Skip to main content
  • Research Article
  • Open access
  • Published:

Regulation of Male Fertility by the Opioid System

Abstract

Endogenous opioid peptides are substances involved in cell communication. They are present in various organs and tissues of the male and female reproductive tract, suggesting that they may regulate some of the processes involved in reproductive function. In fact, the opioid system that operates as a multi-messenger system can participate in the regulation of reproductive physiology at multiple levels, for example, at the levels of the central nervous system, at the testes level and at sperm level. A better understanding of the implication of the opioid system in reproductive processes may contribute to clarifying the etiology of many cases of infertility and the effect of opiate abuse on fertility. Indeed, a novel biochemical tool for the diagnosis and treatment of male infertility could be based upon components of the opioid system. The presence of the opioid system in sperm cells also represents a novel opportunity for reproductive management, for either enhancing the probability of fertilization or reducing it through the development of novel targeted contraceptives.

The Opioid System

The opioid system is a biological communication system for which activity is mediated by the so-called endogenous opioid peptides (EOPs). These peptides are synthesized from the processing of their precursors, which are encoded by three different genes: pro-enkephalin (PENK), pro-opiomelanocortin (POMC) and pro-dynorphin (PDYN) (1). Each PENK molecule contains seven EOP amino acid sequences and mainly produces met- and leuenkephalin (2). The POMC precursor, which in part contains the β-endorphin peptide, is a precursor of adrenocorticotropin hormone (ACTH), α- and β-melanotropin (MSH), and α-, β- and γ-lipotropin (LPH), among others (3). Each molecule of PDYN contains three sequences of leuenkephalin and other larger peptides such as neo-endorphins and A and B dynorphins (2) (Figure 1).

Figure 1
figure 1

Control of male reproductive function by the opioid system at multiple levels. (1) At the level of the CNS, EOPs inhibit the secretion of GnRH, thereby suppressing the release of LH from the pituitary. (2) At the testes level, EOPs are synthesized mainly in Leydig cells after LH stimulation, and they exert an inhibitory effect on Sertoli cells. In particular, EOPs can regulate the levels of testosterone (T) indirectly, inhibiting the production of ABP that is stimulated by FSH in Sertoli cells. (3) Genes encoding opioid peptide precursors are differentially expressed in germ cells, and somatic cells of the testes and their transcripts are not efficiently translated in spermatogenic germ cells. (4) In spegrmatozoa, the opioid system regulates sperm motility in a distinct manner by the activation of distinct receptors.

EOPs exert their action through opioid receptors. Opioid receptors are G-protein-coupled receptors that are monomeric and exhibit seven transmembrane domains. There are three principal types of opioid receptors: the delta-opioid receptor (DOR), the muopioid receptor (MOR) and the kappa-opioid receptor (KOR) (4–7). More recently, the orphanin 1 (ORL1) receptor (also known as the nociceptin receptor) was discovered and found to share a large homology with opioid receptors (8,9) (Table 1).

Table 1 Profiles of affinity of opioid ligands on the basis of binding studies.

EOPs exhibit different affinities for these opioid receptors. Met- and leuenkephalin are considered to be endogenous ligands for DOR, since they have higher affinity for DOR. Nevertheless, they are capable of binding with lower affinity to the MOR. β-Endorphins bind to DOR and MOR with similar affinity, whereas dynorphins preferentially bind to KOR (10). Nociceptin, which is derived from the processing of the precursor pro-nociceptin (PNOC) (11), has been reported to be an endogenous ligand of the ORL1 receptor and has very weak activity at other opioid receptors (12). MOR exhibit high affinity for morphine, whereas KOR have preferential affinity for ketocyclazocine. Concerning antagonist selectivity, naloxone behaves as an antagonist at all opioid receptors, with highest affinity for MOR (10) (Table 1).

Opioid receptors are known to couple to G0 and Gi proteins, since signaling by these receptors is effectively blocked by pertussis toxin. Opioid signaling pathways involve the following: (a) inhibition of the adenylate cyclase enzyme (13), (b) inactivation of voltage-dependent calcium channels (14,15), (c) activation of potassium channels (16), (d) mobilization of intracellular Ca2+ reserves through the activation of phospholipase Cβ (17,18) and (e) stimulation of MAP kinase pathways (19).

The most well-known physiological effect associated with EOPs is their efficacy in pain reduction or analgesia, although their effect on a variety of other physiological functions has become apparent in recent years (20). In particular, evidence of the widespread presence of opioid peptides and receptors in different organs and tissues of the male reproductive system indicates that EOPs likely participate in the regulation of male reproductive function.

Hormonal Control of Reproductive Function By the Opioid System

Sterility is a problem that affects a large percentage of couples of reproductive age. It is estimated that in 40–45% of cases, the principal cause of sterility resides in the male partner. Anatomical defects, genetic diseases and injuries, as well as testicular sperm and hormonal dysfunction are major causes of male infertility.

The opioid system is one of a host of neuroendocrine products (dopamine, arginine vasopressin [AVP], corti-cotropin-releasing hormone [CRH], cholecystokinin [CCK] and more) that are involved in the control of releasing gonadotropin-releasing hormone (GnRH) and thus the sex hormones follicle-stimulating hormone (FSH) and luteinizing hormone (LH). Both FSH and LH are secreted by the anterior pituitary and act directly on the testes to stimulate the somatic cells that contribute to spermatogenesis. The synthesis and release of these hormones is controlled by GnRH, which is secreted by the hypothalamus. The FSH receptor is expressed exclusively in Sertoli cells (21). In contrast, the LH receptor is expressed mainly in Leydig cells but is also mildly expressed in spermatogenic cells (22,23). The main function of FSH is to stimulate the proliferation of Sertoli cells during puberty, whereas LH regulates the synthesis of testosterone in the adult testes (24). Administration of morphine is associated with a suppression of LH release, whereas opioid antagonists such as naloxone and naltrexone produce increased serum levels of LH in humans and many animal species (25,26). The opioid-induced inhibition of LH release is mediated by a hypothalamic action because, on the one hand, opioid receptors have been described in purified gonadotrophs and, on the other hand, in vivo treatment with GnRH antagonists completely blocked the release of GnRH induced by naloxone (27–29). Therefore, opioids generally decrease not only LH but also testosterone and estradiol, which have effects on testicular function (30,31). In fact, opioid abuse primarily leads to hypogonadism and includes decreased libido and erectile dysfunction in men and infertility (32). Administration of opioid antagonists such as naltrexone can improve symptoms of hypogonadism and improved erectile function, although the antagonist did not increase testosterone or LH levels, suggesting regulation at the central rather than the peripheral level (33).

