ISSN: 1734-1922
Archives of Medical Science Special Issues
Current issue Archive Archives of Medical Science
1/2009
 
Share:
Share:

Invited review
The role of insulin and leptin in male reproduction

Fanuel Lampiao
,
Ashok Agarwal
,
Stefan S. du Plessis

Arch Med Sci 2009; 5, 1A: S48–S54
Online publish date: 2009/06/10
Article file
- The role of insulin.pdf  [0.12 MB]
Get citation
 
 

Introduction
A growing body of research has been focusing on obesity and its pathophysiology. Obesity is a cardinal feature of metabolic syndrome, a condition characterized by a group of abnormalities that also includes dyslipidemia, hypertension, and impaired glucose metabolism. In reproductive biology, metabolic syndrome has garnered considerable attention because of the connection that exists between diabetes mellitus (DM), hyperleptinemia, and infertility. Infertility is a common phenomenon in modern societies, affecting an estimated 15% of couples attempting to conceive who are not able to do so within one year. Male factors are believed to play a role in 20 to 50% of infertility cases [1].
Diabetes mellitus is characterized by poor glucose control leading to hyperglycemia. The two types of DM are type I DM, or insulin-dependent diabetes mellitus (IDDM), a condition characterized by an absolute or relative lack of insulin due to autoimmune destruction of the insulin secreting b-cells in the islets of Langerhans in the pancreas; and type II DM, non-insulin dependent diabetes mellitus (NIDDM), characterized by cellular insulin insensitivity despite sufficient insulin levels [2]. Both types of DM are well recognized as a cause of sexual dysfunction, which in turn also contributes to infertility [3]. Diabetes mellitus is thought to affect male reproductive function at multiple levels due to its effects on the endocrine control of the spermatogenesis process and spermatogenesis itself, as well as impairing penile erection and ejaculation [4]. Many studies involving diabetic animal models have demonstrated an impairment of sperm quality [5, 6], which leads to a reduction in fecundity [6-9]. Furthermore, researchers have reported that men affected with IDDM have sperm with severe structural defects, significantly lower motility [10] and lower ability to penetrate zona free hamster eggs [11]. In recent years, the incidence of NIDDM has increased due to an increase in obesity [3]. An increase in the prevalence of DM will pose a significant problem to human fertility.
Obese individuals also are reported to have higher circulating leptin levels as well as a higher prevalence of infertility [12, 13] than non-obese individuals. Leptin is a 16-kDa protein that is produced mainly by adipose tissue and encoded by the ob gene [14]. It also is produced by the placenta [15], stomach [16], and skeletal muscles [17]. Leptin’s tertiary structure resembles that of cytokines and lactogenic hormones [18]. Leptin is best known as a regulator of food intake and energy expenditure via hypothalamic-mediated effects [19]. An increasing body of data suggests that leptin also acts as a metabolic and neuroendocrine hormone. It is involved in glucose metabolism as well as in normal sexual maturation and reproduction [20]. Thus, changes in plasma leptin concentrations can have important and wide-ranging physiological implications. This review aims to highlight the roles of both insulin and leptin in male reproduction as well as focus on their possible effects at various reproductive levels that contribute to male infertility.
Endocrine effects of insulin on male reproduction
The importance of insulin has been demonstrated in male rat reproduction by using streptozotocin to deplete the b-cells of the pancreas, thereby inducing IDDM [7]. Insulin deficiency in these rats led to a decrease in Leydig cell number as well as an impairment in Leydig cell function. This consequently translated to a decrease in androgen biosynthesis and serum testosterone levels. The impaired Leydig cell function and subsequent decrease in testosterone in IDDM could be explained by the absence of the direct stimulatory effects of insulin on Leydig cells, as well as by an insulin-dependent decrease in FSH and LH levels [17].
It also has been reported [10] that insulin plays a central role in regulation of the hypothalamic-pituitary-testicular axis by the reduction in secretion of LH and FSH in diabetic men, as well as in knockout mice lacking the insulin receptor in the hypothalamus. Both the diabetic men and the knockout mice had notably impaired spermatogenesis, increased germ cell depletion, and Sertoli cell vacuolization [10, 21]. Figure 1 show that insulin is required to stimulate the hypothalamus to release gonadotrophin releasing hormone (GnRH), which instructs the release of LH and FSH from the pituitary gland. Higher insulin concentrations, such as those found in NIDDM, have been reported to lead to hypogonadism [22] as well as decreased serum testosterone levels [23]. Furthermore, Pitteloud et al. [24] also reported than insulin resistance lead to a decrease in testosterone secretion at the testicular level (Leydig cell) that was not due to changes in hypothalamic or pituitary function. These findings point to a direct action of insulin at the gonadal level (see Figure 1).
