Sei sulla pagina 1di 4

Journal of Neuroendocrinology 20 (Suppl.

1), 90–93
ª 2008 The Authors. Journal Compilation ª 2008 Blackwell Publishing Ltd
REVIEW ARTICLE

The Cannabinoid System and Male Reproductive Functions


M. Rossato, C. Pagano and R. Vettor
Endocrine-Metabolic Laboratory, Clinica Medica 3, Department of Medical and Surgical Sciences, University of Padova, Padova, Italy.

Journal of Cannabinoids, the main active components of marijuana, have been shown to exert different
adverse effects on male reproduction both in vertebrates and invertebrates. In vivo, cannabinoids
Neuroendocrinology exert negative effects on hypothalamic–hypophyseal reproductive hormone secretion and testi-
cular endocrine and exocrine functions. Furthermore, a large amount of experimental data
obtained in vitro have clearly shown that cannabinoids negatively influence important sperm
functions, including motility and acrosome reaction, two fundamental processes necessary for
oocyte fertilisation. These inhibitory effects are mediated by the direct action of cannabinoids on
Correspondence to: sperm through the activation of the cannabinoid receptor subtype CNR1 that has been shown
Dr Marco Rossato, Endocrine- to be expressed in mature sperm. In the present paper, we briefly review the effects of cannabi-
Metabolic Laboratory, Clinica Medica noids and endocannabinoids, a particular group of endogenously produced cannabinoids, on
3, Department of Medical and male reproductive function.
Surgical Sciences, University of
Padova, Via Ospedale 105, 35128 Key words: cannabinoids, sperm, male reproduction.
Padova, Italy (e-mail: marco.rossato@
unipd.it). doi: 10.1111/j.1365-2826.2008.01680.x

Marijuana, the most widely used recreational drug worldwide (1), function (Table 1) with important final effects on fertility in the
contains a large number of different active compounds known as male (10–12).
cannabinoids, and D9-tetrahydocannabinol (THC) has been recog-
nised as the main active molecule (2). Beside natural cannabi-
Cannabinoids and spermatogenesis
noids, different tissues can synthesise a group of related
unsaturated fatty acid derivatives named endocannabinoids that
Cannabinoids and hypothalamus-pituitary-testis axis
act as endogenous ligands for the cannabinoid receptors. This so
called ‘endocannabinoid system’ regulates many different func- Spermatogenesis is a long a complex process controlled by a num-
tions in all tissues via interaction with two specific receptors, ber of different mechanisms (13). Gonadotrophins (FSH [follicle
cannabinoid receptor subtype 1 (CNR1) and cannabinoid receptor stimulating hormone] and LH [luteinising hormone]) play a crucial
subtype 2 (CNR2), whose tissue distribution and function have role in the induction and maintenance of spermatogenesis in mam-
been previously described (3–5). A number of different endocann- mals including man (14), involving a stimulatory effect of Sertoli
abinoids have been isolated within biological fluids, anandamide and Leydig cells and a complex series of secreting events that fol-
(N-arachidonoylethanolamine, AEA) and 2-arachidonoyl glycerol low. It is known that in males, cannabis smoking decreases plasma
(2-AG) being the main components (3). In particular, AEA is the LH levels, leading to decreased plasma testosterone (15), although
most abundant in reproductive secretions, thus suggesting a pos- contradictory results have been reported (16, and references
sible role for the endocannabinoid system in reproduction (6). therein). FSH secretion has also been shown to be modulated by
CNR receptors have been identified along male reproductive tract: cannabinoids (17) and the combined effects on the gonadotrophins
CNR1 receptors have been detected in the testis, prostate and lead to decreased sperm count in males that has been consistently
vas deferens (7–10). Furthermore, functional CNR1 receptors have observed in men who smoke cannabis (18).
been found in sperm in different vertebrate and invertebrate spe- The effects of cannabinoids on gonadotrophin secretion seem to
cies (10–12). To date, unequivocal data have been reported show- be localised upstream to the pituitary, and, in particular, at the pre-
ing that cannabinoids control male reproductive functions both optic area of the hypothalamus where CNR1 receptors have been
directly and indirectly, depending of the expression of the CNR localised and where LH releasing hormone (LHRH)-secreting
receptor subtypes along the tissues controlling the testicular neurones reside (19). At this level, cannabinoids seem to modulate
Cannabinoids and male reproduction 91

Table 1. Expression of the Cannabinoid Receptor Subtypes in the Tissues grating brain and testicular messages are modulated by cannabi-
Participating in the Control of the Mammalian Male Reproductive Tract noids at both the hypothalamic–pituitary and the testicular level.
Functions.

