You are on page 1of 14

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/280060634

Impact of Metformin on Male Reproduction

Article in Current Pharmaceutical Design · July 2015


DOI: 10.2174/1381612821666150710150041 · Source: PubMed

CITATIONS READS
31 4,707

6 authors, including:

Carolina Ferreira Ana Rabaca


University of Minho University of Porto
14 PUBLICATIONS 178 CITATIONS 6 PUBLICATIONS 59 CITATIONS

SEE PROFILE SEE PROFILE

Pedro F Oliveira Marco G. Alves


University of Aveiro University of Aveiro
470 PUBLICATIONS 8,155 CITATIONS 433 PUBLICATIONS 7,917 CITATIONS

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Marco G. Alves on 16 September 2015.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.ae
Current Pharmaceutical Design, 2015, 21, 3621-3633 3621

Impact of Metformin on Male Reproduction

Carolina Ferreira1,2, Mário Sousa1,3, Ana Rabaça1,4, Pedro F. Oliveira1,5, Marco G. Alves5 and Rosália Sá1*

1
Department of Microscopy, Laboratory of Cell Biology, Institute of Biomedical Sciences Abel Salazar (ICBAS)
and Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, Rua Jorge Viterbo Ferreira
228, 4050-313 Porto, Portugal; 2University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-
801 Vila Real, Portugal; 3Centre for Reproductive Genetics Prof. Alberto Barros, Av. do Bessa 240, 1° Dto.
Frente, 4100-009 Porto, Portugal; 4Faculty of Medicine of University of Porto (FMUP), Al. Prof. Hernâni Mon-
teiro, 4200-319 Porto, Portugal; 5CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, Uni-
versity of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal

Abstract: Male infertility has been increasing over the last decades being nowadays a pressing health problem.
Diabetes mellitus (DM) can contribute directly or indirectly to male infertility due to an abnormal spermatogenesis,
which results in a decreased sperm quality. Type 2 Diabetes mellitus (T2DM) is responsible for the vast majority of
DM cases, being frequently treated with oral antidiabetic drugs. Metformin is the most cost-effective therapy for the treatment of T2DM.
This biguanide is an oral insulin-sensitizing agent capable of increasing insulin sensitivity and decreasing plasma fasting insulin levels.
The main metabolic action of this drug occurs in the liver. However, it has been shown that metformin acts on a variety of organs includ-
ing the male reproductive system. With the rising numbers of diabetic individuals among younger populations, there is an increase in the
consumption of metformin in individuals of this age group. As a result, it is important to discuss the role of metformin in male fertility.
This review presents the most recent data available from studies on the effects of metformin on male reproductive system. Together with
the discussion of these effects, their significance to male fertility is also debated.

Keywords: Diabetes mellitus, metabolism, metformin, male fertility, Sertoli cell, spermatogenesis.

INTRODUCTION effects of this disease and to the eventual effects of its treatment
Human infertility is a serious health problem and a social con- [19].
cern that affects approximately 15% of couples of reproductive age The first aim for DM management is the proper control of gly-
[1, 2]. On average, 50% of all infertility cases can be attributed to a cemia, in order to minimize the patient’s risk of developing vascu-
male factor [3]. Male reproductive health has been decreasing over lar and neurologic complications. Lifestyle interventions (including
the last few decades with a high prevalence of infertile men below diet, physical activity and weight control) are cost-effective means
their forties [4-7]. Male infertility is a multifactorial disorder clini- to improve DM outcome. Still, for the majority of patients, a life-
cally characterized by a low or absent sperm count and/or the pres- style modification alone is not sufficient to maintain normal blood
ence of nonfunctional sperm cells due to abnormal spermatogenesis glucose levels. Thereby, pharmaceutical interventions are required
[8]. Spermatogenesis is a complex process both dependent and to maintain these levels and reduce the risk of clinical complica-
controlled by different somatic cell types present in the testes. tions. Oral antidiabetic agents that are currently in clinical use can
Therefore, Sertoli cells (SCs), Leydig cells (LCs) and developing be categorized into insulin-sensitizing agents (e.g., metformin and
germ cells are strictly associated in order to maintain a normal thiazolidinediones), enhancers of insulin secretion (e.g., sulfony-
spermatogenesis, with production of mature spermatozoa capable of lureas and meglitinides), agents that modify intestinal absorption of
fertilizing an oocyte [9]. carbohydrate or fat (e.g., a-glucosidase inhibitors, lipase inhibitors),
Systemic diseases, such as Diabetes mellitus (DM), may irre- and exogenous insulin [20, 21]. Metformin (1, 1-dimethylbigua-
versibly affect spermatogenesis leading to a decrease in sperm qual- nide) belongs to the insulin sensitizers class and is a synthetic drug
ity, which can directly or indirectly contribute to male infertility implemented as the first-line oral treatment for individuals with
[10, 11]. DM is a metabolic disorder characterized by a defective T2DM [17, 22]. This agent has been clinically used for over 50
insulin production, insulin resistance, or both [12, 13]. Patients with years, and is now consumed daily by more than 100 million indi-
this disease exhibit reduced antioxidant activity [14] and dysfunc- viduals worldwide [23]. Notably, in the last years several other
tion of the hypothalamic-pituitary-gonadal axis [15, 16]. Type 1 and effects have been attributed to metformin. For instance, metformin
type 2 Diabetes mellitus (T1DM and T2DM, respectively), are the administration has been reported to inhibit the proliferation of neo-
most commonly found types of DM in the general population. plastic cells [24, 25] and, when given in combination with
Nonetheless, T2DM is responsible for the vast majority of DM clomifene, to improve the fertility of women with polycystic ovary
cases. In the clinical practice, T2DM is regularly treated with a syndrome (PCOS) through the induction of ovulation [26]. These
combination of oral antidiabetic drugs, healthy food regimens and reports showed that metformin, not only has anti-hyperglycaemic
physical activity [17]. T2DM has been proliferating in the younger capacity, but can also act in various body tissues, including the
population [18]. As a result, there is an increase in the consumption reproductive system [27, 28].
of oral antidiabetics by this age group. Consequently, there is a high DM impact on male fertility is widely known. Nevertheless,
percentage of males in reproductive age exposed to the adverse DM is a chronic disease and more research is needed to identify the
effects of its continued therapy on male reproductive functions.
*Address correspondence to this author at Department of Microscopy,
Hence, it is necessary to further investigate the effects of metformin
Laboratory of Cell Biology, ICBAS-UP, UMIB, Rua Jorge Viterbo Ferreira on this system, evaluating its potentially harmful or beneficial ef-
228, Building 1, Floor 2, Room 03, 4050-313 Porto, Portugal; Tel: +351-22 fects and thus establishing its safety. Herein, we aim to discuss the
042 8000x5242; Fax: +351-22 042 80 90; E-mail: rmsa@icbas.up.pt

1873-4286/15 $58.00+.00 © 2015 Bentham Science Publishers


3622 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Ferreira et al.

recent literature available on the effects of this drug on male fertil- somatic cells are also responsible for water transport from the inter-
ity. stitial space into the lumen, facilitating the movement of sperm
from the testis to the epididymis [41]. SCs secrete multiple nutri-
SPERMATOGENESIS AND MAINTENANCE OF FUNC- ents, peptides and metabolic intermediates necessary for the effi-
TIONAL SPERM PRODUCTION cient development of germ cells during spermatogenesis [40].
The male reproductive system is of great complexity. This sys- Among these substances, lactate plays a key role, being the pre-
tem comprises the testes, the accessory glands and the penis. The ferred fuel for germ cells development and survival [42]. Research
perfect coordination between physical and hormonal components is conducted on this subject by Courtens and peers, concluded that the
necessary to the normal occurrence of spermatogenesis. In addition, infusion of lactate in rat testes significantly improves the spermato-
this process is dependent on a strict regulation by the brain [29]. genesis index [43]. In addition, other studies reported that lactate
Testes are the central reproductive organs, being responsible for exerts anti-apoptotic effects [44], stimulates RNA and protein syn-
two essential functions: synthesis of sex steroid hormones (steroi- thesis [45, 46], and increases the respiratory rate of male germ cells
dogenesis) and production of spermatozoa (spermatogenesis) [30, [46].
31]. Steroidogenesis is mainly maintained through LCs’ steroid- Spermatogenesis is under strong hormonal regulation, particu-
secreting activity, being highly important for the development of larly by the pituitary hormones, follicle-stimulating hormone (FSH)
spermatogenesis [32]. This process occurs within the seminiferous and luteinizing hormone (LH). These hormones promote the func-
tubules (ST) of the testes of pubertal men. ST basically consist of tional connection between the brain and testis through the hypo-
an association of SCs with germ cells. SCs create tight junctions thalamus-pituitary-testis axis [47]. FSH and LH are dependent on
between the neighboring cells giving rise to the Sertoli/blood-testis the release of the gonadotropin-releasing hormone (GnRH) by the
barrier (BTB) [33]. This barrier physically divides the seminiferous hypothalamus. When FSH and LH are produced, they are responsi-
epithelium into basal and apical compartments, each containing ble for the stimulation of SCs and LCs, respectively. However,
germ cells in different stages of development. The basal compart- when the stimulation index is at its highest, a negative-feedback
ment consists of type A and B spermatogonia and leptotene sperma- mechanism takes place. Hormones produced by LCs and SCs, tes-
tocytes I, while the apical region comprises pachytene spermato- tosterone and inhibin, respectively, act on the hypothalamus and
cytes I, spermatocytes II and spermatids [34]. The BTB also con- pituitary inhibiting the production of GnRH, FSH and LH [48].
trols the entry of some substances from the blood into the This complex regulatory mechanism is able to maintain the appro-
seminiferous epithelium luminal environment, controlling their priate levels of hormones for the normal course of spermatogenesis.
levels, and immunologically isolating the germ cells [35-37]. Each Consequently, any disruption on these mechanisms may affect male
SC is responsible for the structural maintenance and nurturing of fertility.
multiple germ cells at various stages of maturation [38-40]. These

Interstitial Space

Basement membrane Leydig


cell

Myoid cell

Spermatogonium B
Spermatogonium A
Leptotene
Spermatocyte I
BTB
Pachytene
Endoplasmic Spermatocyte I
reticulum
Sertoli cell
nucleus
Spermatocyte II

Round
Spermatids

Spermatozoa

Mitochondria

Sertoli cell

Fig. (1). Schematic illustration of spermatogenesis. Spermatogonia type A divide and develop into spermatogonia type B, which enter mei-
otic prophase and differentiate into primary spermatocytes that undergo meiosis I and form the haploid secondary spermatocytes. Meiosis II
yields four equalized spermatids that migrate toward the lumen where fully formed spermatozoa are finally released. BTB - Sertoli/blood-testis
barrier.
Metformin and Male Fertility Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3623