These studies clearly indicate that EOPs can regulate reproductive function by inhibiting the secretion of GnRH at the level of the central nervous system (CNS) (26,28,34) (Figure 2).

Figure 2
figure 2

Opioid peptide precursor proteins: PENK (A), POMC (B) and PDYN (C). Opioid peptides are shown in red.

Regulation of Testicular Function by the OPIOID System

Numerous studies have demonstrated the presence of EOPs in different testicular cell types (35–37). In addition, binding studies have revealed the presence of all three types of opioid receptors (DOR, MOR and KOR) in rat testes. Moreover, the genes encoding the opioid precursors PENK, PDYN and POMC have also been found to be expressed in the testes of the rat and mouse (38,39), providing further evidence that the opioid system may play a role in regulating testicular function.

Spermatogenesis consists of a series of processes involving the proliferation and differentiation of developing germ cells into sperm. In addition to being regulated by endocrine mechanisms of the hypothalamic-pituitary axis, spermatogenesis is controlled by paracrine and autocrine processes that take place between different cell types present in the testicles.

Sertoli cells provide the structural and metabolic support that is necessary for the differentiation of germ cells. Dysfunctional FSH receptors lead to a decrease in the size of the testicles due to a decrease in the number of Sertoli cells (40). Dysfunctions that occur during spermatogenesis can lead to pathologies such as oligozoospermia and terato-zoospermia, although males may nevertheless be fertile (41).

Leydig cells are responsible for the production of testicular testosterone. Testosterone and its metabolites (dihydrosterone and estradiol) are male sex hormones that regulate reproductive function, since spermatogenesis depends on the presence of an adequate level of intratesticular testosterone (42). The concentration of testosterone in the seminiferous tubule is 100 times higher than in peripheral blood (43). These high intratesticular testosterone levels are achieved thanks to the LH-induced production of testosterone around the tubules in the Leydig cells and to the activity of androgen-binding protein (ABP), which is synthesized in Sertoli cells and which transports testosterone into the lumen of the seminiferous tubule (44).

As previously mentioned, binding studies found all three types of opioid receptors (DOR, MOR and KOR) in the rat testis, although subsequent higher-resolution localization studies found that these receptors are exclusively expressed by Sertoli cells (45). With regard to EOPs, the genes encoding the opioid precursors PENK, POMC and PDYN were found to be expressed in the rat and mouse testes (38,39,46), supporting the idea that testicular function could be regulated through local synthesis of EOPs. The finding that transgenic mice who overexpress PENK in the testes have impaired fertility, morphologically abnormal testicles and low sperm motility has provided further evidence suggesting that the opioid system participates in an important way in the regulation of testicular function (47).

In rats and mice, the PENK gene is differentially expressed in germ cells and somatic cells of the testes (48). Testicular germ cells express a large 1.7-kb transcript that is expressed under the control of a specific promoter (49), whereas Leydig cells, Sertoli cells and peritubular cells express a smaller 1.45-kb mRNA (39,48–50). Both transcripts contain an intact coding sequence and differ in the 5′ noncoding sequence (48). Polysome expression profiles indicate that the PENK transcript, which is specifically expressed in germ cells, is not translated efficiently, whereas the PENK transcript expressed by Leydig cells is efficiently translated. This result would explain why the concentration of PENK products detected in rat testes homogenates is relatively low (<2 pmol/g), compared with the levels of mRNA detected for the PENK gene (39). Moreover, in Sertoli cells exposed to FSH in vitro, the expression of the PENK gene, as well as levels of metenkephalin, were found to be upregulated (46), suggesting that not only Leydig cells, but also Sertoli cells, contribute to the de novo synthesis of EOPs in the testes. On the other hand, it was suggested that germ cells may also regulate levels of EOPs during spermatogenesis by controlling the expression of PENK in the surrounding cells; however, the underlying mechanism has not yet been characterized (51).

POMC gene expression was localized to Leydig cells (38) and to germ cells (46) by in situ hybridization. In the mouse testis, the POMC transcript has two different sizes. Leydig cells and some stages of germ cells expressed a long POMC transcript of about 675–750 nucleotides, but pachytene spermatozoa expressed a shorter POMC transcript. As occurs with PENK, the POMC transcript is efficiently transcribed in Leydig cells, so that it is likely to be responsible for synthesizing POMC-derived peptides in the testis. However, the small POMC transcript in pachytene spermatozoa was found to be inefficiently transcribed during spermatogenesis (46). These findings are consistent with the detection of β-endorphin immunoreactivity in Leydig cells in many species (37). The synthesis of β-endorphin is controlled by LH secreted by the pituitary in adult rats (52) and also by corticotropin-releasing factor (CRF). Leydig cells in culture exposed to CRF also produce higher levels of β-endorphin, and this effect is completely reversed in the presence of an α-helix CRF antagonist, confirming its specificity of action (53). CRF regulates the function of Leydig cells in an autocrine manner (54), since the levels of CRF increased in Leydig cells after LH treatment, and these cells also specifically expressed the corresponding receptor (55,56).