Endocrine effects of leptin on male reproduction
Three leptin receptor isoforms have been reported to be present in gonadal tissue, suggesting that leptin could exert a direct endocrine action on the gonads [25-27]. Indeed, studies have shown that treatment of infertile ob/ob knockout mice with leptin restored reproductive ability [28]. Injecting these ob/ob mice with leptin reportedly caused an elevation in FSH levels and also stimulated gonadal development [29]. Chronic administration of anti-leptin antibody to rats was shown to inhibit LH release [30].
Humans deficient in leptin exhibit effects similar to those observed in animal models. A case study regarding a male with a homozygous leptin mutation reported that he was still pre-pubertal and showed clinical traits typical of hypogonadism and androgen deficiency despite being 22 years of age [31]. Another male subject with a leptin receptor deficiency reportedly showed no pubertal development at either 13 or 19 years of age [32]. Reports like these emphasize the importance of leptin in the onset of puberty in humans.
The mechanisms through which leptin acts are not clearly elucidated as yet but probably involve the hypothalamus and its subsequent effects on the pituitary and gonadal axis. Administration of GnRH to leptin-deficient men has been shown to induce a normal increase in serum LH and FSH levels, while the administration of gonadotrophins increased testosterone levels [31]. As illustrated in Figure 1, this effect may be the result of leptin stimulating GnRH synthesis or secretion from the hypothalamic neurons or secretion of gonado-trophins by the pituitary gland [33].
Effects of insulin on spermatogenesis
Morphological abnormalities have been reported in IDDM human testicular biopsies. These abnormalities included increasing tubule-wall thickness, germ cell depletion and Sertoli cell vacuolization [34]. Morphological and functional spermatozoal abnormalities that have been observed in diabetic animal models appear to be reversible with insulin administration [35, 36]. A significantly lower sperm count and epididymal sperm motility were reported in diabetic rats in comparison to controls [36]. In vitro insulin administration to these retrieved epididymal spermatozoa restored their motility to that of normal levels, suggesting a direct effect on spermatozoa due to defective carbohydrate metabolism. Studies have reported that insulin as well as insulin-like growth factor I (IGF-I) and IGF-II promote the differentiation of spermatozoa into primary spermatocytes by binding to the IGF-I receptor [37]. Evidence also suggests that both the sperm membrane and the acrosome represent cytological targets for insulin [38].
Effects of leptin on spermatogenesis
The importance of leptin during the process of spermatogenesis was demonstrated by the observation that a leptin deficiency in mice was associated with impaired spermatogenesis, increased germ cell apoptosis, and up-regulated expression of pro-apoptotic genes within the testes [39]. This resulted in a reduction in germ cell numbers and the absence of mature spermatozoa in the seminiferous tubules. This finding adds further support to the importance of physiological leptin levels in the normal production of male gametes.
Insulin and ejaculated spermatozoa
Insulin has been shown to play a central role in the regulation of gonadal function; however, its significance in male fertility is not completely understood and properly elucidated [40]. Until recently, insulin was thought to be produced only by the b-cells in the pancreas of adult mammals [41].
Newer studies, however, have demonstrated that insulin is expressed in and secreted by human ejaculated spermatozoa. Both insulin mRNA as well as the actual protein were detected in ejaculated human sperm [41]. Capacitated spermatozoa were found to secrete more insulin than noncapacitated spermatozoa [41], suggesting a possible role for insulin in sperm capacitation.
Our group, furthermore, has shown the importance of insulin on ejaculated human spermatozoa in vitro [42]. Insulin administration to the medium (10 mIU) was found to significantly increase total and progressive motility and enhance hyperactivation characteristics (VCL and ALH) significantly. In vitro insulin administration also led to an increase in spontaneous acrosome reaction, as well as enhanced sensitivity to the progesterone-induced acrosome reaction. Whether this increase was due to the agonists’ effect on capacitation or the acrosome reaction itself is unclear. Our group also demonstrated that insulin increased nitric oxide (NO) production in human spermatozoa, possibly via the phosphoinositide 3-kinase (PI3K) signaling pathway as evidenced by the reduction in NO production when the PI3K inhibitor wortmannin was administered. Insulin may play a role in enhancing the fertilization capacity of human spermatozoa by increasing motility, NO production and acrosome reaction sensitivity [42].