In mammals, spermatogenesis (Fig. 1) is a complex event of GLUTs accountable for the glucose uptake in these cells. GLUT8 is
cellular differentiation that features three main continuous steps: 1) located in the membrane of intracellular organelles and is not ex-
mitotic spermatogonia proliferation and differentiation; 2) meiotic pected to contribute for the glucose intake from the extracellular
phase; and 3) metamorphosis of round spermatids [49]. In the first milieu [63, 64, 66, 67]. In these cells, glucose intake is highly regu-
phase, the male stem cell, spermatogonium (2n), undergoes mitosis. lated by a group of different substances (hormones, cytokines and
At the end of mitosis, this cell originates two different cell types: growth factors) that have the ability to modulate the mRNA and
spermatogonium A (2n) and B (2n). While spermatogonium A re- protein expression of GLUTs, thereby ensuring an appropriate lac-
mains undifferentiated, ensuring the germ cell line maintenance, tate production [61, 62, 64]. In fact, it was shown that in SCs, when
spermatogonium B originates primary spermatocyte which under- the levels of glucose or insulin are decreased, GLUTs expression
goes meiosis, continuing the spermatogenesis process [49, 50]. At may be altered [67, 68]. In human and rat SCs, insulin deprivation
the end of meiosis I, the primary spermatocyte (2n) originates two [67] or glucose deprivation [68] increases GLUT1 and decreases
secondary spermatocytes (n). These go through the second meiotic GLUT3 mRNA expression. Similarly in rats SCs, factors such as
division and form four round spermatids (n). Spermatids undergo FSH, interleukin-1 (IL1) and the basic fibroblast growth factor
maturation, becoming more elongated, compact and motile in a (bFGF), modulate the mRNA expression of GLUT1 during pubertal
mechanism known as spermiogenesis [49, 51]. Finally, spermiation development [64]. Once inside the SCs, the majority of glucose
occurs, the release of immature spermatozoa into the lumen of ST. follows the glycolytic pathway being metabolised into pyruvate
Subsequently, the last vestiges of cytoplasm are removed as the which is subsequently converted to lactate with the oxidation of
spermatozoon transits to the rete testis and epididymis where the NADH to NAD+ [69]. Once produced, lactate is exported to the
final maturation process occurs [49, 51]. extracellular environment, where it can be used by the developing
germ cells. This transport through the SCs plasma membrane to
RELEVANCE OF THE NUTRITIONAL SUPPORT OF DE- germ cells is mediated by specific set of carriers called monocar-
VELOPING GERM CELLS AND SPERMATOZOA TO boxylate transporters (MCTs), isoforms MCT1, MCT2 and MCT4
SPERMATOGENESIS [58, 61, 67, 68, 70, 71]. While MCT4 exports lactate of SCs, MCT1
During spermatogenesis, germ cells present very specific needs and MCT2 are responsible for lactate uptake by germ cells [70, 71].
for their development into mature spermatozoa. Both germ cells Notably, SCs are also capable of producing this substrate in the
and spermatozoa rely on specific nutrients for their development absence of glucose. Indeed, SCs are able to produce lactate through
and to obtain the necessary energy for several cellular functions. the metabolism of lipids [72], amino acids or glycogen [68, 73] in
Sugars (mainly hexoses like glucose and fructose) and other me- order to ensure the metabolic requirements of spermatogenesis.
tabolites (such as lactate and citrate) are the preferred substrate of After spermatogenesis is completed, the newly formed sper-
germ cells and spermatozoa for ATP production [52, 53]. In fact, matozoa are stored in the epididymis for a final maturation process.
glucose is the main fuel for the normal maintenance of spermato- Hither and once required, it is important for spermatozoa to obtain
genesis and, subsequently, a normal production of mature, motile energy necessary to acquire and maintain motility [74]. The major-
spermatozoa [30]. Furthermore, glucose metabolism is responsible ity of functional characteristics, like motility and fertilization capac-
for the SCs production of lactate, which is then exported into the ity, rely on a high-energy production [52]. Several studies have
intra-tubular fluid, where it is the main metabolite used by germ been conducted on the existing GLUTs on spermatozoa. In fact,
cell during spermatogenesis [42, 54]. several authors have reported that GLUT1-3 and GLUT5 are pre-
Glucose is a polar molecule, thus requiring the presence of sent in spermatozoa, both in humans as in numerous other mam-
transport proteins to effectively cross the lipid bilayer. Glucose mals like rats, bulls, dogs, boars, stallions and donkeys [57, 75-78].
transporters are divided into two groups, sodium-dependent glucose Additionally, mRNA and protein expression of GLUT8 was ob-
transporters (SGLT) and glucose transporters (GLUTs) [55, 56]. served in human mature spermatozoa [79], while both isoforms of
The SGLT family is composed of six different active glucose trans- GLUT9 (GLUT9a and GLUT9b) were reported in mouse sper-
porter proteins, SGLT 1 to 6, that spend energy to transfer glucose matozoa [80, 81]. Contrastingly, the presence of GLUT4 was not
across the plasma membrane [55]. On the contrary, GLUTs are reported in any of the above mentioned species [57, 75-78]. Con-
passive transporters that despite their name, may carry other mole- cerning cellular distribution, GLUTs can be found in different re-
cules in addition to glucose, such as fructose, mannitol, vitamins gions along the spermatozoa. GLUT1 is present in the initial and
[57] and glucosamine [52]. The GLUTs family encompasses 14 final portion of the tail and in the apical region of the spermatozoa
proteins that are grouped into 3 classes, I (GLUT1-4 and 14); II head. GLUT2 is mainly concentrated in the acrosomal region,
(GLUT5, 7, 9 and 11); and III (GLUT6, 8, 10 and H+-coupled myo- whereas GLUT3 and GLUT5 are located in the head and midpiece
inositol transporter (HMIT)), varying on their tissue distribution, of spermatozoa. Besides, GLUT8 can be found in the acrosomal
hexose affinity and structure [52]. However, only some of these membrane, whereas GLUT9a and GLUT9b are localised on the tail
passive transporters (GLUT 1-3,5,8 and 9) are present in mammal- of spermatozoa [79-81].
ian testicular cells and are responsible for the uptake of glucose Even when subjected to anaerobic conditions, mature spermato-
[52]. zoa have the ability to use distinct substrates to obtain energy, such
Being located in the basal compartment of ST, undifferentiated as glucose and fructose [82-84]. Indeed, two distinct mechanisms
spermatogonia are supplied with nutrients directly from the intersti- can be used by spermatozoa to obtain energy: oxidative phosphory-
tial fluid and use glucose as fuel for ATP production [58]. Mature lation (midpiece) and glycolysis (principal piece) [85]. These
germ cells, particularly pachytene spermatocytes and round sper- mechanisms can act together or independently, depending on the
matids, express all enzymes of the glycolytic pathway. However energetic needs of the spermatozoa [84]. In spermatozoa, glucose
they are unable to use glucose and require lactate from the extracel- undergoes the glycolytic pathway and is converted to lactate by a
lular environment for energy production and survival [42, 46, 54, specific isoform of lactate dehydrogenase (LDH), present only in
58]. SCs play a crucial role in lactate production for the mature the testes (LDHC) [86]. Lactate is then accumulated in these cells,
germ cells and are able to produce lactate via glycolysis at high where it can further be used for energy production and for sper-
rates [46, 58-62]. Glucose is imported into the SCs by a number of matozoa motility enhancement [83, 84, 87]. Consequently, glucose
different GLUTs [59]. GLUT1, 2, 3 and 8 are reported as the metabolism in SCs and in mature spermatozoa are closely related
GLUTs responsible for the glucose transport on these cells, how- with male fertility, thus being essential processes for the regular
ever, their contribution is not equal [63-65]. Since GLUT1-3 are development of germ cells during spermatogenesis and for the final
located in the plasma membrane, they are suggested as the main maturation of spermatozoa.
3624 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Ferreira et al.

BASIC ASPECTS OF DIABETES Several studies have reported that DM has a detrimental effect
Systemic diseases, especially DM, are highly prevalent disor- on male reproductive system, commonly resulting in infertility or
ders that tremendously affect today’s world health. The number of subfertility by the induction of a number of alterations associated
individuals with diabetes has been increasing over the last decades, with testicular dysfunction [107]. Male DM patients present numer-
and it is expected that by the year 2030 more than 552 million peo- ous alterations on their reproductive system, being that changes in
ple will be affected by this disease [13]. There are numerous factors sperm parameters are the most noticeable. A decrease in testicular
that contribute to its high prevalence, particularly those closely weight, together with a reduction of testicular and epididymal
related to obesity and a generally unhealthy lifestyle [12, 13]. DM sperm content and a regression of the epididymis, have been consis-
is a metabolic disorder, characterized by an intensified oxidative tently observed in animals with DM [108-110]. The metabolic al-
stress and a hyperglycaemic state, which induce structural and func- terations induced by DM can instigate various abnormalities on
tional alterations in cells, tissues and organs [88]. Individuals af- male fertility, which can ultimately lead to the production of ab-
fected by this disorder present a defective insulin production, insu- normal spermatozoa. Sperm motility, count and volume were the
lin resistance, or both factors combined [12, 13]. Moreover, DM is mainly altered properties, being significantly reduced on individu-
accompanied with a reduced antioxidant activity [14] and dysfunc- als with T1DM and T2DM [11, 111-114]. Clinical studies reported
tion of the hypothalamic-pituitary-gonadal axis [15, 16]. Notably, alterations of sperm morphology and an increase of nuclear and
DM is associated with a wide range of other health issues, like heart mitochondrial DNA fragmentation in human spermatozoa of dia-
diseases, stroke, nerve damage, and kidney or eye problems [89]. betic individuals [11, 93]. Furthermore, there are multiple animal
Although there are several types of DM, T1DM and T2DM are the studies reporting a decrease on sperm parameters quality on dia-
most widespread. betic subjects, which are in clear agreement with most results ob-
tained in humans [108, 110, 115-117].
T1DM is characterized by a complete absence of insulin due to
an autoimmune destruction of pancreatic -cells, which are the With the growth of T2DM among children and adolescents
insulin-producing cells. Hence, patients with this disorder need [92], there is a definite need to fully understand the exact mecha-
exogenous insulin to control their blood glucose levels. As a result, nisms involved in the subfertility or infertility of these male dia-
T1DM patients are commonly referred as insulin-dependent [90- betic patients, and the correlation between the two pathological
92]. Although T1DM can affect a wide range of age groups, chil- states. With the rising number of diabetic individuals there is also a
dren and young people are undoubtedly the most affected [93]. growth in the consumption of antidiabetics [118]. There are several
oral antidiabetics available, categorized into different groups ac-
T2DM is responsible for the great majority of all DM cases. It cording to their means of action. Individuals suffering from T2DM
is characterised by insulin resistance and impaired insulin secretion can be treated with different classes of oral antidiabetics, such as
[89, 92], associated with a continued decline in -cell function and insulin sensitizers, insulin secretagogues, and external insulin deliv-
an increase of -cell apoptosis [94]. The incomplete suppression of ery [20]. Among these, metformin is one of the most frequently
hepatic glucose production associated with a decreased efficiency prescribed drugs for T2DM and is usually recommended in combi-
of the liver and muscle glucose uptake promotes an increase in nation with a healthy lifestyle [17]. This drug is the first line treat-
blood glucose levels of patients with this disorder [95]. Cells are ment for this type of DM, being able to control the glycemic levels,
unable to respond to the abnormal blood insulin levels, and pancre- reduce the risk of micro- and macrovascular complications, and
atic cells display a limited capacity to produce higher amounts of significantly decrease mortality rates [119]. In spite of all the dis-
insulin in response to insulin resistance. All these factors combined coveries made on the mechanisms of action of this agent, little is
lead to a failure in the control of the blood glucose levels. Patients known about its effects on the male reproductive system and spe-
with T2DM do not require exogenous insulin to control their blood cifically on germ cell development. Consequently, it is crucial to
glucose levels, thus being referred to as non-insulin dependent [89]. further investigate the implications of metformin consumption on
There are various individuals with pre-diabetes, an intermediate male fertility.
state between a normal control of glucose levels and T2DM [96]
that have a higher predisposition to develop T2DM. Interestingly, METFORMIN AND ITS THERAPEUTIC RELEVANCE
pre-diabetic patients exhibit important metabolic alterations, like
Metformin (1, 1-dimethylbiguanide) is a synthetic drug origi-
impaired fasting glucose, impaired glucose tolerance and/or dis-
nated from a medicinal herb used since the medieval Europe,
turbed insulin secretion [97]. Although there is a high incidence of
Galega officinalis [120]. This agent is the most cost-effective ther-
pre-diabetic patients, age seems to be the crucial factor for T2DM
apy for the treatment of individuals with T2DM [17, 22] and is now
development, with most patients being from the adult and elder age
used by more than 100 million individuals [23]. In the clinical prac-
groups [89]. Nonetheless, the number of young patients with T2DM
tice, metformin has been used for over 50 years, being characterised
has been increasing over the last years, mainly because of the high
as an oral biguanide insulin-sensitizing agent capable of increasing
prevalence of obesity that is now found in the younger population
insulin sensitivity and significantly decreasing plasma fasting insu-
[12, 13, 95, 98]. So, it is expected that the number of individuals
lin levels [121]. This drug exerts its principal metabolic action, the
suffering from T2DM and its associated complications will con-
glucoregulatory, in the liver. However, it may act on a variety of
tinue to rise, affecting more individuals each year.
body tissues like skeletal muscles, adipose tissue, endothelium and
DIABETES: A THREAT FOR MALE FERTILITY reproductive tissues [27]. Therefore, owing to its multiple action on
several body regions, which are affected by insulin resistance and
Diabetes is associated with a wide range of health complica- hyperinsulinemia, metformin has gained interest for its multiple
tions. Male fertility is no exception. Males with DM are at high risk potential therapeutic uses [27, 122]. When administered, metformin
for fertility issues, due to alterations induced on their reproductive is not metabolised and is excreted in the urine and bile [123]. Thus,
potential [15, 99]. In addition, depending on the patient’s age, his this agent does not affect the action of other drugs prescribed to
control of blood glucose levels and the duration of the disease, DM patients.
is reported to cause erectile dysfunction [100, 101]. Other compli-
cations associated with DM in males are impotence, retrograde The best-known effects mediated by metformin are associated
ejaculation and decreased sexual desire [101-104], as well as ab- with its anti-hyperglycemic action. Its major effects are: 1) blocking
normalities in testicular functions and disruption of the spermato- of liver gluconeogenesis; 2) increasing skeletal muscle uptake of
genesis endocrine control, leading to an abnormal sperm production glucose [124]; 3) reduction of the absorption of glucose in intestinal
[105, 106]. mucosa; and 4) decreasing lipogenesis in the liver, which can re-
duce lipotoxicity and improve insulin signaling reducing glu-
Metformin and Male Fertility Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3625