Thus, EOPs are mainly synthesized de novo by Leydig cells in the testis, although EOPs have no effect on testosterone production in cultured Leydig cells or in rat testicular tissue after human chorionic gonadotropin (hCG) treatment (37,57). However, intratesticular administration to rats of the opioid antagonist naloxone and nalmefene was found to decrease basal secretion of testosterone and reduce levels of serum testosterone (58). Binding studies have not detected opioid receptors in Leydig cells (45), which suggests that EOPs indirectly regulate the secretion of testosterone.

On the other hand, β-endorphins synthesized by Leydig cells produce an inhibitory paracrine effect on Sertoli cell function (58,59). Specifically, β-endorphins inhibit the production of ABP stimulated by FSH in Sertoli cells, and this inhibition is reversed in the presence of naloxone (36). ABP can regulate intratubular testosterone levels since, as mentioned before, it is responsible for testosterone transport into the lumen of the seminiferous tubule (44). Consequently, β-endorphin is involved in the control of the precise levels of testosterone that are required for proper spermatogenesis. Furthermore, β-endorphins have been reported to inhibit the proliferation and differentiation of Sertoli cells stimulated by FSH in the testes (60) and to alter the proliferative dynamics of Sertoli cells and enhance the levels of ABP after treatment of the testes of newborn rats with the opioid antagonist nalmefene (58). Taken together, these findings suggest that EOPs inhibit the development of Sertoli cells and thus may contribute to maintaining the quiescent sexual state of the testes before puberty (61).

Dynorphin A and B immunoreactivity was detected in testicular extracts using chromatography (62). The PDYN transcript in rat testes is smaller than that described in brain, as a result of alternative splicing of mRNA, but the same final protein is transcribed. Polysome expression profiles indicate that the mRNA of testicular PDYN is translated as efficiently as the larger brain transcript (48). This precursor is expressed mainly in Sertoli cells. Because PDYN products have been immunolocalized to Sertoli cells (63) and to the interstitial compartment of the rat testis, it was suggested that dynorphins secreted by Sertoli cells may regulate the function of these cells in an autocrine manner (62).

In summary, EOPs are present in different cells of the male gonads and are likely to intervene in the mechanisms that regulate spermatogenesis. Opioid precursors are expressed differentially in somatic and germ cells of the testes, indicating that EOPs may regulate testicular function locally by de novo synthesis. In particular, LH and CRF stimulate the production of EOPs in Leydig cells, and these opioid peptides inhibit the role of Sertoli cells in a paracrine manner (Figure 2).

Regulation of Sperm Fertility By the OPIOID System

The presence of three types of opioid receptors (MOR, DOR and KOR) on human sperm membranes (64,65) (Figure 3) indicates that EOPs may contribute to the regulation of human reproductive function by means of a direct effect on sperm. In addition to these opioid receptors, leuenkephalin, metenkephalin and β-endorphin immunoreactivity has been detected in spermatozoa (66; Subirán, unpublished data, 2011), and immunoreactivity to a variety of PENK products was found in the acrosome region (67). Expression of the opioid peptide precursors PENK, PDYN and POMC was also detected by immunohistochemistry in different subcellular regions of human sperm (Subirán, unpublished data, 2011). EOPs such as enkephalins and endorphins are present in seminal fluid, and their concentration is between 6 and 12 times higher than that detected in blood plasma (68). EOP levels are controlled by enzymatic degradation. The two enzymes that specifically degrade enkephalins are aminopeptidase N (APN) and endopeptidase neutral N 24.11 (NEP) (Figure 3). These enzymes are present in both sperm and seminal fractions (69), and their activity in semen is particularly high with respect to other body tissues (70). Interestingly, APN activity levels were found to be altered in semen from subfertile patients, suggesting that this enzyme may play an important role in male fertility (71).

Figure 3
figure 3

Immunofluorescence analysis of opioid receptors in human sperm cells. Distribution of DOR (A), MOR (B) and KOR (C) in spermatozoa is shown. DNA staining was done with Hoechst 33342 (D). Scale bar = 10μm (modified from ref. 64).

Although the three types of opioid receptors are present in human spermatozoa, these receptors show differential expression and localization patterns. MOR and KOR transcripts have been detected in mature sperm by reverse transcription-polymerase chain reaction (RT-PCR), whereas DOR mRNA was absent (64). Selective RNA degradation was reported in human semen, in which specific RNA populations appear to be protected. This result points to the existence of a stable population of RNAs that have previously been synthesized during spermatogenesis (72), since sperm are transcriptionally and translationally inactive. Although the function of RNA in spermatozoa is currently unknown, the following functions for this RNA have been suggested: (a) to facilitate chromatin packing; (b) a role in gene imprinting; (c) to be translated into proteins by mitochondrial polysomes; (d) involvement in the activation of the zygote during early development; and (e) to establish spatial cues in the developing embryo (73,74). However, the three types of opioid receptors (DOR, MOR and KOR) are present in the membranes of sperm cells (64), suggesting that opioid receptors may be expressed during spermatogenesis, although opioid receptors are found exclusively in Sertoli cells in rats (45). Future experiments should focus on characterizing the expression and activation of opioid receptors during spermatogenesis and the function of opioid receptor transcripts in mature sperm.

Sperm motility is an essential feature for reproductive success, since both the journey that the sperm has to make to reach the oocyte and the penetration of the extracellular matrix required for fertilization depends on sperm movement. As a result, sperm motility is considered to be one of the key functions that control reproduction (75). The opioid system could be involved in the control of sperm movement, since asthenozoospermia (reduced motility) is a common pathology found in the sperm of opiate drug addicts (76), and asthenozoospermic patients show reduced levels of metenkephalin in seminal plasma (77).