Leptin and ejaculated spermatozoa
Despite the fact that leptin has been implicated in the regulation of reproduction in humans and animal models and that its specific role in the female reproductive system has been well established, its exact role (s) in the male reproductive system remains to be clarified [43, 44]. Leptin expression in ejaculated human spermatozoa has been demonstrated by identifying its transcripts by means of reverse transcription-polymerase chain reaction; its protein presence was evidenced by Western blot analysis and its localization by immunostaining techniques [45].
The significance of leptin in male reproduction will remain ambiguous for at least a while as results from studies are quite controversial and contradictory. Some studies have indicated positive effects [46], whereas others have reported negative effects of leptin on gonadal function [47]. Seminal plasma leptin levels have been shown to be significantly lower in normozoospermic patients compared with pathological semen samples, and higher leptin levels have shown a negative correlation with sperm function [48]. Conversely, other reports show no correlation between leptin levels and sperm motility or morphology [49]. Capacitated spermatozoa were reported to secrete more leptin than noncapacitated spermatozoa, suggesting that leptin plays a role in the process of capacitation [45]. Moreover, leptin receptors were detected by immunohistochemistry in ejaculated spermatozoa and were localized on the tail area [50]. Similar to what we observed with insulin, our group has demonstrated that in vitro leptin administration increased various motility parameters and NO production and also increased the sensitivity of spontaneous and progesterone-induced acrosome reactions [42].
GLUT8 as a glucose transporter in human spermatozoa
Glucose uptake and metabolism are essential for cell proliferation and survival and usually is carried out through glucose transporters (GLUTs). In mammals there are 14 known members of GLUT proteins [51]. Insulin regulation of glucose transport in target tissues is known to involve the specialized GLUT4 isoforms, which are localized only in insulin-responsive tissues [51].
Glucose metabolism is recognized as essential for germ cell fertility, and disruptions to it such as those occurring in DM are known to impair spermatogenesis, causing infertility [10, 11]. Until recently, the assumption was that GLUT5 was the major sugar transporter in the sperm cell [52]. However, researchers now have shown that GLUT5 is a very specific fructose transporter [53] and does not transport glucose to a significant extent. Because GLUT5 was not detected in rat testis, other sugar transporters, presumably GLUT3, have been suggested for catalyzing the fuel supply of the rat sperm cell [54]. In recent years, a novel 447-amino-acid glucose transporter protein, GLUT8 has been described [55-57]. GLUT8 is expressed to some extent in insulin-sensitive tissues, e. g., brain, adrenal gland, spleen, adipose tissue, muscle, heart, and liver [55, 56, 58]. GLUT8 mRNA expression was determined to be highest in testicular tissue and linked to circulating gonadotrophin levels [56, 59].
GLUT8 was found to be located specifically in the head of mouse and human spermatozoa predominantly within the acrosome of mature sperm [60]. Coincidentally, immunohistochemical studies have shown that insulin also is located predominantly in these areas of human spermatozoa [38]. The intracellular localization of GLUT8 is similar to that of the insulin-sensitive glucose transporter GLUT4, and it has indeed been suggested that insulin could produce a trans-location of GLUT8 to the plasma membrane of the blastocyst [57]. In addition, GLUT8 has been shown to recycle in a dynamic-dependent manner between internal membranes and the plasma membrane in rat adipocytes and COS-7 cells [61]. As illustrated in Figure 2, both insulin and leptin stimulation converges at the level of PI3K during the intracellular signaling pathway. PI3K activation leads to protein kinase B (PKB/Akt) phospho-rylation, which in turn causes GLUT8’s translocation and insertion into the cell membrane. This allows increased glucose uptake, fueling glucose metabolism necessary for increased motility and the acrosome reaction. Simultaneously the PI3K and PKB/Akt pathway activated by insulin and leptin also can diverge and stimulate the endothelial nitric oxide synthase (eNOS) enzyme of spermatozoa to increase NO generation NO’s ability to increase sperm motility and acrosome reaction also has been demonstrated [62]. Therefore, we hypothesize that insulin and leptin can act via two possible methods (GLUT8 translocation; NO production) to influence human sperm motility and acrosome reaction.