coneogenesis [125]. In addition, it has also been shown that met- have been conducted on this subject took use of animal models.
formin can be used as an antineoplastic agent. Indeed, metformin Rodents were the most frequently used animal models, although
restricts the growth and proliferation of various neoplastic cells other animals have been exploited, as is the case of chickens and
both in vitro and in vivo. These results were described in different rabbits. Furthermore, in vivo and in vitro approaches were also
tumors, such as pulmonary adenocarcinoma [24], gastric [25], ovar- employed in order to evaluate the effects of metformin on the male
ian [126-129], endometrial [130], prostate and colon cancer [131], reproductive tract. Data were obtained from several cell types, like
bladder neoplastic cells [132] and different types of breast cancer fetal testicular cells, adult germ cells, SCs or spermatozoa. How-
[133-135]. Additionally, metformin increases the effect of the che- ever, the existing results remain controversial with some conflicting
motherapeutic agents in cultures of ovarian cancer cells, when co- results. We have summarized the different metformin-related ef-
administered with cisplatin [126, 128, 129], carboplatin [136, 137], fects on the male reproductive system in Table 1.
phenethyl isothiocyanate [138], and in cultures of breast cancer Tartarin and collaborators conducted an extensive study on the
cells, when co-administered with doxorubicin [135]. Notably, it has effect of metformin in human and murine testes using in vitro ex-
also been reported that this drug diminishes the incidence of breast perimental models [162]. The authors evaluated the impact of this
cancer in diabetic women [136]. Recently, Singhal and collabora- drug by exposing organotypic cultures of fetal testes to increasing
tors established as well metformin as a possible adjuvant for anti- doses of metformin (50 μM to 5 mM) for 3 days. They reported that
tuberculosis therapy, by reduction of intracellular growth of Myco- both murine and human systems were affected, exhibiting de-
bacterium tuberculosis [139]. Metformin can also be used in the creased testosterone production and increased lactate secretion with
treatment of anovulatory infertility in women with PCOS [140], the supra-pharmacological (500 M) concentration, in the human
inducing ovulation and increasing pregnancy rates [26, 141]. Like- model, and a 500 M and 5 mM concentrations in mice. Further-
wise, was proven that metformin modulates the reproductive axis, more, the mRNA expression levels of key players involved in ster-
affecting the release of GnRH and LH [142]. Moreover, reactive oid secretion were also reduced after exposure to 5 mM of met-
oxygen species (ROS) can be reduced by metformin treatment formin. While for humans the lowest observed effect concentration
[143-145] and the expression of antioxidant genes (i.e. Nrf2) can be on testosterone secretion was with the pharmacological concentra-
increased [146]. tion of metformin (50 M), in mouse it was only verified at 500
Although this drug has been clinically available since the 50s, M. With these concentrations, the production of testosterone was
little is known about its exact mechanisms of action. Even so, it is reduced, which suggests that its pharmacological dose may cause
known that metformin requires different isoforms of “Organic impaired steroidogenesis [162]. In mouse cultures, it was also ob-
cation transporters” (OCT1 and OCT2) or of “multidrug and toxic served a significant increase in the mRNA levels of LDH at a con-
compound extrusion type” transporters (MATE1 and MATE2), to centration of 5 mM. A similar stimulation of the LDH mRNA ex-
be transported into the various tissues. OCT1 is mainly responsible pression has also been described in rat SCs after treatment with
for the transport of metformin in the liver and gut, whereas OCT2 is aminoimidazole carboxamide ribonucleotide, another AMPK in-
primarily found in the kidney [147, 148]. Metformin is rapidly ab- ducer [9]. The increase of lactate production promoted by AMPK
sorbed across the intestinal epithelium and transported to the liver, activation on fetal murine testis in vitro, thus seems to be resultant
where it accumulates [123]. In the hepatocytes, the predominant of augmented LDH levels. Due to this increased lactate production,
expression of OCT1 facilitates cellular uptake of metformin into the particularly at high concentrations of metformin, it has been hy-
liver [148] and as a result, there is a higher accumulation of this pothesized that this drug may also have a protective effect on sper-
drug in the liver than in other tissues [149]. Once inside the cytoso- matogenesis, which could lead to a promotion of sperm production,
lic compartment, mitochondria constitute the primary target of this caused by an increase of the glycolysis index. In fact, to better
drug. At this point, metformin inhibits the complex I of the mito- comprehend the conflicting results obtained in vitro, the authors
chondrial respiratory chain, without affecting any other steps of its also conducted an in vivo study, where pregnant mice were exposed
machinery [150, 151]. However, the exact mechanism by which to a daily dose of metformin (300 mg/kg the dose that is needed to
this drug promotes this inhibition remains unknown. After the spe- obtain a similar therapeutic effect in diabetic animals as in humans -
cific inhibition of complex I, an increase in the intracellular 50 M). The treatment took place between embryonic day 0.5 and
AMP/ATP ratio occurs leading to serine/threonine protein kinase 13.5 or 16.5. The results obtained revealed that the pre-natal met-
(AMPK) activation in the liver and in other tissues. This activation formin exposure reduces the testes size and the ST diameter. A
promotes a decrease in lipid synthesis along with an increase of reduction in the SC population, due to a substantial decrease in the
fatty acid oxidation [152, 153]. Furthermore, it also inhibits glu- proliferation indexes of these cells, was also observed. This can
coneogenesis [152-155], mitochondrial ROS production [156] and also be explained by the activation of the AMPK pathway since it
cell proliferation [127, 157]. The decrease in ROS production re- may lead to a decrease in cell proliferation. However, this decline in
sults in an increase of glutathione (GSH), catalase (CAT), and su- the SCs number did not affect the total number of germ cells pre-
peroxide dismutase (SOD) levels [158]. As AMPK is present in sent in the testes [162]. In contrast with the in vitro studies, met-
many tissues, metformin effects are designated as pleiotropic [28]. formin did not affect the intratesticular lactate concentration.
Therefore, in energy-rich environments, such as is the case of obe- Hence, the authors concluded that some alterations in fetal testicu-
sity and insulin resistance, AMPK is frequently inactivated and lar function due to metformin were obtained at concentrations equal
high levels of ROS can be found [156, 159, 160]. However met- to or 10 times higher than the therapeutic doses used, since they
formin can also act independently from AMPK activation. Experi- were able to promote a deleterious effect on several parameters
ments with AMPK and the upstream liver kinase K1 (LKB1) associated with the male reproductive function both in vivo and in
knockout (KO) mice, as well as wild type mice, have shown that vitro.
serum glucose levels were reduced after metformin treatment. This An in vivo study where male rats were exposed to a metformin
effect may be caused by alterations in AMP/ATP ratio, which can dose equivalent to that used for the treatment of T2DM patients (30
modulate the hepatic glucose output upstream of AMPK, inhibiting mg/kg/day for 21 days by oral gavage), reported a significant de-
the AMPK pathway [161]. crease in sperm motility and sperm count [165]. The authors also
described an increase in testicular injury and testicular dysfunction,
IMPACT OF METFORMIN ON MALE REPRODUCTIVE
concurrent with high testicular lipid peroxidation (LPO) levels.
SYSTEM
Metformin administration resulted in a general decrease in the anti-
While the effects of metformin on several organs have been oxidant status of the rat testes due to a reduction in the activities of
broadly studied, little is known about its effects on the human male testicular SOD, CAT and GSH. The histological study of the rat
reproductive system. The vast majority of the in vivo studies that
3626 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Ferreira et al.

Table 1. Summary of main studies reporting metformin-associated effects on male reproductive system.

