic Modifications in Rats

Yuksel Agca, John K. Critser, in The Laboratory Rat (Second Edition), 2006

ii. Flow Cytometric Sperm Analysis.

Although sperm motility and morphology are the most commonly used parameters for the assessment of sperm viability, neither is an accurate indicator of function. Spermatozoa gross morphology is insufficient to detect damage to the plasma membrane, mitochondria, and acrosome. Flow cytometric analysis using specific fluorescent probes is a valuable method of determining these structural changes that may hamper normal fertilization and subsequent embryo development. This method provides a rapid and accurate means of determining the functional status of large numbers of spermatozoa.

Gravance et al. (2001) used flow cytometric analysis of JC-1 staining patterns of rat spermatozoa to detect chemical-induced alterations of sperm mitochondrial membrane potential. They found that the mid-piece (mitochondrial location) of live, highly motile spermatozoa stained bright orange, whereas the mid-piece of live, non-motile spermatozoa stained green. The mid-piece of slightly or non-progressively motile spermatozoa stained a faint orange-green. Gravance et al. (2003) were able to assess frozen-thawed rat sperm viability using SYBR-14 and propidium iodide. Sperm mitochondria were differentially labeled with JC-1. Motile sperm stained with JC-1 appeared orange in the mid-piece, indicating a high mitochondrial membrane potential, whereas immotile sperm with a low membrane potential stained green.

View chapterPurchase book

Read full chapter

URL: https://www.sciencedirect.com/science/article/pii/B9780120749034500108

Evaluation of the Fertility of Breeding Males

Michael McGowan, in Veterinary Reproduction and Obstetrics (Tenth Edition), 2019

Assessment of Sperm Motility

The assessment of sperm motility should be done within a couple of minutes of semen collection whenever possible. In the event that semen cannot be assessed immediately, then it should be appropriately extended and stored. For those species that produce highly concentrated semen (ram, bull), a small drop of semen is often first placed on a warm slide and examined directly using the 4x lens (with the condenser turned down) for evidence of mass activity (often scored from no swirling wave to rapid swirling wave on a 1–5 or 1–10 scale). This examination enables rapid determination of whether the sample collected is consistent with the findings from examination of the scrotal contents (i.e., is representative of what the male would ejaculate if mated). However, it is important to note that mass activity is a function of sperm concentration and percentage motile sperm, and when samples have low sperm concentration, typical of some species and in bulls and rams due to variable response to electroejaculation, a swirl may not be observed even though the sample contains a high proportion of motile sperm. Further, observing a swirl does not allow the veterinarian to be able to always conclude that the sample contains a high proportion of progressively motile sperm, as a swirl can be observed in samples containing a high proportion of sperm swimming backwards due to the distal-midpiece reflex defect or looped tails.

To assess the percentage of progressively motile sperm, a small drop (~3 mm in diameter) of semen is placed on a prewarmed slide, and a warm coverslip is placed over it. It is critical that the droplet is small enough to ensure that a single layer of sperm can be observed. If the microscope being used only has bright field capacity, then the condenser must be turned down, the light reduced, and the 10x or 40x lens used. If the semen sample is quite concentrated, then it is advisable to dilute a drop of semen with several drops of either phosphate buffered saline or 2.9% sodium citrate solution on a warm slide, and then draw up a drop of the diluted semen and make a wet mount preparation as described previously. It is very important that the diluent is in-date and has been appropriately stored; otherwise, the pH or osmolality may have changed, and this can negatively bias the estimate of percentage progressively motile sperm. Some experience is required to accurately estimate the percentage progressively motile sperm. The veterinarian should focus on estimating the proportion of sperm that are not moving forward and initially broadly categorise the sample as having poor, fair, or good progressive motility (< 30%, 30%–59%, and ≥ 60%, respectively). The ‘golden rule’ when assessing sperm motility is, if in doubt, think ‘there might be a problem with how I have managed the sample, conducted the evaluation or, there could be a problem with the sample itself or the bull’. Therefore, if the mass activity or percentage progressively motile sperm is inconsistent with the findings of the physical examination of the scrotal contents, examine one or more additional droplets/wet mount preparations and/or collect another sample of semen. Also, sometimes with males who are examined out of season or have not been serving, the first sample/ejaculate may contain a relatively high number of dead sperm. In these cases the motility observed in the second sample is usually much better than initially observed.

What provides energy for sperm movement in mammals?

Flagellar dyneins drive sperm motility, which accounts for the consumption of high amounts of ATP. The two main ATP-producing metabolic pathways are compartmentalized in sperm: oxidative phosphorylation in the midpiece and glycolysis in the principal piece.

What provides energy for movement?

The source of energy that is used to power the movement of contraction in working muscles is adenosine triphosphate (ATP) – the body's biochemical way to store and transport energy. However, ATP is not stored to a great extent in cells. So once muscle contraction starts, the making of more ATP must start quickly.