EOPs can regulate sperm motility, but paradoxical results have been reported. On the one hand, high concentrations of enkephalins and β-endorphins were found to decrease sperm motility (66). Furthermore, synthetic analogs of metenkephalin [D-Ala2, MePhe4, Met(o)5-ol]enkephalin (DAMME)] decreased sperm motility, without affecting sperm viability (78). On the other hand, low concentrations of enkephalins appear to be necessary to maintain sperm motility (77), whereas other studies did not find any effect of metenkephalin on motility (79). Other studies suggested that an adequate level of enkephalins may be necessary to maintain sperm motility and that the effect of these peptides on sperm motility is concentration dependent. Hence, enkephalin-degrading enzymes that participate in regulating the levels of EOP in seminal fluid may affect sperm motility, since the inhibition of both enzymes was found to attenuate the natural, time-dependent decrease in sperm motility. This effect, moreover, was antagonized by naloxone, indicating that the regulation of sperm motility by these enzymes is specifically mediated by an opioid receptor pathway (69).

Experiments carried out with agonists and antagonists of opioid receptors suggest that EOPs have a biphasic effect on sperm motility. On the one hand, sperm motility decreased after morphine (an MOR agonist) treatment, and on the other hand, naltrindole (a DOR antagonist) inhibited sperm motility also (64,80). The inhibitory effect of naltrindole could be due to the displacement of endogenous enkephalin from the delta receptor, which may be necessary to maintain motility. However, the inhibitory action of higher doses of enkephalins may be the result of their effect on MOR (64). This result is also consistent with the effects of the different doses of the opioid antagonist naloxone on sperm motility, where progressive motility was significantly reduced after incubation with high doses of naloxone, whereas it increased significantly after low doses of naloxone (65). Thus, the opioid system can regulate sperm motility in a variety of ways by the activation of distinct receptors (Table 2 and Figure 2). However, recent studies carried out with rats have shown that oxymorphone does not affect sperm count, motility or reproductive organ weights in males (81). The latter finding may be due to a compensatory effect of DOR and MOR, since oxymorphone displays almost equal binding affinity at both receptors (82).

Table 2 Effect of the opioid system on sperm motility, capacitation and acrosome reaction.

Ejaculated sperm cells are immature and infertile and must undergo many modifications to become fertilization competent. These sperm modifications include different processes such as activation of sperm motility, capacitation and hyperactivation. Capacitation is a process that involves sperm membrane reorganization (83), whereas hyperactivation involves changes to the motility pattern. Hyperactive motility is characterized by an increase in the amplitude and asymmetry of flagellar beat, which leads to sudden changes in the direction of travel (84). Only capacitated and hyper-activated sperm can join to and penetrate the zona pellucida of the oocyte; thus, both processes are essential for spermatozoa fertility (85). The union of sperm cells to the oocyte induces multiple signals that induce the sperm acrosome reaction, which involves sperm fusion to the oolemma (plasma membrane of the oocyte) and oocyte penetration (83). Mammalian spermatozoa become capacitated and hyperactivated in the female oviduct (86). It is believed that both processes are due to signals originating in the oviduct and that substances released by the egg would chemotactically attract the sperm toward the egg (83,86). The concentration of metenkephalins and β-endorphins was found to vary along the different parts of the cow reproductive tract (87,88), raising the possibility that the mechanisms regulating these processes in humans may involve EOPs. Nevertheless, the role of the opioid system in capacitation, hyperactivation and acrosome reaction is poorly understood. In vitro studies of horse sperm have found that naloxone can induce the capacitation of sperm cells without affecting the acrosome reaction (65). Moreover, the DOR antagonist naltrindol was found to induce capacitation and acrosome reaction, whereas the corresponding agonist DPDPE ([d-Pen2,d-Pen5]-enkephalin) inhibited the acrosome reaction without affecting capacitation (80). Furthermore, other studies have shown that DAMME, a synthetic analog of metenkephalin, inhibits the spontaneous acrosome reaction in a dose-dependent manner (78) (Table 2). To date, no in vivo studies have been performed in this regard. Therefore, future experiments will need to characterize the participation of the opioid system in these processes.

Closing Remarks

Here, we have briefly reviewed current knowledge about the role of the opioid system in male fertility. The activity of the opioid signaling system is mediated by endogenous opioid peptides. The extensive distribution of opioid peptides and receptors in different organs and in tissues of the male reproductive system indicates that the opioid system operates as a multi-messenger system in the CNS and testes and exerts direct effects on spermatozoa.

In summary, at the level of the CNS, EOPs regulate reproductive function by inhibiting the secretion of GnRH, thereby suppressing the release of LH and sex hormonal steroids such as testosterone and estradiol. Because of the shortcomings of currently available methods of male contraception, opioid system may contribute to develop additional nonhormonal male contraceptive since currently available methods require the administration of exogenous testosterone (89). In the testis, EOPs are mainly synthesized de novo by Leydig cells and in Sertili cells and appear to be able to inhibit Sertoli cell function in an autocrine and paracrine manner. However, opioid precursors are expressed differentially in somatic and germ cells, and the role of EOPs in germline sperm cells is still unknown. Finally, the detection in sperm cells of EOPs, specific enzymes for their degradation and opioid receptors suggests that the opioid system may contribute to sperm fertility, and EOPs may be used as a biochemical tool for the diagnosis and treatment of the human male fertility (90). These findings open up a novel area of therapeutic exploitation of the treatment of male infertility.

Interestingly, different EOPs are also present in the cow reproductive tract (87,88). Recent studies have demonstrated that MOR is expressed in the bovine (91), human (92), canine (93) and equine (94) cumulus-oocyte complex (COC) (95). MOR, by inducing an increase in intracellular calcium, has been shown to participate in the cumulus-oocyte coupled signaling associated with oocyte maturation (94,95). In the mare, a seasonal breeder, MOR was found to be expressed with higher intensity in anoestrous specimens (96). However, at the moment, the role of the opioid system in female fertility is still largely unknown. A thorough understanding of how the opioid system participates in the regulation of reproduction will also require a better understanding of the implication of this system in female reproductive processes.

Disclosure

The authors declare that they have no competing interests as defined by Molecular Medicine, or other interests that might be perceived to influence the results and discussion reported in this paper.