Biological Study Effects on Male Reproduction


Species Studied Reference
Material Type Metabolic Physiologic

 lactate production
Mouse
In vitro  testosterone levels
Fetal
Human  steroidogenic factors expression
Testicular [162]
Cells  ST diameter
In vivo Mouse  number of SCs
 testes size

 testosterone levels
 ST degeneration
 LH levels
 thickness of the germinal epithelium [163]
 FSH levels
 spermatogenesis index
 insulin levels

 sperm motility
 testes antioxidant status [164]
 sperm concentration
Testes In vivo Rat

 sperm motility
 sperm count
 testes antioxidant status  ST degeneration [165]
 spermatocytes defoliation
 cells involved in spermatogenesis

 lactate production
Sertoli Cells In vitro Rat  GLUT1, GLUT3, MCT4 expression [166]
 antioxidant activity

 sperm motility
 lactate production
Chicken  sperm viability [167]
 AMPK phosphorylation
 acrosome reaction
In vitro
 sperm motility
Mouse  AMPK phosphorylation  sperm viability [156]
 membrane integrity

 sperm motility
Spermatozoa  oxidative stress  sperm count [168]
Rat  normal sperm morphology

 sperm count
 oxidative stress [169]
In vivo  normal sperm morphology

 sperm motility
 testosterone levels  sperm count
Rabbit [170]
 normal sperm morphology
 testicular weight
ST- Seminiferous tubules, SCs - Sertoli cells, GLUT - glucose transporter, MCT - monocarboxylate transporter, LH - luteinizing hormone, FSH - follicle-stimulating hormone,
 - increase,  - decrease.

testes, where the testicular dysfunction was accessed, revealed a sumption of glucose and/or decrease ROS production in these ani-
noticeable necrosis of the testicular tissue, with degeneration of the mals. However, the metformin’s blood glucose lowering effect was
ST and spermatocyte defoliation. Additionally, metformin admini- not taken into account in this study, which may explain (at least) the
stration caused ST atrophy and a decrease in the number of cells results obtained. In addition, the previous effect should be further
involved in spermatogenesis, indicating that the production of ma- evaluated and different concentrations of metformin tested since the
ture spermatozoa was seriously affected in the metformin-treated treated animals do not show blood glucose levels similar to those
animals [165]. Thus, it can be concluded that the dose used may not found in patients with diabetes. Nevertheless, the results described
be able to activate AMPK pathway and thereby increase the con- in these studies were obtained by exposing healthy animals with
Metformin and Male Fertility Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3627

normal (lower) levels of blood glucose to metformin, which may formin on male reproductive function in diabetic and obese rats, by
explain (at least partly) the deleterious reproductive effects ob- improving steroidogenesis and the antioxidant ability of the testes,
served. Therefore, several studies have been performed to deter- leading to an increase in the spermatogenesis index and increased
mine the effect of this therapeutic drug on testicular cells or tissue sperm concentration and motility [163, 164]. Consequently, these
and on male reproductive function in diabetic conditions. In a re- data illustrates that metformin acts primarily by decreasing the oxi-
cent study, Alves and collaborators evaluated the effect of the expo- dative stress in the testis, and not by improving glucose consump-
sure to increasing doses (5, 50 and 500 M) of metformin, for 50 tion.
hours, on rat SCs metabolism, in a hyperglycemic medium [166]. Some studies have also been performed to evaluate the impact
These cells create the BTB and are targeted by the numerous sub- of the in vitro exposure of spermatozoa to metformin. In a study
stances present in the blood, as is the case of metformin. In this in where mouse spermatozoa were incubated with different doses of
vitro study, the authors showed that 50 M of metformin reduces metformin (50, 500 and 5000 M) for 30 minutes, the authors re-
the mRNA and protein expression of glucose (GLUT1 and GLUT3) ported that this drug did not induce any deleterious effect on sper-
and lactate (MCT4) transporters. For the same concentration of matozoa quality either sperm viability, membrane integrity or acro-
metformin, authors also found an increase in LDH activity accom- some reaction, except a slight reduction in sperm motility at 5000
panied by an increased production of lactate and alanine [166], M of metformin due to a reduction of active mitochondria [156].
which indicates a possible AMPK pathway activation by the phar- These authors also investigated the effects of metformin incubation
macological concentration of metformin (Fig. 2). In addition, even (30 minutes) before the sperm cryopreservation. They reported that
on cells treated with the supra-pharmacological dose of metformin viability and membrane integrity were increased in cryopreserved
(500 M), no cytotoxic effects were observed. These results illus- sperm subjected to this pre-incubation. This study also evidenced
trate that metformin decreases the number of SCs’ glycolysis- that this pre-incubation before cryopreservation has a positive im-
related transporters, but increases SCs activity, also indicating that pact in the fertilization ability of cryopreserved mouse spermatozoa
the glycolytic pathway is enhanced. However, glucose consumption due to a significantly increase in the percentage of fertilized oocytes
was maintained in these cells. Furthermore, their data showed that [156]. Furthermore, the pre-treatment with 5000 M of metformin
metformin has the capacity to induce the antioxidant activity, in reduced the percentage of fragmented embryos and increased the
response to increased production of alanine, in rat SCs exposed to a number of oocytes reaching the 4 cell-stage embryo. It was also
hyperglycemic condition. Then, the possible activation of AMPK observed that the lower metformin concentrations (50 and 500 M)
by metformin acts primarily by decreasing oxidative stress in rat did not modify the basal phosphorylation of AMPK. However,
SCs and not by glucose consumption. Notably, germ cell develop- frozen spermatozoa pre-treated with 5000 M of metformin pre-
ment is positively regulated by these factors (stimulated lactate sented a 5-fold increase in AMPK phosphorylation [156]. Accord-
production and less oxidative stress) and so the authors suggested ingly, an in vitro study using chicken spermatozoa (1 mM of met-
that metformin may be used as an efficient drug for the treatment of formin for 40 minutes) demonstrated that exposure to metformin
T2DM male individuals on reproductive age, improving their over- significantly improved AMPK phosphorylation, sperm motility,
all fertility status. However, although these results were based on an viability, percentage of sperm able to undergo a successful acro-
in vitro model that attempts to mimic the in vivo conditions they some reaction and lactate production [167], evidencing a possible
need to be further validated, as for instance, by the use of germ beneficial effect of metformin in bird spermatozoa, at least during
cells-Sertoli cells co-cultures. in vitro storage. In fact, the use of metformin in cryopreservation
Still, data obtained in several in vivo studies were concomitant protocols seems to have beneficial effects on the quality of
with the results obtained in this in vitro approach. In fact, several (cryo)preserved sperm, improving its quality and increasing its
authors reported that metformin can improve the spermatogenic fertilization ability, in response to the activation of the AMPK
index and the male reproductive functions of diabetic rats [163, pathway.
164]. A study conducted by Nasrolahi and collaborators evaluated Contradictory results were obtained in in vivo studies. In a
the effects of daily metformin in vivo administration (100 mg/kg), study conducted by Attia and collaborators, authors evaluated the
for a 40 day period, on the testes of streptozotocin-induced diabetic impact of single (100, 500 or 2500 mg/kg) and multiple oral ad-
rats ( 220 mg/dl of blood glucose concentrations) [163]. They ministration (100 or 500 mg/kg/day for 4 and 8 weeks) of met-
reported an increase in testosterone, LH and FSH levels, when formin on sperm quality parameters on a streptozotocin-induced
compared with non-treated rats. In the same study, the authors ob- diabetic rat model (blood glucose levels >350 mg/dL) [168]. In all
served that, when administered as a single agent or in a combina- the doses tested, metformin was neither genotoxic nor cytotoxic for
tion therapy with honey, metformin produced a significant increase the spermatozoa. No significant effects were observed with the
of the percentage of ST with a positive spermatogenesis index and single administration. However, the two doses of metformin used in
of the germinal epithelium thickness. Co-treatment also reduced the multiple treatment attenuated the sperm oxidative stress, commonly
ST degeneration caused by DM and increased its diameter. The observed on diabetic rats caused by ROS overproduction, with in-
results obtained by these authors suggested that the administration creased levels of GSH and decreased levels of LPO. In fact, the
of metformin alone or in combination with honey improves the multiple treatment significantly increased sperm motility, count and
male reproductive function of diabetic rats and has the ability of the percentage of morphologically normal sperm, in a dose-
reducing the detrimental changes induced by DM in testicular tissue dependent manner, with significantly alterations observed with 500
[163]. Fang and collaborators reported concurrent results when mg/kg [168]. Similar data were described by Rabbani and collabo-
evaluating the effect of metformin, for 12 weeks, on the epididymal rators, who reported an increase on sperm count and on the general
sperm quality and testicular antioxidant capacity in obese rodent antioxidant status, and a decrease on the percentage of sperm with
models [164], which is closely associated with the development of abnormal morphology, when metformin (50 mg/kg) is co-
DM. These authors reported that metformin-treated rats presented administered orally daily for 4 weeks with pioglitazone (1 mg/kg)
an increase in sperm concentration and motility. Furthermore, met- to streptozotocin-induced diabetic rats (blood glucose levels >230
formin-treated obese rats displayed an improvement in the oxida- mg/kg) [169]. Although these authors have not evaluated the activa-
tion status of the testes, evidenced by the reduction of LPO levels. tion of AMPK pathway, they observed a reduction in the production
These results illustrate that a combination of metformin with an of ROS. However, these results demonstrate that metformin may
appropriate diet enhances sperm quality and promotes the antioxi- lead to the AMPK pathway activation preferentially leading to a
dant ability of the testes of obese rats, thereby attenuating the ef- reduction of oxidative stress rather than changing glucose metabo-
fects of obesity on the male reproductive function [164]. In fact, the lism. So, in response to this alteration, in the last two studies, met-
data obtained on these studies confirmed the protective role of met-
3628 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Ferreira et al.

Extracellular Space
Glucose
Glucose
T1
GLU GL
UT
3

Glycolysis
PFK LDH
Glucose Pyruvate Lactate

ALT Sertoli Cell

Alanine Lactate
Lactate
-oxidation Pyruvate

4
CT Lactate
Acetyl-CoA M MCT 2

Mitochondria Germ Cell


Lactate

Sertoli cell nucleus


ATP

Fig. (2). Schematic illustration of the main metabolic pathways described in Sertoli cells (SCs) and the metformin actions on the metabolism of these cells.
The extracellular glucose enters via glucose transporters (GLUT1 and GLUT3) and is converted into pyruvate by phosphofructokinase (PFK) action. Pyruvate
can either be transported into the mitochondrial matrix to form Acetyl-coA, or can be converted into lactate or alanine by lactate dehydrogenase (LDH) and
alanine transaminase (ALT), respectively. SCs actively consume glucose producing large quantities of lactate, for the developing germ cells, which is then
exported to the extracellular medium by proton-linked plasma membrane transporters, the monocarboxylate transporters (MCT2 and MCT4). SCs incubation
with metformin results in decreased mRNA and proteins levels of GLUT1, GLUT3 and MCT4, increased activity of LDH and improves the production of
lactate and alanine by these cells.
Dotted lines - metabolism steps affected by metformin action in SCs

formin was able to reduce the genomic alterations and the damages the other hand, the male reproductive capacity of individuals with
induced by oxidative stress. normal glucose levels was negatively affected. As discussed, there
Dissonant results were however described in a study performed are some contradictory results concerning the safety and therapeutic
on normal and alloxan-induced diabetic rabbits (blood glucose lev- potential of metformin in male reproduction. The different out-
els >400 mg/kg) treated with a high dose (120 mg/kg) of met- comes may be explained by the wide variety of species studied and
formin, once a day for 3 months [170]. Treated non-diabetic rabbits the diverse experimental conditions. Nonetheless, it was also possi-
displayed a significant reduction in testicular weight (which can be ble to verify that AMPK phosphorylation appears to play important
explained by decreased cell proliferation triggered by AMPK acti- functions in the regulation of sperm parameters and metabolism.
vation), in sperm motility and count and serum testosterone level Nevertheless, further studies are needed to elucidate the exact influ-
with a significant increase in abnormal and dead sperm. However, ence of metformin on male reproduction. Even so, from the major-
the greatest reduction in these parameters was observed in met- ity of data available it seems evident that metformin improves the
formin-treated diabetic rabbits [170]. In face of their data, authors antioxidant activity of male reproductive system, by reduction of
suggested that this dose of metformin might potentiate the effect of ROS production, due to the AMPK pathway activation. Less fre-
DM, impacting negatively the male reproductive function [170]. quently, the activation of AMPK pathway by metformin treatment
These results are not in agreement with the ones obtained in some was also able to improve the glycolytic pathway, which increased
of the in vivo studies mentioned above using other animal models, lactate production leading to an improvement of the spermatogenic
particularly using rodent models. Still, the method of induction of process. However, it has to be noted that not all studies have evalu-
DM in rabbits was distinct as that used in rodent. Although, in both ated simultaneously the AMPK activation with the glucose metabo-
rodents and rabbits, metformin has been capable of lowering the lism and/or oxidative stress. The most promising results were ob-
glucose plasma levels to control levels, in rabbits the dose used may tained with mouse spermatozoa, where metformin treatment acti-
not have been enough to improve sperm parameters thus leading to vated the AMPK pathway, improving the quality of frozen sperm
adverse effects on male fertility [170]. and consequently the fertilizing capacity of cryopreserved sper-
matozoa. This data is of great importance and additional research is
In vivo exposure to metformin only had beneficial effects in the needed for future application of metformin on human reproduction.
male reproductive function of diabetic animals, particularly in
mammals, except the case of alloxan-induced diabetic rabbits. On
Metformin and Male Fertility Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3629