Cannabinoid receptor Cannabinoids and sperm functions


Tissue subtype Reference
Cannabinoids and sperm motility
Hypothalamus
GnRH secreting cells CNR1 ⁄ CNR2 (20) Since the first report published by Perez et al. (26) in 1981, experi-
Pituitary mental data have accumulated confirming that cannabinoids nega-
Gonadotrophs CNR1 (21) tively influence the motility of sperm in different mammalian
Testis species. Apart from observations obtained in sea urchins showing
Germ cells CNR1 (10) no significant effects of cannabinoids on sperm motility (12), these
Sertoli cells CNR2 (24) substances inhibit sperm motility in different mammalian species,
Leydig cells CNR1 (8) including man (10, 27). The observations on the negative effects of
Epididymis n.d. –
cannabinoids on sperm motility were reported in the mid 1970s
Vas deferens CNR1 (38)
and early 1980s; however, CNR1 and CNR2 were not identified until
Seminal vesicles n.d. –
Prostate CNR1 (37)
the early 1990s, when expression of CNR1 was demonstrated in the
Corpus cavernosum CNR1 (39) testis (28). Only recently, taking a pharmacological experimental
approach, the inhibitory effects of cannabinoids on mammalian
CNR1, cannabinoid receptor subtype 1; CNR2, cannabinoid receptor subtype sperm motility have been attributed to CNR1 activation (10). Very
2; n.d., not determined. recently, Cobellis et al. (11) replicated these results in sperm from
Rana esculenta, taking the same pharmacological approach.
negatively the activity of LHRH-secreting neurones by direct and Furthermore, the same authors extended these observations by
indirect mechanisms (20). Beyond the well known effect of canna- means of a molecular genetic experimental design, showing that
binoids on gonadotrophin secretion at the hypoyhalamic level mod- sperm from CNR1-knockout mice show a dramatic increase in
ulating gonadal function, previous observations have localised motility percentages in the caput epididymis. This clearly demon-
CNR1 in the anterior pituitary, suggesting a direct effect of canna- strates that the lack of CNR1 signalling increases the acquisition of
binoids on LH and FSH secretion at the pituitary level (21). sperm motility (9).
Although the studies evaluating the effects of cannabinoids on
sperm motility are not abundant, it seems that in general the acti-
Cannabinoids and male germ cells
vation of CNR1 inhibits motility in invertebrate and vertebrate
Evidence primarily from animal studies has shown that CNR1 is sperm, at least in vitro. Contradictory results have been reported,
expressed in germ cells, from spermatogonia to mature sperm. For however, regarding the effects of cannabinoids ingestion, as well as
instance Gye et al. (8) demonstrated the presence of this receptor marijuana smoking, on human sperm motility in vivo. Variation in
in spermatogonia, primary spermatocytes and sperm in mouse tes- these results may be owing to the experimental design or the small
tis, and these observations have been replicated by Cobellis et al. number of subjects participating to the different clinical studies
(11) in the testis of invertebrates. Current knowledge clearly shows (29).
that cannabinoids negatively influence the spermatogenetic process,
reducing germ cell proliferation and reproductive organ weight (11,
Cannabinoids and sperm capacitation
22). Sertoli cells constitute one of the main components of the
seminiferous tubule, and their number – fixed at puberty – is clo- After ejaculation, sperm have to undergo a complex – and still
sely related to the total sperm production. Sertoli cells surround unclear – series of biochemical and morphological events collec-
and nurse germinal cells during their maturation. By mediating the tively known as ‘capacitation’, before they are capable of fertilising
actions of all hormonal stimuli regulating the spermatogenetic pro- an egg (30). This includes remodelling the lipid plasma membrane,
cess they provide a finely tuned microenvironment leading to germ which is required to improve the ability of sperm to respond to
cell maturation from spermatogonium to mature sperm (23). stimuli that induce the acrosome reaction, facilitating the interac-
CNR2 has been recently described in Sertoli cells, modulating tion with the egg. These changes are activated by still not fully elu-
apoptosis in these cells (24). Moreover, Leydig cells have been cidated mechanisms involving the adenylate cyclase ⁄ cAMP ⁄ protein
shown to express CNR1, which, when activated, induces a reduction kinase A (PKA) signalling pathway, leading to tyrosine phosphoryla-
of testosterone production, thus further inhibiting the spermatoge- tion of different proteins (31). These processes are inhibited by
netic process (because high local levels of testosterone are required cannabinoids and CNR1 activation (27, 31). These effects are
to maintain spermatogenesis (8)). Furthermore, testosterone is specific: they are counteracted by sperm pre-incubation with the
necessary for epididymis and seminal vesicle activity, which CNR1 antagonist SR141716.
influence important sperm functions such as motility (25). Thus, the
complex events coordinating the male spermatogenesis and inte-

ª 2008 The Authors. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20 (Suppl. 1), 90–93
92 M. Rossato et al.