References

  1. Cesselin F. (1995) Opioid and anti-opioid peptides [review]. Fundam. Clin. Pharmacol. 9:409–33.

    Article  CAS  PubMed  Google Scholar 

  2. Roques BP, Noble F, Fournie-Zaluski M-C. (1999) Endogenous Opioid Peptides and Analgesia. In: Opioids in Pain Control: Basic and Clinical Aspects. Stein C (ed.) Cambridge University Press, Cambridge, UK, pp. 21–45. Available at: https://doi.org/ebooks.cambridge.org/ebook.jsf?bid=CBO9780511575068

    Google Scholar 

  3. Turner AJ. (1986) Processing and metabolism of neuropeptides [review]. Essays Biochem. 2:69–119.

    Google Scholar 

  4. Chen Y, Mestek A, Liu J, Yu L. (1993) Molecular cloning of a rat kappa opioid receptor reveals sequence similarities to the mu and delta opioid receptors. Biochem. J. 295:625–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Merkouris M, et al. (1996) Identification of the critical domains of the delta-opioid receptor involved in G protein coupling using site-specific synthetic peptides. Mol. Pharmacol. 50:985–93.

    CAS  PubMed  Google Scholar 

  6. Jordan B, Devi LA. (1998) Molecular mechanisms of opioid receptor signal transduction [review]. Br. J. Anaesth. 81:12–9.

    Article  CAS  PubMed  Google Scholar 

  7. Gaveriáux-Ruff C, Kieffer BL. (1999) Opioid receptors: gene structure and function. In: Opioids in Pain Control: Basic and Clinical Aspects. Stein C (ed.) Cambridge University Press, Cambridge, UK, pp. 1–20. Available at: https://doi.org/ebooks.cambridge.org/ebook.jsf?bid=CBO9780511575068

    Google Scholar 

  8. Wick MJ, et al. (1994) Isolation of a novel cDNA encoding a putative membrane receptor with high homology to the cloned mu, delta, and kappa opioid receptors. Brain Res. Mol. Brain Res. 27:37–44.

    Article  CAS  PubMed  Google Scholar 

  9. Mollereau C, et al. (1994). ORL1, a novel member of the opioid receptor family: cloning, functional expression and localization. FEBS Lett. 341:33–8.

    Article  CAS  PubMed  Google Scholar 

  10. Stein C, ed. (1999) Opioids in Pain Control: Basic and Clinical Aspects. Cambridge (UK): Cambridge University Press. 359 pp. Available at: https://doi.org/ebooks.cambridge.org/ebook.jsf?bid=CBO9780511575068

    Google Scholar 

  11. Mollereau C, et al. (1996) Structure, tissue distribution, and chromosomal localization of the pre-pronociceptin gene. Proc. Natl. Acad. Sci. U. S. A. 93:8666–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Pan YX, Xu J, Pasternak GW. (1996) Cloning and expression of a cDNA encoding a mouse brain orphanin FQ/nociceptin precursor. Biochem. J. 315:11–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Sharma SK, Klee WA, Nirenberg M. (1977) Opiate-dependent modulation of adenylate cyclase. Proc. Natl. Acad. Sci. U. S. A. 74:3365–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Surprenant A, Shen KZ, North RA, Tatsumi H. (1990) Inhibition of calcium currents by noradrenaline, somatostatin and opioids in guinea-pig submucosal neurons. J. Physiol. 431:585–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. McGivern RF, Henschel DM. (1990) Interaction of naltrexone with postnatal administration of testosterone and estrogen on neurobehavioral sexual differentiation in rats. Horm. Behav. 24:20–39.

    Article  CAS  PubMed  Google Scholar 

  16. North RA, Williams JT, Surprenant A, Christie MJ. (1987) Mu and delta receptors belong to a family of receptors that are coupled to potassium channels. Proc. Natl. Acad. Sci. U. S. A. 84:5487–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chan JS, Chiu TT, Wong YH. (1995) Activation of type II adenylyl cyclase by the cloned mu-opioid receptor: coupling to multiple G proteins. J. Neurochem. 65:2682–9.

    Article  CAS  PubMed  Google Scholar 

  18. Tsu RC, Chan JS, Wong YH. (1995) Regulation of multiple effectors by the cloned delta-opioid receptor: stimulation of phospholipase C and type II adenylyl cyclase. J. Neurochem. 64:2700–7.

    Article  CAS  PubMed  Google Scholar 

  19. Law PY, Wong YH, Loh HH. (2000) Molecular mechanisms and regulation of opioid receptor signaling [review]. Annu. Rev. Pharmacol. Toxicol. 40:389–430.

    Article  CAS  PubMed  Google Scholar 

  20. Bodnar RJ. (2010) Endogenous opiates and behavior: 2009. Peptides. 12:2325–59.

    Article  Google Scholar 

  21. McLachlan RI, Wreford NG, Robertson DM, de Kretser DM. (1995) Hormonal control of spermatogenesis. Trends Endocrinol. Metab. 6:95–101.

    Article  CAS  PubMed  Google Scholar 

  22. Eblen A, Bao S, Lei ZM, Nakajima ST, Rao CV. (2001) The presence of functional luteinizing hormone/chorionic gonadotropin receptors in human sperm. J. Clin. Endocrinol. Metab. 86:2643–8.

    Article  CAS  PubMed  Google Scholar 

  23. Lei ZM, Rao CV. (2001) Neural actions of luteinizing hormone and human chorionic gonadatropin [review]. Semin. Reprod. Med. 19:103–9.

    Article  CAS  PubMed  Google Scholar 

  24. Heckert LL, Griswold MD. (2002) The expression of the follicle-stimulating hormone receptor in spermatogenesis [review]. Recent Prog. Horm. Res. 57:129–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Cicero TJ, Schainker BA, Meyer ER. (1979) Endogenous opioids participate in the regulation of the hypothalamus-pituitary-luteinizing hormone axis and testosterone’s negative feedback control of luteinizing hormone. Endocrinology. 104:1286–91.