MOLECULAR MECHANISMS OF METFORMIN ACTION high percentage of morphological abnormalities. However, no al-
IN THE TESTES teration in testes morphology or sperm production rate was verified.
The molecular pathways by which metformin may employ its These animals also presented high levels of androgens, with in-
potential actions on male reproductive system remain elusive. Cur- creased intra-testicular cholesterol and testosterone concentrations.
rently, few studies are available reporting the effects of metformin Furthermore, the LH and FSH levels were lower in KO mice due to
on male reproduction and, specifically, on human sperm quality negative feedback induced by increased levels of testosterone.
parameters. Nonetheless, some studies on animal models have Therefore, the lack of AMPK activation seems to result in an in-
emerged in an attempt to identify the effect of this drug on male crease of androgens levels, which can influence sperm quality, and
fertility and reproductive health. consequently male fertility [181]. As previously referred, a study
with male chickens reported that AMPK is also present in ejacu-
Several authors reported that metformin can indirectly influence lated spermatozoa. This enzyme was located in the spermatozoon
the activation of the AMPK pathway and an association between acrosomal region, midpiece and flagellum. The AMPK active form,
the improvement of male reproductive functions and AMPK activa- phospho-Thr172-AMPK, was mainly observed in the flagellum and
tion on testicular tissue was recently reported [152, 167]. Accord- acrosome region, while a reduced enzyme protein expression was
ingly, some authors have studied the activity of AMPK in different observed in the midpiece of chikens’ spermatozoa [167]. These
testicular cell types, with and without metformin action, to better results seem to explain why ATP production is not restricted to the
clarify the mechanisms of this drug on testicular tissue. AMPK acts spermatozoa midpiece (mitochondrial respiration), but can likewise
as a sensor and regulator of cell metabolism [171] and is a hetero- occur in other locations along spermatozoa through anaerobic gly-
trimeric complex composed by one catalytic unit (AMPK) and colysis [185, 186]. The expression pattern of AMPK suggests that
two regulatory subunits ( and ) [172, 173]. Isoforms of all three this kinase may be closely related with the promotion of spermato-
subunits (1, 2, 1, 2, 1, 2 and 3) have been identified in zoa motility and acrosome reaction process [167].
mammals [174]. A decline in cellular energy status, and increased
intracellular levels of AMP, lead to the binding of AMP to the regu- Indeed, a mechanism of action involving AMPK has been pro-
latory  subunit of the AMPK complex. This association leads to a posed for metformin on the chicken male reproductive tract. In this
conformational change of the AMPK complex that promotes its model, metformin induced the inhibition of the mitochondrial respi-
activation via phosphorilation of the catalytic subunit in the ratory chain complex I and, in response to the decrease of ATP
threonine 172 residue (Thr172) by an upstream LKB1 (100-fold production and the increase in AMP intracellular level, AMPK was
increase in activity) [173, 175, 176], and inhibition of its dephos- activated [151]. AMPK activation regulates ATP levels, improving
phorilation by protein phosphatases [177, 178]. the metabolic functions needed to ensure high energy consuming
processes, such as sperm motility and acrosome reaction [167].
AMPK can be found in several mammalian tissues were it is Furthermore, activation of the AMPK pathway leads to increased
responsible for the activation of some metabolic pathways involved sperm motility, as well as increased lactate production. Still, this
in the activation of ATP producing pathways (glucose and lipid effect is not equivalent among species. Although metformin acts as
catabolism), and inhibition of ATP consuming anabolic pathways AMPK modulator, altering sperm ATP levels and function in sev-
(protein, fatty acid and cholesterol synthesis) [178, 179]. It has been eral animals, the kinetics and magnitude of these modifications is
shown that AMPK is present in reproductive cells like in boar variable. For instance, in the chickens these modifications occur
ejaculated sperm [171] and tissues of some mammals including the faster than in boars. Therefore, it has been suggested that the rele-
rodent testis [180, 181]. Regardless of the fact that all the subunits vance of AMPK pathway on male reproductive function differs
of AMPK are expressed in rat testis, 1AMPK is responsible for between mammals and birds. Yet, it is widelly accepted that this
most of the total AMPK activity [180]. Notably, it has been de- pathway is susceptible of activation by metformin in the cells of the
scribed that rodent haploid spermatids have a specific isoform of male reproductive tract, particularly in spermatozoa.
upstream LKB1, called LKB1s, responsible for AMPK activation,
illustrating the relevance of this pathway on male fertility. It was In summary, activation of the AMPK pathway can improve
also reported in mice that the lack of LKB1 can result in a decrease some sperm parameters, like motility and morphology, thereby
of spermatozoa production leading to a reduction of mature sper- improving the overall male fertility status. This activation is also
matozoa in the epididymis, with an increase on the amount of mor- able to stimulate the acrosome reaction and lactate production, and
phologically abnormal and non-motile spermatozoa [182]. maintain normal hormone levels, which conduct to an efficient
spermatogenesis.
The existence of high levels of AMPK was noticed while
studying cultures of rat SCs. In these cells, activation of AMPK CONCLUSION
pathway leads to an increase of lactate production, in response to an
Metformin is an oral antidiabetic with anti-hyperglicemic activ-
increase in glucose uptake. The rise in glucose uptake and lactate
ity. Recently, it has been reported that this drug can act in different
production results of an increase in the GLUT1 and MCT4 levels.
organs and tissues including the male reproductive system. How-
These data led to the hypothesis that the AMPK pathway is respon-
ever, the existing literature presents some contradictory results on
sible for the nutritional modulation of SCs function [9, 183]. A
the impact of metformin on this system. While several studies pro-
different study revealed that AMPK is expressed in boar spermato-
vide evidence that metformin improves sperm motility, others do
zoa at higher levels than those found in somatic cells. Moreover, it
not confirm this observation. Analysis of the antioxidant status,
was reported that the inhibition of this enzyme in boar spermatozoa
lactate production, LDH activity and sperm count, likewise re-
resulted in a decrease of sperm motility, and fertilization capacity
vealed opposing results. The differences found between all experi-
[171]. More recently, the same authors reported that AMPK is also
ments can justify the contraditory results observed. Nevertheless, it
involved in plasma membrane organization, since its inhibition
is accepted that metformin improves the reproductive functions of
induced plasma membrane lipid disorganization and reduced the
diabetic individuals, although the same was not always verified in
outward translocation of phosphatidylserine at the plasma mem-
normal individuals. Additionally, the incubation of spermatozoa
brane of spermatozoa [184]. Thus, it has been suggested that
with metformin improves their fertilization capacity after
AMPK is involved in the function of plasma membrane of mam-
cryopreservation. However, data from studies involving humans or
malian spermatozoa at different levels, playing a key role on the
human material are scarse and it is important to investigate the
production of normal spermatozoa. Tartarin and collaborators stud-
pharmacological and toxicological effects of metformin incubation
ied the role of this kinase on male reproductive function using
on human spermatozoa and reproductive function.
1AMPK KO male mice [181]. They reported a significant reduc-
tion in fertility capacity by induction of asthenozoospermia and a
3630 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Ferreira et al.