Physiologically, sperm capacitation occurs within the female gen- tebrates, impairing the fertilisation process and thus reproductive
ital tract and it has been demonstrate that sperm leaving seminal function. This is important, given the prevalence of marijuana
plasma and reaching the uterus and oviduct swim in an external smoking particularly in the young. These observations, together
milieu presenting progressively reduced concentrations of endo- with the demonstration that the male reproductive tract possesses
cannabinoids (6). As sperm approach the egg, they encounter the the whole enzymatic machinery for the synthesis and metabolism
lowest concentrations of endocannabinoids present in the female of endocannabinoids, could suggest the existence of particular clini-
genital tract, thus suggesting that the activation of the endo- cal situations, beyond marijuana smoking, characterised by a patho-
cannabinoid system in sperm seems to maintain the sperm in an logical increase of endocannabinoids within the male reproductive
uncapacitated, quiescent condition before interacting with the egg tract, that could lead to an impairment of sperm function.
(10). Furthermore, if we consider that the use of cannabinoids has
been legally authorised in some countries for the treatment of
some pathological conditions (such as appetite stimulation in
Cannabinoids and sperm acrosome reaction
patients with AIDS, treatment of nausea and vomiting, reduction of
Sperm capacitation is a fundamental pre-requisite for exocytosis of symptoms in some neurological diseases, reducing intraocular pres-
the acrosome, a specialised secretory granule located at the apex of sure in glaucoma patients, treatment of depression and pain), the
sperm head (32). Sperm acrosome reaction occurs as the sperm possible negative effects of these therapeutic cannabinoid agonists
faces the egg, and leads to the release of proteolytic enzymes nec- on human male fertility have to be taken into careful account.
essary for penetration of the egg’s coats, the exposure of specia-
lised sperm membrane sites that allow attachment and fusion with
Conflicts of interest
the egg’s plasma membrane and then to fertilisation (33). Sperm
that do not undergo the acrosome reaction are not able to bind to MR, CP and RV have received speaker’s fees from Sanofi-Aventis.
the egg’s zona pellucida and cannot fertilise it. Substantially homo-
geneous data exist indicating that CNR1 activation reduce the abil- Received: 7 January 2008,
ity of sperm to undergo acrosome reaction both in vertebrates and accepted 8 February 2008
invertebrates (10, 12, 27, 31). These data further confirm the inhibi-
tory effects of cannabinoids on sperm fertilising ability. References
Importantly, since cannabinoids do not affect fertility of eggs,
these effects are directed only to sperm (27). The mechanisms 1 Di Marzo V. A brief history of cannabinoid and endocannabinoid phar-
macology as inspired by the work of British scientists. Trends Pharmacol
involved in cannabinoid inhibition of the sperm acrosome reaction
Sci 2006; 27: 134–140.
are still unknown. It is well known that ion channels and regulation 2 Di Marzo V, De Petrocellis L, Bisogno T. The biosynthesis, fate and phar-
of ion fluxes through the sperm plasma membrane are crucial for macological properties of endocannabinoids. Handb Exp Pharmacol
the acrosome reaction, and the most important physiological regu- 2005; 168: 147–185.
lator appears to be Ca2+ (34). In this regard we have previously 3 Felder CC, Glass M. Cannabinoid receptors and their endogenous agon-
shown that the endocannabinoid AEA did not modify intracellular ists. Annu Rev Pharmacol Toxicol 1998; 38: 179–200.
Ca2+ concentrations in human sperm, possibly ruling out any inter- 4 Pertwee RG. Pharmacology of cannabinoid CB1 and CB2 receptors. Phar-
macol Ther 1997; 74: 129–180.
fering effect of cannabinoids on Ca2+ signalling (10). CNR1 is a
5 McPartland JM, Glass M, Pertwee RG. Meta-analysis of cannabinoid
G-protein-coupled receptor that has been shown to inhibit adenyl- ligand binding affinity and receptor distribution: interspecies differences.
ate cyclase activity (3). Intracellular levels of cAMP rise during acro- Br J Pharmacol 2007; 152: 583–593.
some reaction, cell-permeant cAMP analogues induce the acrosome 6 Schuel H, Burkman LJ, Lippes J, Crickard K, Forester E, Piomelli D, Giuffri-
reaction, and pharmacological inhibitors of cAMP-dependent PKA da A. N-Acylethanolamines in human reproductive fluids. Chem Phys
reduce this exocytotic event (35, 36): these observations suggest Lipids 2002; 121: 211–227.
that the adenylate cyclase ⁄ cAMP ⁄ PKA system plays a primary role 7 Pertwee RG, Ross RA, Craib SJ, Thomas A. Cannabidiol antagonizes can-
nabinoid receptor agonists and noradrenaline in the mouse vas deferens.
in the signalling pathway leading to sperm acrosome reaction and
Eur J Pharmacol 2002; 456: 99–106.
egg fertilisation. The inhibitory effect of cannabinoids on this fun- 8 Gye MC, Kang HH, Kang HJ. Expression of cannabinoid receptor 1 in
damental signalling pathway, as recently described in mammalian mouse testes. Arch Androl 2005; 51: 247–255.
sperm (31), might explain the negative effect these agents have, 9 Ricci G, Cacciola G, Altucci L, Meccariello R, Pierantoni R, Fasano S,
through CNR1 activation, on sperm capacitating and acrosome Cobellis G. Endocannabinoid control of sperm motility: the role of epi-
reaction. didymus. Gen Comp Endocrinol 2007; 153: 320–322.
10 Rossato M, Ion Popa F, Ferigo M, Clari G, Foresta C. Human sperm
express cannabinoid receptor Cb1, the activation of which inhibits motil-
Conclusions ity, acrosome reaction, and mitochondrial function. J Clin Endocrinol
Metab 2005; 90: 984–991.
Marijuana is the most commonly used recreational drug worldwide 11 Cobellis G, Cacciola G, Scarpa D, Meccariello R, Chianese R, Franzoni MF,
and it is now well recognised that the active components of Mackie K, Pierantoni R, Fasano S. Endocannabinoid system in frog and
marijuana – as well as endogenously synthesised endocannabinoids rodent testis: type-1 cannabinoid receptor and fatty acid amide hydro-
– negatively affect sperm functions, both in invertebrates and ver- lase activity in male germ cells. Biol Reprod 2006; 75: 82–89.