    Article  CAS  PubMed  Google Scholar 

  26. Sirinathsinghji DJ, Whittington PE, Audsley A, Fraser HM. (1983) Beta-endorphin regulates lordosis in female rats by modulating LH-RH release. Nature. 301:2–4.

    Article  CAS  PubMed  Google Scholar 

  27. Wilkes MM, Yen SS. (1981) Augmentation by naloxone of efflux of LRF from superfused medial basal hypothalamus. Life Sci. 28:2355–9.

    Article  CAS  PubMed  Google Scholar 

  28. Blank MS, Roberts DL. (1982) Antagonist of gonadotropin-releasing hormone blocks naloxone-induced elevations in serum luteinizing hormone. Neuroendocrinology 35:309–12.

    Article  CAS  PubMed  Google Scholar 

  29. Blank MS, Fabbri A, Catt KJ, Dufau ML. (1985) Direct inhibition of gonadotroph function by opiates. Trans. Assoc. Am. Physicians. 98:1–9.

    CAS  PubMed  Google Scholar 

  30. Bliesener N, et al. (2005) Plasma testosterone and sexual function in men receiving buprenorphine maintenance for opioid dependence. J. Clin. Endocrinol. Metab. 90:203–6.

    Article  CAS  PubMed  Google Scholar 

  31. Daniell HW. (2002) Hypogonadism in men consuming sustained-action oral opioids. J. Pain. 3:377–84.

    Article  PubMed  Google Scholar 

  32. Vuong C, Van Uum SH, O’Dell LE, Lutfy K, Friedman TC. (2010) The effects of opioids and opioid analogs on animal and human endocrine systems [review]. Endocr. Rev. 31:98–132.

    Article  CAS  PubMed  Google Scholar 

  33. Fabbri A, et al. (1989) Endorphins in male impotence: evidence for naltrexone stimulation of erectile activity in patient therapy. Psychoneuroen-docrinology. 14:103–11.

    Article  CAS  PubMed  Google Scholar 

  34. Fraioli F, et al. (1982) Control of gonadotropin secretion in man: role of opioid peptides. Horm. Metab. Res. 14:312–6.

    Article  CAS  PubMed  Google Scholar 

  35. Soverchia L, et al. (2006) Proopiomelanocortin gene expression and beta-endorphin localization in the pituitary, testis, and epididymis of stallion. Mol. Reprod. Dev. 73:1–8.

    Article  CAS  PubMed  Google Scholar 

  36. Fabbri A, Knox G, Buczko E, Dufau ML. (1988) Beta-endorphin production by the fetal Leydig cell: regulation and implications for paracrine control of Sertoli cell function. Endocrinology. 122:749–55.

    Article  CAS  PubMed  Google Scholar 

  37. Tsong SD, et al. (1982) ACTH and beta-endor-phin-related peptides are present in multiple sites in the reproductive tract of the male rat. Endocrinology. 110:2204–6.

    Article  CAS  PubMed  Google Scholar 

  38. Pintar JE, Schachter BS, Herman AB, Durgerian S, Krieger T. (1984) Characterization and localization of proopiomelanocortin messenger RNA in the adult rat testis. Science. 225:632–4.

    Article  CAS  PubMed  Google Scholar 

  39. Kilpatrick DL, Millette CF. (1986) Expression of proenkephalin messenger RNA by mouse spermatogenic cells. Proc. Natl. Acad. Sci. U. S. A. 83:5015–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Dierich A, et al. (1998) Impairing follicle-stimulating hormone (FSH) signaling in vivo: targeted disruption of the FSH receptor leads to aberrant gametogenesis and hormonal imbalance. Proc. Natl. Acad. Sci. U. S. A. 95:13612–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tapanainen JS, Aittomäki K, Min J, Vaskivuo T, Huhtaniemi IT. (1997) Men homozygous for an inactivating mutation of the follicle-stimulating hormone (FSH) receptor gene present variable suppression of spermatogenesis and fertility. Nat. Genet. 15:205–6.

    Article  CAS  PubMed  Google Scholar 

  42. Russell L, Clermont Y. (1976) Anchoring device between Sertoli cells and late spermatids in rat seminiferous tubules. Anat. Rec. 185:259–78.

    Article  CAS  PubMed  Google Scholar 

  43. Morse HC, Horike N, Rowley MJ, Heller CG. (1973) Testosterone concentrations in testes of normal men: effects of testosterone propionate administration. J. Clin. Endocrinol. Metab. 37:882–6.

    Article  CAS  PubMed  Google Scholar 

  44. Huang HF, Pogach LM, Nathan E, Giglio W, Seebode JJ. (1991) Synergistic effects of follicle-stimulating hormone and testosterone on the maintenance of spermiogenesis in hypophysectomized rats: relationship with the androgen-binding protein status. Endocrinology. 128:3152–61.

    Article  CAS  PubMed  Google Scholar 

  45. Fabbri A, Tsai-Morris CH, Luna S, Fraioli F, Dufau ML. (1985) Opiate receptors are present in the rat testis: identification and localization in Sertoli cells. Endocrinology. 117:2544–6.

    Article  CAS  PubMed  Google Scholar 

  46. Kilpatrick DL, Borland K, Jin DF (1987). Differential expression of opioid peptide genes by testicular germ cells and somatic cells. Proc. Natl. Acad. Sci. U. S. A. 84:5695–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. O’Hara BF, et al. (1994) Proenkephalin transgenic mice: a short promoter confers high testis expression and reduced fertility. Mol. Reprod. Dev. 38: 275–84.