The metformin mechanism of action is not yet fully understood. [16] Chiodini I, Di Lembo S, Morelli V, et al. Hypothalamic-pituitary-
However, there are evidences that this drug acts via AMPK path- adrenal activity in type 2 diabetes mellitus: role of autonomic im-
way in numerous tissues, including the male reproductive system. balance. Metabolism 2006; 55: 1135-40.
Activation of the AMPK pathway can improve some sperm pa- [17] Nathan DM, Buse JB, Davidson MB, et al. Medical management of
hyperglycaemia in type 2 diabetes mellitus: a consensus algorithm
rameters, as is the case of motility by activation of anaerobic path- for the initiation and adjustment of therapy: a consensus statement
ways involved in ATP production. The AMPK pathway is also from the American Diabetes Association and the European Asso-
involved in a reduction of morphologically abnormal sperm accom- ciation for the Study of Diabetes. Diabetologia 2009; 52: 17-30.
panied with increased lactate production [9, 167, 182]. These altera- [18] Rosenbloom AL, Joe JR, Young RS, et al. Emerging Epidemic of
tions seem to be caused by a reduction in ROS production in re- Type 2 Diabetes in Youth. Diabetes Care 1999; 22: 345-54.
sponse to AMPK activation, which improves sperm quality. In most [19] Dias TR, Alves MG, Silva BM, et al. Sperm glucose transport and
cases of the reported studies, the action of metformin seems to be metabolism in diabetic individuals. Mol Cell Endocrinol 2014; 396:
responsible for the decrease of oxidative stress rather than for an 37-45.
increase in glycolysis rate. Nonetheless, the studies conducted ob- [20] He ZX, Zhou ZW, Yang Y, et al. Overview of clinically approved
oral antidiabetic agents for the treatment of type 2 diabetes melli-
tained different results for the effects of metformin on male fertility. tus. Clin Exp Pharmacol Physiol 2015; 42: 125-38.
Therefore, additional studies are needed in order to fully understand [21] Chao C, Liu S. Diabetes Mellitus Treatment In: Heggenhougen K,
the influence of metformin on the molecular mechanisms involved Quah S, Eds. International encyclopedia of public health. 2nd ed.
on its effects on the male reproductive system and consequently on San Diego: Academic Press; 2008; pp. 153-60
male fertility and the role of the AMPK pathway on sperm func- [22] Inzucchi SE, Bergenstal RM, Buse JB, et al. Management of hy-
tions. perglycaemia in type 2 diabetes: a patient-centered approach. Posi-
tion statement of the American Diabetes Association (ADA) and
CONFLICT OF INTEREST the European Association for the Study of Diabetes (EASD). Dia-
betologia 2012; 55: 1577-96.
The authors declare that they have no conflict of interest. [23] Rena G, Pearson ER, Sakamoto K. Molecular mechanism of action
of metformin: old or new insights? Diabetologia 2013; 56: 1898-
ACKNOWLEDGEMENTS 906.
UMIB (Pest-OE/SAU/UI0215/2014) and MG Alves (SFRH/ [24] Wu N, Gu HJ, Li Q. Effects of antidiabetic drug metformin on the
BPD/80451/2011) were funded by National Funds through FCT- migration and invasion abilities of human pulmonary adenocarci-
Foundation for Science and Technology and AR by the Master noma A549 cell line in vitro. J Thorac Dis 2010; 2: 76-80.
Course in Molecular Medicine and Oncology, FMUP. [25] Kato K, Gong J, Iwama H, et al. The antidiabetic drug metformin
inhibits gastric cancer cell proliferation in vitro and in vivo. Mol
REFERENCES Cancer Ther 2012; 11: 549-60.
[26] Creanga AA, Bradley HM, McCormick C, et al. Use of metformin
[1] WHO laboratory manual for the examination and processing of in polycystic ovary syndrome: a meta-analysis. Obstet Gynecol
human semen. 5th ed: World Health Organization 2010. 2008; 111: 959-68.
[2] de Kretser DM. Male infertility. Lancet 1997; 349: 787-90. [27] Diamanti-Kandarakis E, Christakou CD, Kandaraki E, et al. Met-
[3] Abid S, Maitra A, Meherji P, et al. Clinical and laboratory evalua- formin: an old medication of new fashion: evolving new molecular
tion of idiopathic male infertility in a secondary referral center in mechanisms and clinical implications in polycystic ovary syn-
India. J Clin Lab Anal 2008; 22: 29-38. drome. Eur J Endocrinol 2010; 162: 193-212.
[4] Punab M, Zilaitiene B, Jorgensen N, et al. Regional differences in [28] Bertoldo MJ, Faure M, Dupont J, et al. Impact of metformin on
semen qualities in the Baltic region. Int J Androl 2002; 25: 243-52. reproductive tissues: an overview from gametogenesis to gestation.
[5] Fernandez MF, Duran I, Olea N, et al. Semen quality and reproduc- Ann Transl Med 2014; 2: 55.
tive hormone levels in men from Southern Spain. Int J Androl [29] Caroppo E. Male reproductive medicine: anatomy and physiology.
2012; 35: 1-10. In: Niederberger I, Craig S, Eds. An introduction to male reproduc-
[6] Jorgensen N, Andersen AG, Eustache F, et al. Regional differences tive medicine. USA, Cambrige, New York: Cambridge University
in semen quality in Europe. Hum Reprod 2001; 16: 1012-9. Press; 2011; pp. 1-28.
[7] Jorgensen N, Carlsen E, Nermoen I, et al. East-West gradient in [30] Rato L, Alves MG, Socorro S, et al. Metabolic regulation is impor-
semen quality in the Nordic-Baltic area: a study of men from the tant for spermatogenesis. Nat Rev Urol 2012; 9: 330-8.
general population in Denmark, Norway, Estonia and Finland. [31] Alves MG, Rato L, Carvalho RA, et al. Hormonal control of Sertoli
Hum Reprod 2002; 17: 2199-208. cell metabolism regulates spermatogenesis. Cell Mol Life Sci 2013;
[8] Tournaye HJ, Cohlen BJ. Management of male-factor infertility. 70: 777-93.
Best Pract Res Clin Obstet Gynaecol 2012; 26: 769-75. [32] Sharpe RM, McKinnell C, Kivlin C, et al. Proliferation and func-
[9] Galardo MN, Riera MF, Pellizzari EH, et al. The AMP-activated tional maturation of Sertoli cells, and their relevance to disorders of
protein kinase activator, 5-aminoimidazole-4-carboxamide-1-b-D- testis function in adulthood. Reproduction 2003; 125: 769-84.
ribonucleoside, regulates lactate production in rat Sertoli cells. J [33] Rato L, Socorro S, Cavaco JE, et al. Tubular fluid secretion in the
Mol Endocrinol 2007; 39: 279-88. seminiferous epithelium: ion transporters and aquaporins in Sertoli
[10] Karagiannis A, Harsoulis F. Gonadal dysfunction in systemic dis- cells. J Membr Biol 2010; 236: 215-24.
eases. Eur J Endocrinol 2005; 152: 501-13. [34] Johnson MH. Making sperm. In: Johnson MH, Eds. Essential re-
[11] Bhattacharya SM, Ghosh M, Nandi N. Diabetes mellitus and ab- production. 7th ed. UK: Blackwell Publishing 2013; pp. 106-21.
normalities in semen analysis. J Obstet Gynaecol Res 2014; 40: [35] Walker WH, Cheng J. FSH and testosterone signaling in Sertoli
167-71. cells. Reproduction 2005; 130: 15-28.
[12] Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the preva- [36] Dym M, Fawcett DW. The blood-testis barrier in the rat and the
lence of diabetes for 2010 and 2030. Diabetes Res Clin Pract 2010; physiological compartmentation of the seminiferous epithelium.
87: 4-14. Biol Reprod 1970; 3: 308-26.
[13] Whiting DR, Guariguata L, Weil C, et al. IDF diabetes atlas: global [37] Setchell BP, Voglmayr JK, Waites GM. A blood-testis barrier
estimates of the prevalence of diabetes for 2011 and 2030. Diabetes restricting passage from blood into rete testis fluid but not into
Res Clin Pract 2011; 94: 311-21. lymph. J Physiol 1969; 200: 73-85.
[14] Nuttall SL, Dunne F, Kendall MJ, et al. Age-independent oxidative [38] Russell L, Ettlin R, Sinha Hikim A, et al. Histological and histopa-
stress in elderly patients with non-insulin-dependent diabetes melli- thological evaluation of the testis. Clearwater: Cache River Press
tus. QJM 1999; 92: 33-8. 1990.
[15] Ballester J, Munoz MC, Dominguez J, et al. Insulin-dependent [39] Griswold M, McLean D. Knobil and Neill’s physiology of repro-
diabetes affects testicular function by FSH- and LH-linked mecha- duction. San Diego: Elsevier 2006.
nisms. J Androl 2004; 25: 706-19. [40] Mruk DD, Cheng CY. Sertoli-Sertoli and Sertoli-germ cell interac-
tions and their significance in germ cell movement in the
Metformin and Male Fertility Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3631

seminiferous epithelium during spermatogenesis. Endocr Rev [65] Ulisse S, Jannini EA, Pepe M, et al. Thyroid hormone stimulates
2004; 25: 747-806. glucose transport and GLUT1 mRNA in rat Sertoli cells. Mol Cell
[41] Setchell BP, Scott TW, Voglmayr JK, et al. Characteristics of Endocrinol 1992; 87: 131-7.
testicular spermatozoa and the fluid which transports them into the [66] Reagan LP, Gorovits N, Hoskin EK, et al. Localization and regula-
epididymis. Biol Reprod 1969; 1: 40-66. tion of GLUTx1 glucose transporter in the hippocampus of strepto-
[42] Mita M, Hall PF. Metabolism of round spermatids from rats: lactate zotocin diabetic rats. Proc Natl Acad Sci USA 2001; 98: 2820-5.
as the preferred substrate. Biol Reprod 1982; 26: 445-55. [67] Oliveira PF, Alves MG, Rato L, et al. Effect of insulin deprivation
[43] Courtens JL, Ploen L. Improvement of spermatogenesis in adult on metabolism and metabolism-associated gene transcript levels of
cryptorchid rat testis by intratesticular infusion of lactate. Biol Re- in vitro cultured human Sertoli cells. Biochim Biophys Acta 2012;
prod 1999; 61: 154-61. 1820: 84-89.
[44] Erkkila K, Aito H, Aalto K, et al. Lactate inhibits germ cell apopto- [68] Riera MF, Galardo MN, Pellizzari EH, et al. Molecular mecha-
sis in the human testis. Mol Hum Reprod 2002; 8: 109-17. nisms involved in Sertoli cell adaptation to glucose deprivation.
[45] Nakamura M, Hino A, Yasumasu I, et al. Stimulation of protein Am J Physiol Endocrinol Metab 2009; 297: 907-14.
synthesis in round spermatids from rat testes by lactate. J Biochem [69] Kreisberg RA. Lactate homeostasis and lactic acidosis. Ann Intern
(Tokyo) 1981; 89: 1309-15. Med 1980; 92: 227-37.
[46] Jutte NH, Grootegoed JA, Rommerts FF, et al. Exogenous lactate is [70] Halestrap AP, Price NT. The proton-linked monocarboxylate trans-
essential for metabolic activities in isolated rat spermatocytes and porter (MCT) family: structure, function and regulation. Biochem J
spermatids. J Reprod Fertil 1981; 62: 399-405. 1999; 343(Pt 2): 281-99.
[47] Cheng CY, Mruk DD. A local autocrine axis in the testes that regu- [71] Adijanto J, Philp NJ. The SLC16A family of monocarboxylate
lates spermatogenesis. Nat Rev Endocrinol 2010; 6: 380-95. transporters (MCTs)--physiology and function in cellular metabo-
[48] Tilbrook AJ, Clarke IJ. Negative feedback regulation of the secre- lism, pH homeostasis, and fluid transport. Curr Top Membr 2012;
tion and actions of gonadotropin-releasing hormone in males. Biol 70: 275-311.
Reprod 2001; 64: 735-42. [72] Xiong W, Wang H, Wu H, et al. Apoptotic spermatogenic cells can
[49] Hess R, Franca L. Sepermatogenesis and Cycle of the Seminiferous be energy sources for Sertoli cells. Reproduction 2009; 137: 469-
Epithelium In: Cheng CY, Eds. Molecular mechanisms in sper- 79.
matogenesis. Advances in Experimental Medicine and Biology. [73] Leiderman B, Mancini RE. Glycogen content in the rat testis from
636. USA: Landes Bioscience and Springer Science+Business Me- postnatal to adult ages. Endocrinology 1969; 85: 607-9.
dia, LLC; 2008. p. 1-15. [74] Yanagimachi R. Mammalian fertilization. New York: Raven Press
[50] Cheng CY, Mruk DD. An intracellular trafficking pathway in the Ltd 2008.
seminiferous epithelium regulating spermatogenesis: a biochemical [75] Rigau T, Rivera M, Palomo MJ, et al. Differential effects of glu-
and molecular perspective. Crit Rev Biochem Mol Biol 2009; 44: cose and fructose on hexose metabolism in dog spermatozoa. Re-
245-63. production 2002; 123: 579-91.
[51] Alukal JP, Lamb DJ, Niederberger CS, et al. Spermatogenesis in [76] Medrano A, Garcia-Gil N, Ramio L, et al. Hexose-specificity of
the adult In: Lipshultz LI, Howards SS, Niederberger CS, Eds. In- hexokinase and ADP-dependence of pyruvate kinase play impor-
fertility in the Male 4th ed. USA: Cambridge University Press 2009; tant roles in the control of monosaccharide utilization in freshly di-
pp. 74-89. luted boar spermatozoa. Mol Reprod Dev 2006; 73: 1179-94.
[52] Bucci D, Rodriguez-Gil JE, Vallorani C, et al. GLUTs and mam- [77] Bucci D, Isani G, Spinaci M, et al. Comparative immunolocaliza-
malian sperm metabolism. J Androl 2011; 32: 348-55. tion of GLUTs 1, 2, 3 and 5 in boar, stallion and dog spermatozoa.
[53] Grootegoed JA, Oonk RB, Jansen R, et al. Metabolism of radiola- Reprod Domest Anim 2010; 45: 315-22.
belled energy-yielding substrates by rat Sertoli cells. J Reprod Fer- [78] Bucci D, Spinaci M, Vallorani C, et al. Detection and localization
til 1986; 77: 109-18. of GLUTs 1, 2, 3 and 5 in donkey spermatozoa. Reprod Domest
[54] Nakamura M, Okinaga S, Arai K. Metabolism of round spermatids: Anim 2010; 45: 217-20.
evidence that lactate is preferred substrate. Am J Physiol 1984; [79] Schurmann A, Axer H, Scheepers A, et al. The glucose transport
247: 234-42. facilitator GLUT8 is predominantly associated with the acrosomal
[55] Scheepers A, Joost HG, Schurmann A. The glucose transporter region of mature spermatozoa. Cell Tissue Res 2002; 307: 237-42.
families SGLT and GLUT: molecular basis of normal and aberrant [80] Gomez O, Romero A, Terrado J, et al. Differential expression of
function. JPEN J Parenter Enteral Nutr 2004; 28: 364-71. glucose transporter GLUT8 during mouse spermatogenesis. Repro-
[56] Joost HG, Thorens B. The extended GLUT-family of sugar/polyol duction 2006; 131: 63-70.
transport facilitators: nomenclature, sequence characteristics, and [81] Kim ST, Moley KH. The expression of GLUT8, GLUT9a, and
potential function of its novel members (review). Mol Membr Biol GLUT9b in the mouse testis and sperm. Reprod Sci 2007; 14: 445-
2001; 18: 247-56. 55.
[57] Angulo C, Rauch MC, Droppelmann A, et al. Hexose transporter [82] Frenkel G, Peterson RN, Freund M. Changes in the metabolism of
expression and function in mammalian spermatozoa: cellular local- guinea pig sperm from different segments of the epididymis. Proc
ization and transport of hexoses and vitamin C. J Cell Biochem Soc Exp Biol Med 1973; 143: 1231-6.
1998; 71: 189-203. [83] Mann T. Metabolism of semen. Adv Enzymol Relat Areas Mol
[58] Boussouar F, Benahmed M. Lactate and energy metabolism in Biol 1947: 329-83.
male germ cells. Trends Endocrinol Metab 2004; 15: 345-50. [84] Storey BT. Mammalian sperm metabolism: oxygen and sugar,
[59] Robinson R, Fritz IB. Metabolism of glucose by Sertoli cells in friend and foe. Int J Dev Biol 2008; 52: 427-37.
culture. Biol Reprod 1981; 24: 1032-41. [85] Gomez M, Navarro-Sabate A, Manzano A, et al. Switches in 6-
[60] Jutte NH, Jansen R, Grootegoed JA, et al. Regulation of survival of phosphofructo-2-kinase isoenzyme expression during rat sperm
rat pachytene spermatocytes by lactate supply from Sertoli cells. J maturation. Biochem Biophys Res Commun 2009; 387: 330-5.
Reprod Fertil 1982; 65: 431-8. [86] Coonrod S, Vitale A, Duan C, et al. Testis-specific lactate dehy-
[61] Oliveira PF, Alves MG, Rato L, et al. Influence of 5alpha- drogenase (LDH-C4; Ldh3) in murine oocytes and preimplantation
dihydrotestosterone and 17beta-estradiol on human Sertoli cells embryos. J Androl 2006; 27: 502-9.
metabolism. Int J Androl 2011; 34: 612-20. [87] Li SS, O'Brien DA, Hou EW, et al. Differential activity and syn-
[62] Rato L, Alves MG, Socorro S, et al. Metabolic modulation induced thesis of lactate dehydrogenase isozymes A (muscle), B (heart),
by oestradiol and DHT in immature rat Sertoli cells cultured in and C (testis) in mouse spermatogenic cells. Biol Reprod 1989; 40:
vitro. Biosci Rep 2012; 32: 61-69. 173-80.
[63] Carosa E, Radico C, Giansante N, et al. Ontogenetic profile and [88] Alves MG, Oliveira PF, Socorro S, et al. Impact of diabetes in
thyroid hormone regulation of type-1 and type-8 glucose transport- blood-testis and blood-brain barriers: resemblances and differences.
ers in rat Sertoli cells. Int J Androl 2005; 28: 99-106. Curr Diabetes Rev 2012; 8: 401-12.
[64] Galardo MN, Riera MF, Pellizzari EH, et al. Regulation of expres- [89] American Diabetes A. Diagnosis and classification of diabetes.
sion of Sertoli cell glucose transporters 1 and 3 by FSH, IL1 beta, Diabetes Care 2014; 37: 590.
and bFGF at two different time-points in pubertal development. [90] Grieco FA, Sebastiani G, Spagnuolo I, et al. Immunology in the
Cell Tissue Res 2008; 334: 295-304. clinic review series; focus on type 1 diabetes and viruses: how viral
3632 Current Pharmaceutical Design, 2015, Vol. 21, No. 25 Ferreira et al.