ª 2008 The Authors. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20 (Suppl. 1), 90–93
Cannabinoids and male reproduction 93

12 Schuel H, Goldstein E, Mechoulam R, Zimmerman AM, Zimmerman S. 26 Perez LE, Smith CG, Asch RH. delta 9-tetrahydrocannabinol inhibits fruc-
Anandamide (arachidonylethanolamide), a brain cannabinoid receptor tose utilization and motility in human,rhesus monkey, and rabbit sperm
agonist, reduces sperm fertilizing capacity in sea urchins by inhibiting in vitro. Fertil Steril 1981; 35: 703–705.
the acrosome reaction. Proc Natl Acad Sci U S A 1994; 91: 7678–7682. 27 Schuel H, Burkman LJ, Lippes J, Crickard K, Mahony MC, Giuffrida A,
13 Foresta C, Bettella A, Merico M, Garolla A, Plebani M, Ferlin A, Rossato Picone RP, Makriyannis A. Evidence that anandamide-signaling regulates
M. FSH in the treatment of oligozoospermia. Mol Cell Endocrinol 2000; human sperm functions required for fertilization. Mol Reprod Dev 2002;
161: 89–97. 63: 376–387.
14 Foresta C, Bettella A, Rossato M, La Sala G, De Paoli M, Plebani M. Inhi- 28 Gérard CM, Mollereau C, Vassart G, Parmentier M. Molecular cloning of
bin B plasma concentrations in oligozoospermic subjects before and a human cannabinoid receptor which is also expressed in testis. Biochem
after therapy with follicle stimulating hormone. Hum Reprod 1999; 14: J 1991; 279: 129–134.
906–912. 29 Whan LB, West MC, McClure N, Lewis SE. Effects of delta-9 tetrahydro-
15 Kolodny RC, Masters WH, Kolodner RM, Toro G. Depression of plasma cannabinol, the primary psychoactive cannabinoid in marijuana, on
testosterone levels after chronic intensive marihuana use. N Engl J Med human sperm function in vitro. Fertil Steril 2006; 85: 653–660.
1974; 290: 872–874. 30 Salicioni AM, Platt MD, Wertheimer EV, Arcelay E, Allaire A, Sosnik J, Vis-
16 Park B, McPartland JM, Glass M. Cannabis, cannabinoids and reproduc- conti PE. Signalling pathways involved in sperm capacitation. Soc Reprod
tion. Prostaglandins Leukot Essent Fatty Acids 2004; 70: 189–197. Fertil Suppl 2007; 65: 245–259.
17 Wenger T, Rettori V, Snyder GD, Dalterio S, McCann SM. Effects of D-9- 31 Maccarrone M, Barboni B, Paradisi A, Bernabò N, Gasperi V, Pistilli MG,
tetrahydrocannabinol on the hypothalamic-pituitary control of luteiniz- Fezza F, Lucidi P, Mattioli M. Characterization of the endocannabinoid
ing hormone and follicle-stimulating hormone secretion in adult male system in boar spermatozoa and implications for sperm capacitation and
rats. Neuroendocrinology 1987; 46: 488–493. acrosome reaction. J Cell Sci 2005; 118: 4393–4404.
18 Hembree WC III, Nahas GG, Zeidenberg P, Huang HF. Changes in human 32 Yanagimachi R. Mammalian fertilization. In: Knobil E, Neill JD, eds. The