    Article  PubMed  Google Scholar 

  48. Garrett JE, Collard MW, Douglass JO. (1989) Translational control of germ cell-expressed mRNA imposed by alternative splicing: opioid peptide gene expression in rat testis. Mol. Cell. Biol. 9:4381–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kilpatrick DL, et al. (1990) Transcription of the rat and mouse proenkephalin genes is initiated at distinct sites in spermatogenic and somatic cells. Mol. Cell. Biol. 10:3717–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kew D, Kilpatrick DL. (1989) Expression and regulation of the proenkephalin gene in rat Sertoli cells. Mol. Endocrinol. 3:179–84.

    Article  CAS  PubMed  Google Scholar 

  51. Mehta ND, et al. (1994) Proenkephalin gene expression in testicular interstitial cells is down-regulated coincident with the appearance of pachytene spermatocytes. Endocrinology. 135:1543–50.

    Article  CAS  PubMed  Google Scholar 

  52. Chen CL, Madigan MB. (1987) Regulation of testicular proopiomelanocortin gene expression. Endocrinology. 121:590–6.

    Article  CAS  PubMed  Google Scholar 

  53. Eskeland NL, Lugo DI, Pintar JE, Schachter BS. (1989) Stimulation of beta-endorphin secretion by corticotropin-releasing factor in primary rat Leydig cell cultures. Endocrinology. 124:2914–9.

    Article  CAS  PubMed  Google Scholar 

  54. Audhya T, Hollander CS, Schlesinger DH, Hutchinson B. (1989) Structural characterization and localization of corticotropin-releasing factor in testis. Biochim. Biophys. Acta. 995: 10–6.

    Article  CAS  PubMed  Google Scholar 

  55. Ulisse S, Fabbri A, Dufau ML. (1989) Corticotropin-releasing factor receptors and actions in rat Leydig cells. J. Biol. Chem. 264:2156–63.

    CAS  PubMed  Google Scholar 

  56. Fabbri A, Tinajero JC, Dufau ML. (1990) Corticotropin-releasing factor is produced by rat Leydig cells and has a major local antireproductive role in the testis. Endocrinology. 127:1541–3.

    Article  CAS  PubMed  Google Scholar 

  57. Garrett JE, Douglass JO. (1989) Human chorionic gonadotropin regulates expression of the proenkephalin gene in adult rat Leydig cells. Mol. Endocrinol. 3:2093–100.

    Article  CAS  PubMed  Google Scholar 

  58. Gerendai I, Shaha C, Thau R, Bardin CW. (1984) Do testicular opiates regulate Leydig cell function? Endocrinology. 115:1645–7.

    Article  CAS  PubMed  Google Scholar 

  59. Gerendai I. (1991) Modulation of testicular functions by testicular opioid peptides [review]. J. Physiol. Pharmacol. 42:427–37.

    CAS  PubMed  Google Scholar 

  60. da Silva VA Jr, et al. (2006) Neonatal treatment with naloxone increases the population of Sertoli cells and sperm production. Reprod. Nutr. Dev. 46:157–66.

    Article  PubMed  Google Scholar 

  61. Fabbri A, et al. (1989) Neuroendocrine control of male reproductive function: the opioid system as a model of control at multiple sites. J. Steroid Biochem. 32:145–50.

    Article  CAS  PubMed  Google Scholar 

  62. Douglass J, Cox B, Quinn B, Civelli O, Herbert E. (1987) Expression of the prodynorphin gene in male and female mammalian reproductive tissues. Endocrinology. 120:707–13.

    Article  CAS  PubMed  Google Scholar 

  63. Collard MW, Day R, Akil H, Uhler MD, Douglass JO. (1990) Sertoli cells are the primary site of prodynorphin gene expression in rat testis: regulation of mRNA and secreted peptide levels by cyclic adenosine 3′,5′-monophosphate analogs in cultured cells. Mol. Endocrinol. 4:1488–96.

    Article  CAS  PubMed  Google Scholar 

  64. Agirregoitia E, et al. (2006) Expression and localization of delta-, kappa-, and mu-opioid receptors in human spermatozoa and implications for sperm motility. J. Clin. Endocrinol. Metab. 91:4969–75.

    Article  CAS  PubMed  Google Scholar 

  65. Albrizio M, Guaricci AC, Calamita G, Zarrilli A, Minoia P. (2006) Expression and immunolocalization of the mu-opioid receptor in human sperm cells. Fertil. Steril. 86:1776–9.

    Article  CAS  PubMed  Google Scholar 

  66. Sastry BV, Janson VE, Owens LK, Tayeb OS. (1982) Enkephalin- and substance P-like immunoreactivities of mammalian sperm and accessory sex glands. Biochem. Pharmacol. 31:3519–22.

    Article  CAS  PubMed  Google Scholar 

  67. Kew D, Muffly KE, Kilpatrick DL (1990). Proenkephalin products are stored in the sperm acrosome and may function in fertilization. Proc. Natl. Acad. Sci. U. S. A. 87:9143–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Sharp B, Pekary AE. (1981) Beta-endorphin 61–91 and other beta-endorphin-immunoreactive peptides in human semen. J. Clin. Endocrinol. Metab. 52: 586–8.

    Article  CAS  PubMed  Google Scholar 

  69. Subirán N, et al. (2008) Expression of enkephalin-degrading enzymes in human semen and implications for sperm motility. Fertil. Steril. 89 Suppl 5:1571–7.

    Article  PubMed  Google Scholar 

  70. Fernández D, Valdivia A, Irazusta J, Ochoa C, Casis L. (2002) Peptidase activities in human semen. Peptides. 23:461–8.

    Article  PubMed  Google Scholar 

  71. Irazusta J, et al. (2004) Enkephalin-degrading enzymes in normal and subfertile human semen. J. Androl. 25:733–9.

    Article  CAS  PubMed  Google Scholar 

  72. Miller D, et al. (1999) A complex population of RNAs exists in human ejaculate spermatozoa: implications for understanding molecular aspects of spermiogenesis. Gene. 237:385–92.

    Article  CAS  PubMed  Google Scholar 

  73. Miller D, Ostermeier GC. (2006) Spermatozoal RNA: Why is it there and what does it do? Gynecol. Obstet. Fertil. 34:840–6.