In conclusion, insulin levels, leptin levels, and male infertility are associated. Decreased insulin levels have been shown to exert adverse effects on reproductive endocrine function and gonadal function, as well as on ejaculated spermatozoa function (Table I). On the other hand, decreased leptin levels negatively affect the male’s reproductive capacity by delaying puberty; higher leptin levels have been reported to correlate negatively with human sperm function (Table I).
Insulin and leptin concentrations are a double-edged sword, and a proper balance must be struck for normal reproductive function. Insulin and leptin impairment due to pathologies such as DM, obesity, and metabolic syndrome explain why infertility is connected to these conditions. Despite the fact that the relationship between obesity, metabolic syndrome, DM, and male infertility has been established, the exact mechanisms by which they act have not been elucidated to the fullest. This brief review has focused only on two hormones i.e., insulin and leptin, that possibly can be implicated under these conditions. Further studies are needed not only to tease out the exact roles each plays, but also to help find possible in vivo and in vitro solutions and treatment regimes for male infertility patients.
References
1. Sharlip ID, Jarow JP, Belker AM, et al. Best practice policies for male infertility. Fertil Steril 2002; 77: 873-82.
2. Atkinson MA, Maclaren NK. The pathogenesis of insulin-dependent diabetes mellitus. N Engl J Med 1994; 331: 1428-36.
3. Agbaje IM, Rogers DA, McVicar CM, et al. Insulin dependent diabetes mellitus: implications for male reproductive function. Hum Reprod 2007; 22: 1871-7.
4. Sexton WJ, Jarow JP. Effects of diabetes mellitus upon male reproductive function. Urology 1997; 49: 508-13.
5. Amaral S, Moreno AJ, Santos MS, Seica R, Ramalho-Santas J. Effects of hyperglycemia on sperm and testicular cells of Goto-Kakizaki and streptozotocin-induced rat model for diabetes. Theriogenology 2006; 66: 2056-67.
6. Scarano WR, Messias AG, Oliva SU, Klinefelter GR, Kempinas WG. Sexual behaviour, sperm quantity and quality after short-term streptozotocin-induced hyperglycaemia in rats. Int J Androl 2006; 29: 482-8.
7. Murray FT, Cameron DF, Orth JM. Gonadal dysfunction in the spontaneously diabetic BB rat. Metabolism 1983; 32 (7 Suppl 1): 141-7.
8. Cameron DF, Rountree J, Schultz, et al. Sustained hyperglycemia results in testicular dysfunction and reduced fertility potential in BBWOR diabetic rats. Am J Physiol 1990; 259: E881-9.
9. Ballester J, Munoz MC, Dominguez J, Rigau T, Guinovart JJ, Rodríguez-Gil JE. Insulin-dependent diabetes affects testicular function by FSH- and LH-linked mechanisms. J Androl 2004; 25: 706-19.
10. Baccetti B, La Marca A, Piomboni P, et al. Insulin-dependent diabetes in men is associated with hypothalamo-pituitary derangement and with impairment in semen quality. Hum Reprod 2002; 17: 2673-7.
11. Shrivastav P, Swann J, Jeremy JY, Thompson C, Shaw RW, Dandona P. Sperm function and structure and seminal plasma prostanoid concentrations in men with IDDM. Diabetes Care 1989; 12: 742-4.
12. Mantzoros CS. The role of leptin in human obesity and disease: a review of current evidence. Ann Intern Med 1999; 130: 671-80.
13. Kasturi SS, Tannir J, Brannigan RE. The metabolic syndrome and male infertility. J Androl 2008; 29: 251-9.
14. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 1994; 372: 425-32.
15. Masuzaki H, Ogawa Y, Hosoda K, et al. Glucocorticoids regulation of leptin synthesis and secretion in humans: elevated plasma leptin levels in Cushing’s syndrome. J Clin Endocrinol Metab 1997; 82: 2542-7.
16. Bado A, Levasseur S, Attoub S, et al. The stomach is a source of leptin. Nature 1998; 394: 790-3.
17. Wang MY, Zhou YT, Newgard CB, Unger RH. A novel leptin receptor isoform in rat. FEBS Letters 1998; 392: 87-90.
18. Zabeau L, Lavens D, Peelman F, Eyckerman S, Vandekerckhove J, Tavernier J. The ins and outs of leptin receptor activation. FEBS Letters 2003; 546: 45-50.