infections modulate beta cell function. Clin Exp Immunol 2012; [118] Union OE. Pharmaceutical Consumption Health at a Glance:
168: 24-29. Europe 2014 Paris: OECD Publishing 2014. pp. 61-84.
[91] Burul-Bozkurt N, Pekiner C, Kelicen P. Diabetes alters aromatase [119] Hemmingsen B, Lund SS, Wetterslev J, et al. Oral hypoglycaemic
enzyme levels in gonadal tissues of rats. Naunyn Schmiedebergs agents, insulin resistance and cardiovascular disease in patients
Arch Pharmacol 2010; 382: 33-41. with type 2 diabetes. Eur J Endocrinol 2009; 161: 1-9.
[92] Silink M. Childhood Diabetes: A Global Perspective. Horm Res [120] Bailey CJ, Campbell IW, Chan JCN, et al. Metformin. The gold
2002; 57: 1-5. standard. A scientific handbook. UK: Wiley (Chichester) 2008.
[93] Agbaje IM, Rogers DA, McVicar CM, et al. Insulin dependant [121] Gunton JE, Delhanty PJ, Takahashi S, et al. Metformin rapidly
diabetes mellitus: implications for male reproductive function. increases insulin receptor activation in human liver and signals
Hum Reprod 2007; 22: 1871-7. preferentially through insulin-receptor substrate-2. J Clin Endocri-
[94] Kahn SE. Clinical review 135: The importance of beta-cell failure nol Metab 2003; 88: 1323-32.
in the development and progression of type 2 diabetes. J Clin En- [122] Palomba S, Falbo A, Zullo F, et al. Evidence-based and potential
docrinol Metab 2001; 86: 4047-58. benefits of metformin in the polycystic ovary syndrome: a compre-
[95] Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity hensive review. Endocr Rev 2009; 30: 1-50.
to insulin resistance and type 2 diabetes. Nature 2006; 444: 840-6. [123] Sivalingam VN, Myers J, Nicholas S, et al. Metformin in reproduc-
[96] Edelstein SL, Knowler WC, Bain RP, et al. Predictors of progres- tive health, pregnancy and gynaecological cancer: established and
sion from impaired glucose tolerance to NIDDM: an analysis of six emerging indications. Hum Reprod Update 2014: 1-16.
prospective studies. Diabetes 1997; 46: 701-10. [124] Martineau LC. Large enhancement of skeletal muscle cell glucose
[97] Bock G, Dalla Man C, Campioni M, et al. Pathogenesis of pre- uptake and suppression of hepatocyte glucose-6-phosphatase activ-
diabetes: mechanisms of fasting and postprandial hyperglycemia in ity by weak uncouplers of oxidative phosphorylation. Biochim
people with impaired fasting glucose and/or impaired glucose tol- Biophys Acta 2012; 1820: 133-50.
erance. Diabetes 2006; 55: 3536-49. [125] Sakar Y, Meddah B, Faouzi MA, et al. Metformin-induced regula-
[98] Rosenbloom AL, Joe JR, Young RS, et al. Emerging epidemic of tion of the intestinal D-glucose transporters. J Physiol Pharmacol
type 2 diabetes in youth. Diabetes Care 1999; 22: 345-54. 2010; 61: 301-7.
[99] Baccetti B, La Marca A, Piomboni P, et al. Insulin-dependent dia- [126] Shank JJ, Yang K, Ghannam J, et al. Metformin targets ovarian
betes in men is associated with hypothalamo-pituitary derangement cancer stem cells in vitro and in vivo. Gynecol Oncol 2012; 127:
and with impairment in semen quality. Hum Reprod 2002; 17: 390-7.
2673-7. [127] Rattan R, Giri S, Hartmann LC, et al. Metformin attenuates ovarian
[100] Sexton WJ, Jarow JP. Effect of diabetes mellitus upon male repro- cancer cell growth in an AMP-kinase dispensable manner. J Cell
ductive function. Urology 1997; 49: 508-13. Mol Med 2011; 15: 166-78.
[101] Schoeffling K, Federlin K, Ditschuneit H, et al. Disorders of sexual [128] Rattan R, Graham RP, Maguire JL, et al. Metformin suppresses
function in male diabetics. Diabetes 1963; 12: 519-27. ovarian cancer growth and metastasis with enhancement of cis-
[102] Bourne RB, Kretzschmar WA, Esser JH. Successful artificial in- platin cytotoxicity in vivo. Neoplasia 2011; 13: 483-91.
semination in a diabetic with retrograde ejaculation. Fertil Steril [129] Yasmeen A, Beauchamp MC, Piura E, et al. Induction of apoptosis
1971; 22: 275-7. by metformin in epithelial ovarian cancer: involvement of the Bcl-2
[103] Fedele D. Therapy Insight: sexual and bladder dysfunction associ- family proteins. Gynecol Oncol 2011; 121: 492-8.
ated with diabetes mellitus. Nat Clin Pract Urol 2005; 2: 282-90. [130] Cantrell LA, Zhou C, Mendivil A, et al. Metformin is a potent
[104] De Young L, Yu D, Bateman RM, et al. Oxidative stress and anti- inhibitor of endometrial cancer cell proliferation--implications for a
oxidant therapy: their impact in diabetes-associated erectile dys- novel treatment strategy. Gynecol Oncol 2010; 116: 92-98.
function. J Androl 2004; 25: 830-36. [131] Zakikhani M, Dowling RJ, Sonenberg N, et al. The effects of adi-
[105] Murray FT, Cameron DF, Orth JM. Gonadal dysfunction in the ponectin and metformin on prostate and colon neoplasia involve
spontaneously diabetic BB rat. Metabolism 1983; 32: 141-7. activation of AMP-activated protein kinase. Cancer Prev Res
[106] Gazzaruso C, Coppola A, Giustina A. Erectile dysfunction and (Phila) 2008; 1: 369-75.
coronary artery disease in patients with diabetes. Curr Diabetes Rev [132] Zhang T, Guo P, Zhang Y, et al. The antidiabetic drug metformin
2011; 7: 143-7. inhibits the proliferation of bladder cancer cells in vitro and in vivo.
[107] Cameron DF, Rountree J, Schultz RE, et al. Sustained hyperglyce- Int J Mol Sci 2013; 14: 24603-18.
mia results in testicular dysfunction and reduced fertility potential [133] Zakikhani M, Dowling R, Fantus IG, et al. Metformin is an AMP
in BBWOR diabetic rats. Am J Physiol 1990; 259: 881-9. kinase-dependent growth inhibitor for breast cancer cells. Cancer
[108] Hassan AA, Hassouna MM, Taketo T, et al. The effect of diabetes Res 2006; 66: 10269-73.
on sexual behavior and reproductive tract function in male rats. J [134] Zakikhani M, Blouin MJ, Piura E, et al. Metformin and rapamycin
Urol 1993; 149: 148-54. have distinct effects on the AKT pathway and proliferation in
[109] Soudamani S, Malini T, Balasubramanian K. Effects of streptozo- breast cancer cells. Breast Cancer Res Treat 2010; 123: 271-9.
tocin-diabetes and insulin replacement on the epididymis of prepu- [135] Hirsch HA, Iliopoulos D, Tsichlis PN, et al. Metformin selectively
bertal rats: histological and histomorphometric studies. Endocr Res targets cancer stem cells, and acts together with chemotherapy to
2005; 31: 81-98. block tumor growth and prolong remission. Cancer Res 2009; 69:
[110] Scarano WR, Messias AG, Oliva SU, et al. Sexual behaviour, 7507-11.
sperm quantity and quality after short-term streptozotocin-induced [136] Landman GW, Kleefstra N, van Hateren KJ, et al. Metformin asso-
hyperglycaemia in rats. Int J Androl 2006; 29: 482-8. ciated with lower cancer mortality in type 2 diabetes: ZODIAC-16.
[111] Padron RS, Dambay A, Suarez R, et al. Semen analyses in adoles- Diabetes Care 2010; 33: 322-6.
cent diabetic patients. Acta Diabetol Lat 1984; 21: 115-21. [137] Erices R, Bravo ML, Gonzalez P, et al. Metformin, at concentra-
[112] Ranganathan P, Mahran AM, Hallak J, et al. Sperm cryopreserva- tions corresponding to the treatment of diabetes, potentiates the cy-
tion for men with nonmalignant, systemic diseases: a descriptive totoxic effects of carboplatin in cultures of ovarian cancer cells.
study. J Androl 2002; 23: 71-5. Reprod Sci 2013; 20: 1433-46.
[113] Bartak V. Sperm quality in adult diabetic men. Int J Fertil 1979; 24: [138] Chan DK, Miskimins WK. Metformin and phenethyl isothiocy-
226-32. anate combined treatment in vitro is cytotoxic to ovarian cancer
[114] Bartak V, Josifko M, Horackova M. Juvenile diabetes and human cultures. J Ovarian Res 2012; 5: 19.
sperm quality. Int J Fertil 1975; 20: 30-32. [139] Singhal A, Jie L, Kumar P, et al. Metformin as adjunct antitubercu-
[115] Seethalakshmi L, Menon M, Diamond D. The effect of streptozoto- losis therapy. Sci Transl Med 2014; 6: 1-10.
cin-induced diabetes on the neuroendocrine-male reproductive tract [140] Palomba S, Orio F, Jr., Falbo A, et al. Effects of metformin and
axis of the adult rat. J Urol 1987; 138: 190-4. clomiphene citrate on ovarian vascularity in patients with polycys-
[116] Amaral S, Moreno AJ, Santos MS, et al. Effects of hyperglycemia tic ovary syndrome. Fertil Steril 2006; 86: 1694-701.
on sperm and testicular cells of Goto-Kakizaki and streptozotocin- [141] Lord JM, Flight IH, Norman RJ. Metformin in polycystic ovary
treated rat models for diabetes. Theriogenology 2006; 66: 2056-67. syndrome: systematic review and meta-analysis. BMJ 2003; 327:
[117] Kim ST, Moley KH. Paternal effect on embryo quality in diabetic 951-3.
mice is related to poor sperm quality and associated with decreased [142] Tosca L, Froment P, Rame C, et al. Metformin decreases GnRH-
glucose transporter expression. Reproduction 2008; 136: 313-22. and activin-induced gonadotropin secretion in rat pituitary cells:
Metformin and Male Fertility Current Pharmaceutical Design, 2015, Vol. 21, No. 25 3633