    Article  CAS  PubMed  Google Scholar 

  74. Miller D, Ostermeier GC, Krawetz SA. (2005) The controversy, potential and roles of spermatozoal RNA [review]. Trends Mol. Med. 11:156–63.

    Article  CAS  PubMed  Google Scholar 

  75. Quill TA, Wang D, Garbers DL. (2006) Insights into sperm cell motility signaling through sNHE and the CatSpers [review]. Mol. Cell. Endocrinol. 250:84–92.

    Article  CAS  PubMed  Google Scholar 

  76. Ragni G, De Lauretis L, Bestetti O, Sghedoni D, Gambaro V. (1988) Gonadal function in male heroin and methadone addicts. Int. J. Androl. 11:93–100.

    Article  CAS  PubMed  Google Scholar 

  77. Fujisawa M, Kanzaki M, Okada H, Arakawa S, Kamidono S. (1996) Metenkephalin in seminal plasma of infertile men. Int. J. Urol. 3:297–300.

    Article  CAS  PubMed  Google Scholar 

  78. Foresta C, Tramarin A, Scandellari C, Arslan P. (1985) Effects of a met-enkephalin analogue on motility, O2 consumption, and ATP content of human spermatozoa. Arch. Androl. 14:247–52.

    Article  CAS  PubMed  Google Scholar 

  79. Fraioli F, et al. (1984) Beta-endorphin, Metenkephalin, and calcitonin in human semen: evidence for a possible role in human sperm motility. Ann. N. Y. Acad. Sci. 438:365–70.

    Article  CAS  PubMed  Google Scholar 

  80. Albrizio M, et al. (2010) Delta opioid receptor on equine sperm cells: subcellular localization and involvement in sperm motility analyzed by computer assisted sperm analyzer (CASA). Reprod. Biol. Endocrinol. 8:78.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Shuey DL, Stump DG, Carliss RD, Gerson RJ. (2008) Effects of the opioid analgesic oxymorphone hydrochloride on reproductive function in male and female rats. Birth Defects Res. B. Dev. Reprod. Toxicol. 83:12–8.

    Article  CAS  PubMed  Google Scholar 

  82. Ananthan S, et al. (2004) Identification of opioid ligands possessing mixed micro agonist/delta antagonist activity among pyridomorphinans derived from naloxone, oxymorphone, and hydromorphone. J. Med. Chem. 47:1400–12.

    Article  CAS  PubMed  Google Scholar 

  83. Flesch FM, Gadella BM. (2000) Dynamics of the mammalian sperm plasma membrane in the process of fertilization. Biochim. Biophys. Acta. 1469:197–235.

    Article  CAS  PubMed  Google Scholar 

  84. Suarez SS. (2008) Control of hyperactivation in sperm [review]. Hum. Reprod. Update. 14:647–57.

    Article  CAS  PubMed  Google Scholar 

  85. Mortimer ST. (2000) CASA: practical aspects [review]. J. Androl. 1:515–24.

    Google Scholar 

  86. Suarez SS. (2008) Regulation of sperm storage and movement in the mammalian oviduct [review]. Int. J. Dev. Biol. 52:455–62.

    Article  PubMed  Google Scholar 

  87. Petraglia F, et al. (1986) Endogenous opioid peptides in uterine fluid. Fertil. Steril. 46:247–51.

    Article  CAS  PubMed  Google Scholar 

  88. Facchinetti F, et al. (1986) Met-enkephalin enhances follicle-stimulating hormone-dependent progesterone production from cultured granulosa cells. J. Clin. Endocrinol. Metab. 63:1222–4.

    Article  CAS  PubMed  Google Scholar 

  89. Amory JK. (2005) Male hormonal contraceptives: current status and future prospects [review]. Treat. Endocrinol. 4:333–41.

    Article  CAS  PubMed  Google Scholar 

  90. Garrido N, et al. (2008) Contribution of sperm molecular features to embryo quality and assisted reproduction success [review]. Reprod. Biomed. Online. 17:855–65.

    Article  CAS  PubMed  Google Scholar 

  91. Dell’Aquila ME, et al. (2002) Effects of beta-endorphin and Naloxone on in vitro maturation of bovine oocytes. Mol. Reprod. Dev. 63:210–22.

    Article  PubMed  Google Scholar 

  92. Dell’Aquila ME, et al. (2003) Immunofluorescence detection of the mu-opioid receptor in human oocytes. Hum. Reprod. Suppl. 18:180.

    Google Scholar 

  93. Pavone L, Albrizio M, Minoia R. (2008) Expression and localization of the mu-opioid receptor in canine oocytes. Reprod. Fertil. Dev. 20:172.

    Article  Google Scholar 

  94. Dell’Aquila ME, et al. (2008) Expression and localization of the mu-opioid receptor (MOR) in the equine cumulus-oocyte complex and its involvement in the seasonal regulation of oocyte meiotic competence. Mol. Reprod. Dev. 75:1229–46.

    Article  PubMed  Google Scholar 

  95. Iorga AI, et al. (2009) Expression of the mu opioid receptor and effects of the opioid antagonist naloxone on in vitro maturation of oocytes recovered from anoestrous bitches. Reprod. Domest. Anim. 44 Suppl 2:263–8.

    Article  PubMed  Google Scholar 

  96. Desantis S, et al. (2010) Changes in the expression of the mu-opioid receptor in the mare oviduct during oestrus and anoestrus. Anim. Reprod. Sci. 119: 40–9.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This review was supported by a grant from the Basque Government (GIC10/95). The authors thank BioEntelechia Translations for improving the English in this paper.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nerea Subirán.

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Subirán, N., Casis, L. & Irazusta, J. Regulation of Male Fertility by the Opioid System. Mol Med 17, 846–853 (2011). https://doi.org/10.2119/molmed.2010.00268

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2119/molmed.2010.00268

Keywords