19. Schwartz MW, Baskin DG, Kaiyala KJ, Woods SC. Model for the regulation of energy balance and adiposity by the central nervous system. Am J Clin Nutr 1999; 69: 584-96.
20. Wauters M, Considine RV, Van Gaal LF. Human leptin: from an adipocyte hormone to an endocrine mediator. Eur J Endocrinol 2000; 143: 293-311.
21. Brüning JC, Gautam D, Burks DJ, et al. Role of brain insulin receptor in control of body weight and reproduction. Science 2000; 289: 2122-5.
22. Barrett-Connor E, Khaw KT, Yen SS. Endogenous sex hormone levels in older adult men with diabetes mellitus. Am J Epidemiol 1990; 132: 895-901.
23. Dhindsa S, Prabhakar S, Sethi M, Bandyopadhyay A, Chaudhuri A, Dandona P. Frequent occurrence of hypogonadotropic hypogonadism in type 2 diabetes. J Clin Endocrinol Metab 2004; 89: 5462-8.
24. Pitteloud N, Hardin M, Dwyer AA, et al. Increasing insulin resistance is associated with decrease in Leydig cell testosterone secretion in men. J Clin Endocrinol Metab 2005; 90: 2636-41.
25. Cioffi JA, Shafer AW, Zupancic TJ, et al. Novel B219/OB receptor isoforms: possible role of leptin in hematopoiesis and reproduction. Nat Med 1996; 2: 585-9.
26. Cioffi JA, Van Blerkom J, Antczak M, Shafer A, Wittmer S, Snodgrass H. The expression of leptin and its receptors in pre-ovulatory human follicles. Mol Hum Reprod 1997; 3: 467-72.
27. Karlsson C, Lindell K, Svensson E, et al. Expression of functional leptin receptors in the human ovary. J Clin Endocrin Metab 1997; 82: 4144-8.
28. Mounzih K, Lu R, Chehab FF. Leptin treatment rescues the sterility of genetically obese ob/ob males. Endocri-nology 1997; 138: 1190-3.
29. Barash IA, Cheung CC, Weigle DS, et al. Leptin is a metabolic signal to the reproductive system. Endocrinology 1996; 137: 3144-7.
30. Carro E, Pinilla L, Seoane LM, et al. Influence of endogenous leptin tone on the estrous cycle and luteinizing hormone pulsatility in female rats. Neuroendocrinology 1997; 66: 375-7.
31. Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD. A leptin missence mutation associated with hypogonadism and morbid obesity. Nat Genet 1998; 18: 213-5.
32. Clément K, Vaisse C, Lahlou N, et al. A mutation in a human leptin receptor gene causes obesity and pituitary dysfunction. Nature 1998; 392: 398-401.
33. Yu W, Kimura M, Walczewska A, Karanth S, McCann S. Role of leptin in hypothalamic-pituitary function. Proc Natl Acad Sci U S A 1997; 94: 1023-8.
34. Cameron DF, Murray FT, Drylie DD. Interstitial compartment pathology and spermatogenic disruption in testes from impotent diabetic men. Anat Rec 1985; 213: 53-62.
35. Howland BE, Zebrowski EJ. Some effects of experimentally-induced diabetes on pituitary-testicular relationships in rats. Horm Metab Res 1976; 8: 465-9.
36. Seethalakshmi L, Menon M, Diamond D. The effects of streptozotocin-induced diabetes on the neuro-endocrine-male reproductive tract axis of the adult rat. J Urol 1987; 138: 190-4.
37. Nakayama Y, Yamamoto T, Abe SI. IGF-I, IGF-II and insulin promote differentiation of spermatogonia to primary spermatocytes in organ culture of newt testes. Int J Dev Biol 1999; 43: 343-7.
38. Silvestroni L, Modesti A, Sartori C. Insulin-sperm interaction: effects on plasma membrane and binding to acrosome. Arch Androl 1992; 28: 201-11.
39. Bhat GK, Sea TL, Olatinwo MO, et al. Influence of leptin deficiency on testicular morphology, germ cell apoptosis, and expression levels of apoptosis-related genes in the mouse. J Androl 2006; 27: 302-10.
40. Aquila S, Gentile M, Middea E, Calatano S, Ando` S. Autocrine regulation of insulin secretion in human ejaculated spermatozoa. Endocrinology 2005; 146: 552-7.