potential involvement of adenosine 5' monophosphate-activated [163] Nasrolahi O, Khaneshi F, Rahmani F, et al. Honey and metformin
protein kinase (PRKA). Biol Reprod 2011; 84: 351-62. ameliorated diabetes-induced damages in testes of rat; correlation
[143] Esteghamati A, Eskandari D, Mirmiranpour H, et al. Effects of with hormonal changes. Iran J Reprod Med 2013; 11: 1013-20.
metformin on markers of oxidative stress and antioxidant reserve in [164] Fang X, Xu QY, Jia C, et al. Metformin improves epididymal
patients with newly diagnosed type 2 diabetes: a randomized clini- sperm quality and antioxidant function of the testis in diet-induced
cal trial. Clin Nutr 2013; 32: 179-85. obesity rats. Zhonghua Nan Ke Xue 2012; 18: 146-9.
[144] Martin-Montalvo A, Mercken EM, Mitchell SJ, et al. Metformin [165] Adaramoye O, Akanni O, Adesanoye O, et al. Evaluation of toxic
improves healthspan and lifespan in mice. Nat Commun 2013; 4: 1- effects of metformin hydrochloride and glibenclamide on some or-
9. gans of male rats. Niger J Physiol Sci 2012; 27: 137-44.
[145] Mirmiranpour H, Mousavizadeh M, Noshad S, et al. Comparative [166] Alves MG, Martins AD, Vaz CV, et al. Metformin and male repro-
effects of pioglitazone and metformin on oxidative stress markers duction: effects on Sertoli cell metabolism. Br J Pharmacol 2014;
in newly diagnosed type 2 diabetes patients: a randomized clinical 171: 1033-42.
trial. J Diabetes Complications 2013; 27: 501-7. [167] Nguyen TM, Alves S, Grasseau I, et al. Central role of 5'-AMP-
[146] Onken B, Driscoll M. Metformin induces a dietary restriction-like activated protein kinase in chicken sperm functions. Biol Reprod
state and the oxidative stress response to extend C. elegans Health- 2014; 91: 121.
span via AMPK, LKB1, and SKN-1. PLoS ONE 2010; 5: e8758. [168] Attia SM, Helal GK, Alhaider AA. Assessment of genomic insta-
[147] Graham GG, Punt J, Arora M, et al. Clinical pharmacokinetics of bility in normal and diabetic rats treated with metformin. Chem
metformin. Clin Pharmacokinet 2011; 50: 81-98. Biol Interact 2009; 180: 296-304.
[148] Shu Y, Sheardown SA, Brown C, et al. Effect of genetic variation [169] Rabbani SI, Devi K, Khanam S. Role of pioglitazone with met-
in the organic cation transporter 1 (OCT1) on metformin action. J formin or glimepiride on oxidative stress-induced nuclear damage
Clin Invest 2007; 117: 1422-31. and reproductive toxicity in diabetic rats. Malays J Med Sci 2010;
[149] Wilcock C, Bailey CJ. Accumulation of metformin by tissues of the 17: 3-11.
normal and diabetic mouse. Xenobiotica 1994; 24: 49-57. [170] Naglaa ZHE, Hesham AM, Hosny Abdel F, et al. Impact of met-
[150] El-Mir MY, Nogueira V, Fontaine E, et al. Dimethylbiguanide formin on immunity and male fertility in rabbits with alloxan-
inhibits cell respiration via an indirect effect targeted on the respi- induced diabetes. J Am Sci 2010; 6(Suppl 11): 1-10.
ratory chain complex I. J Biol Chem 2000; 275: 223-8. [171] Hurtado de Llera A, Martin-Hidalgo D, Gil MC, et al. AMP-
[151] Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts activated kinase AMPK is expressed in boar sperm and regulates
its anti-diabetic effects through inhibition of complex 1 of the mi- motility. PLoS ONE 2012; 7: 1-10.
tochondrial respiratory chain. Biochem J 2000; 348(Pt 3): 607-14. [172] Long YC, Zierath JR. AMP-activated protein kinase signaling in
[152] Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein metabolic regulation. J Clin Invest 2006; 116: 1776-83.
kinase in mechanism of metformin action. J Clin Invest 2001; 108: [173] Hardie DG, Hawley SA, Scott JW. AMP-activated protein kinase--
1167-74. development of the energy sensor concept. J Physiol 2006; 574: 7-
[153] Stephenne X, Foretz M, Taleux N, et al. Metformin activates AMP- 15.
activated protein kinase in primary human hepatocytes by decreas- [174] Carling D. The AMP-activated protein kinase cascade--a unifying
ing cellular energy status. Diabetologia 2011; 54: 3101-10. system for energy control. Trends Biochem Sci 2004; 29: 18-24.
[154] Li Y, Xu S, Mihaylova MM, et al. AMPK phosphorylates and [175] Carling D, Sanders MJ, Woods A. The regulation of AMP-
inhibits SREBP activity to attenuate hepatic steatosis and athero- activated protein kinase by upstream kinases. Int J Obes 2008;
sclerosis in diet-induced insulin-resistant mice. Cell Metab 2011; 32(Suppl 4): 55-59.
13: 376-88. [176] Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy
[155] McGarry JD, Brown NF. The mitochondrial carnitine palmitoyl- sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol
transferase system. From concept to molecular analysis. Eur J Bio- 2012; 13: 251-62.
chem 1997; 244: 1-14. [177] Carling D. AMP-activated protein kinase. Curr Biol 2004; 14: 220.
[156] Bertoldo MJ, Guibert E, Tartarin P, et al. Effect of metformin on [178] Hardie DG, Hawley SA. AMP-activated protein kinase: the energy
the fertilizing ability of mouse spermatozoa. Cryobiology 2014; 68: charge hypothesis revisited. Bioessays 2001; 23: 1112-9.
262-8. [179] Kahn BB, Alquier T, Carling D, et al. AMP-activated protein
[157] Kayampilly PP, Menon KM. AMPK activation by dihydrotestos- kinase: ancient energy gauge provides clues to modern understand-
terone reduces FSH-stimulated cell proliferation in rat granulosa ing of metabolism. Cell Metab 2005; 1: 15-25.
cells by inhibiting ERK signaling pathway. Endocrinology 2012; [180] Cheung PC, Salt IP, Davies SP, et al. Characterization of AMP-
153: 2831-8. activated protein kinase gamma-subunit isoforms and their role in
[158] Skrha J, Prazny M, Hilgertova J, et al. Oxidative stress and endo- AMP binding. Biochem J 2000; 346(Pt 3): 659-69.
thelium influenced by metformin in type 2 diabetes mellitus. Eur J [181] Tartarin P, Guibert E, Toure A, et al. Inactivation of AMPKalpha1
Clin Pharmacol 2007; 63: 1107-14. induces asthenozoospermia and alters sperm morphology. Endocri-
[159] Weber JE, Russell LD, Wong V, et al. Three-dimensional recon- nology 2012; 153(Suppl 7): 3468-81.
struction of a rat stage V Sertoli cell: II. Morphometry of Sertoli-- [182] Towler MC, Fogarty S, Hawley SA, et al. A novel short splice
Sertoli and Sertoli--germ-cell relationships. Am J Anat 1983; 167: variant of the tumour suppressor LKB1 is required for spermio-
163-79. genesis. Biochem J 2008; 416: 1-14.
[160] Cheng CY, Wong EW, Yan HH, et al. Regulation of spermato- [183] Tosca L, Chabrolle C, Dupont J. [AMPK: a link between metabo-
genesis in the microenvironment of the seminiferous epithelium: lism and reproduction?]. Med Sci (Paris) 2008; 24: 297-300.
new insights and advances. Mol Cell Endocrinol 2010; 315: 49-56. [184] Hurtado de Llera A, Martin-Hidalgo D, Rodriguez-Gil JE, et al.
[161] Foretz M, Hebrard S, Leclerc J, et al. Metformin inhibits hepatic AMP-activated kinase, AMPK, is involved in the maintenance of
gluconeogenesis in mice independently of the LKB1/AMPK path- plasma membrane organization in boar spermatozoa. Biochim Bio-
way via a decrease in hepatic energy state. J Clin Invest 2010; 120: phys Acta 2013; 1828: 2143-51.
2355-69. [185] Mukai C, Okuno M. Glycolysis plays a major role for adenosine
[162] Tartarin P, Moison D, Guibert E, et al. Metformin exposure affects triphosphate supplementation in mouse sperm flagellar movement.
human and mouse fetal testicular cells. Hum Reprod 2012; 27: Biol Reprod 2004; 71: 540-7.
3304-14. [186] Takei GL, Miyashiro D, Mukai C, et al. Glycolysis plays an impor-
tant role in energy transfer from the base to the distal end of the
flagellum in mouse sperm. J Exp Biol 2014; 217: 1876-86.

Received: April 23, 2015 Accepted: July 8, 2015

View publication stats

You might also like