spermatozoa associated with high dose marihuana smoking. Adv Biosci Physiology of Reproduction. New York: Raven Press, 1994: 189–281.
1978; 23: 429–439. 33 Wassarman PM, Jovine L, Qi H, Williams Z, Darie C, Litscher ES. Recent
19 Matsuda LA, Bonner TI, Lolait SJ. Localization of cannabinoid receptor aspects of mammalian fertilization research. Mol Cell Endocrinol 2005;
mRNA in rat brain. J Comp Neurol 1993; 327: 535–550. 234: 95–103.
20 Gammon CM, Freeman GM Jr, Xie W, Petersen SL, Wetsel WC. Regula- 34 Darszon A, Labarca P, Nishigaki T, Espinosa F. Ion channels in sperm
tion of gonadotropin-releasing hormone secretion by cannabinoids. physiology. Physiol Rev 1999; 79: 481–510.
Endocrinology 2005; 146: 4491–4499. 35 Beltrán C, Vacquier VD, Moy G, Chen Y, Buck J, Levin LR, Darszon A. Par-
21 Wenger T, Fernández-Ruiz JJ, Ramos JA. Immunocytochemical demon- ticulate and soluble adenylyl cyclases participate in the sperm acrosome
stration of CB1 cannabinoid receptors in the anterior lobe of the pitui- reaction. Biochem Biophys Res Commun 2007; 358: 1128–1135.
tary gland. J Neuroendocrinol 1999; 11: 873–878. 36 De Jonge CJ, Han HL, Lawrie H, Mack SR, Zaneveld LJ. Modulation of the
22 Maccarrone M, Falciglia K, Di Rienzo M, Finazzi-Agrò A. Endocannabi- human sperm acrosome reaction by effectors of the adenylate cyclase ⁄
noids, hormone-cytokine networks and human fertility. Prostaglandins cyclic AMP second-messenger pathway. J Exp Zool 1991; 258: 113–125.
Leukot Essent Fatty Acids 2002; 66: 309–317. 37 Tokanovic S, Malone DT, Ventura S. Stimulation of epithelial CB1 recep-
23 Brinster RL. Male germline stem cells: from mice to men. Science 2007; tors inhibits contractions of the rat prostate gland. Br J Pharmacol
316: 404–405. 2007; 150: 227–234.
24 Maccarrone M, Cecconi S, Rossi G, Battista N, Pauselli R, Finazzi-Agrò A. 38 Christopoulos A, Coles P, Lay L, Lew MJ, Angus JA. Pharmacological
Anandamide activity and degradation are regulated by early postnatal analysis of cannabinoid receptor activity in the rat vas deferens. Br J
aging and follicle-stimulating hormone in mouse Sertoli cells. Endocri- Pharmacol 2001; 132: 1281–1291.
nology 2003; 144: 20–28. 39 Ghasemi M, Sadeghipour H, Dehpour AR. Anandamide improves the
25 Higgins SJ, Burchell JM. Effects of testosterone on messenger ribonucleic impaired nitric oxide-mediated neurogenic relaxation of the corpus cav-
acid and protein synthesis in rat seminal vesicle. Biochem J 1978; 174: ernosum in diabetic rats: involvement of cannabinoid CB1 and vanilloid
543–551. VR1 receptors. BJU Int 2007; 100: 1385–1390.

ª 2008 The Authors. Journal Compilation ª 2008 Blackwell Publishing Ltd, Journal of Neuroendocrinology, 20 (Suppl. 1), 90–93

Potrebbero piacerti anche