41. Throsby M, Homo-Delarche F, Chevenne D, Goya R, Dardenne M, Pleau JM. Pancreatic hormone expression in the murine thymus: localization in dendritic cells and macrophages. Endocrinology 1998; 139: 2399-406.
42. Lampiao F, Du Plessis SS. Insulin and leptin enhance human sperm motility, acrosome reaction and nitric oxide production. Asian J Androl 2008; 10: 799-807.
43. Camin~a JP, Lage M, Menendez C, et al. Evidence of free leptin in human seminal plasma. Endocrine 2002; 17: 169-74.
44. Fietta P. Focus on leptin, a pleiotropic hormone. Minerva Med 2005; 96: 65-75.
45. Aquila S, Gentile M, Middea E. Leptin secretion by human ejaculated spermatozoa. J Clin Endocrinol Metab 2005; 90: 4753-61.
46. Caprio M, Fabbrini E, Isidori AM, Aversa A, Fabbri A. Leptin in reproduction. Trends Endocrinol Metab 2001; 12: 65-72.
47. Clarke IJ, Henry BA. Leptin and reproduction. Rev Reprod 1999; 4: 48-55.
48. Glander HJ, Lammert A, Paasch U, Glasow A, Kratzsch J. Leptin exists in tubuli seminiferi and in seminal plasma. Andrologia 2002; 34: 227-33.
49. Zorn B, Osredkar J, Meden-Vrtovec H, Majdic G. Leptin levels in infertile male patients are correlated with inhibin B, testosterone and SHBG but not with sperm characteristics. Int J Androl 2007; 30: 439-44.
50. Jope T, Lammert A, Kratzsch J, Paasch U, Glander HJ. Leptin and leptin receptor in human seminal plasma and in human spermatozoa. Int J Androl 2003; 26: 335-41.
51. Yang J, Holman GD. Comparison of GLUT4 and GLUT1 subcellular trafficking in basal and insulinstimulated 3T3-L1 cells. J Biol Chem 1993; 268: 4600-3.
52. Burant CF, Takeda J, Brot-Laroche E, Bell GI, Davidson NO. Fructose transporter in human spermatozoa and small intestines is GLUT5. J Biol Chem 1992; 267: 14523-6.
53. Kane S, Seatter MJ, Gould GW. Functional studies of human GLUT5: effect of pH on substrate selection and an analysis of substrate interactions. Biochem Biophys Res Commun 1997; 238: 503-5.
54. Burant CF, Davidson NO. GLUT3 glucose transporter isoform in rat testis: localization, effect of diabetes mellitus, and comparison to human testis. Am J Physiol 1994; 267: 1488-95.
55. Ibberson M, Uldry M, Thorens B. GLUTX1, a novel mammalian glucose transporter expressed in the central nervous system and insulin-sensitive tissues. J Biol Chem 2000; 275: 4607-12.
56. Doege H, Schürmann A, Bahrenberg G, Brauers A, Joost HG. GLUT8, a novel member of the sugar transport facilitator family with glucose transport activity. J Biol Chem 2000; 275: 16275-80.
57. Carayannopoulos MO, Chi MM, Cui Y, et al. GLUT8 is glucose transporter responsible for insulin-stimulated glucose uptake in blastocyst. Proc Natl Acad Sci USA 2000; 97: 7313-8.
58. Reagan LP, Gorovits N, Hoskin EK et al. Localization and regulation of GLUTX1 glucose transporter in the hippocampus of streptozotocin diabetic rats. Proc Natl Acad Sci USA 2001; 98: 2820-5.
59. Scheepers A, Doege H, Joost HG, Schürmann A. Mouse GLUT8: genomic organization and regulation of expression in 3T3-L1 adipocytes by glucose. Biochem Biophys Res Commun 2001; 288: 969-74.
60. Schürmann A, Axer H, Scheepers A, Doege H, Joost H. The glucose transport facilitator GLUT8 is predominantly associated with the acrosomal region of mature spermatozoa. Cell Tissue Res 2002; 307: 237-42.
61. Lisinski I, Schürmann A, Joost HG, Cushman SW, Al-Hasani H. Targeting of GLUT6 (formerly GLUT9) and GLUT8 in rat adipose cells. Biochem J 2001; 358: 517-22.
62. Wu TP, Huang BM, Tsai HC, Lui MC, Liu MY. Effects of nitric oxide on human spermatozoa activity, fertilization and mouse embryonic development. Arch Androl 2004; 50: 173-9.
Copyright: © 2009 Termedia & Banach. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.