SciELO - Scientific Electronic Library Online

 
vol.3 número2Actividad ovicida de extractos de cuatro especies de plantas contra el nematodo gastrointestinal Cooperia punctata índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • No hay artículos similaresSimilares en SciELO

Compartir


Veterinaria México OA

versión On-line ISSN 2448-6760

Veterinaria México OA vol.3 no.2 Ciudad de México abr./jun. 2016

https://doi.org/10.21753/vmoa.3.2.364 

Original Research Articles

A single neonatal administration of soybean oil and/or tamoxifen permanently affects testis histomorphology in adult rats

Alicia González-González1 

Everardo González-Padilla1 

Francisco Fierro2 

Ma. De Lourdes Juarez-Mosqueda3 

Juan José Perez-Rivero4 

Clara Ortega-Camarillo5 
http://orcid.org/0000-0001-8709-6727

Mónica Salas-Rojas6 
http://orcid.org/0000-0001-7046-0967

Marcela Vergara Onofre4  * 

1Departamento de Reproducción Facultad de Medicina Veterinaria y Zootecnia, UNAM Ciudad Universitaria. Del. Coyoacán CP. 04510 Ciudad de México.

2Departamento de Biotecnología Universidad Autónoma Metropolitana-Unidad Iztapalapa. Av. San Rafael Atlixco 186 Col. Vicentina. 19340 Del. Iztapalapa Ciudad de México.

3Departamento de Morfología Facultad de Medicina Veterinaria y Zootecnia, UNAM. Ciudad Universitaria Del. Coyoacán CP. 04510 Ciudad de México.

4Departamento de Producción Agrícola y Animal Universidad Autónoma Metropolitana-Unidad Xochimilco. Calzada del Hueso 1100 Col. Villa quietud CP. 04960 Del. Xochimilco Ciudad de México.

5Unidad de Investigación Médica en Bioquímica Hospital de Especialidades Centro Médico Nacional Siglo XXI Instituto Mexicano del Seguro Social Av. Cuauhtémoc 330 Col. Doctores CP. 06720 Del Cuauhtémoc. Ciudad de México.

6Unidad de Investigación Médica en Inmunología Hospital de Pediatría Centro Médico Nacional Siglo XXI Instituto Mexicano del Seguro Social México. Av. Cuauhtémoc 330 Col. Doctores CP. 06720 Del Cuauhtémoc. Ciudad de México.


Abstract

The aim of this study was to evaluate the effects of administration of tamoxifen (Tx) and its vehicle, soybean oil (SO), during the critical period of hypothalamic sexual differentiation in newborn male rats in the context of gonadal histomorphology during adulthood. The animals were randomly divided into 3 groups (n = 5 each). One hour after birth, one group was treated subcutaneously with 200 μg of Tx using commercial SO (20 µL) as a vehicle, another group was treated with only 20 μL of SO, and the control group received no treatment. All rats were weighed and sacrificed by cervical dislocation on day 90 post-treatment. Testicles were removed, weighed and processed for histological evaluation. The single administration of Tx and/or SO during the critical period of sexual differentiation of the hypothalamus permanently altered testicular histomorphology, spermatogenesis, and body weight in adulthood. Alterations included vacuolization and reduction in the number of spermatogonia and Sertoli cells. The administration of Tx reduced testicular weight, the diameter and area of the seminiferous tubules, and the height of the germinal epithelium and increased the intertubular space. Soybean oil by itself reduced the number of spermatocytes and spermatids to a greater extent than Tx. There was no effect on the number of Leydig cells. The possibility that soybean oil can act as an endocrine disruptor deserves greater attention and opens the possibility for the development of new pest control methods.

Keywords: Endocrine disruptors; Tamoxifen; Soybean oil; Sexual differentiation of the hypothalamus; Testicular morphology

Resumen

El objetivo de este estudio fue evaluar el efecto de la administración de tamoxifeno (Tx) y aceite de soya (SO) durante el periodo crítico de la diferenciación sexual hipotalámica en ratas macho recién nacidas, en el contexto de la histomorfología gonadal durante la edad adulta. Los animales se dividieron al azar en tres grupos (n = 5 cada uno). Una hora después del nacimiento, un grupo fue tratado por vía subcutánea con 200 μg de Tx y 20 μL de SO comercial como vehículo; mientras otro grupo fue tratado solo con 20 μL de SO, y el grupo control no recibió ningún tratamiento. Todas las ratas se pesaron y sacrificaron por dislocación cervical a los 90 días postratamiento. Se extrajeron los testículos, se pesaron y procesaron para la evaluación histológica. La administración única de Tx y de SO durante el periodo crítico de la diferenciación sexual del hipotálamo altera de forma permanente la histomorfología testicular, la espermatogénesis y el peso corporal en la edad adulta. Las alteraciones incluyen: vacuolización, reducción del número de espermatogonias y de células de Sertoli. La administración de Tx redujo el peso testicular, el diámetro y el área de los túbulos seminíferos, así como la altura del epitelio germinal, e incrementó el espacio intertubular. El SO redujo el número de espermatocitos y espermátides, incluso más que el Tx. No se afectó el número de células de Leydig. La posibilidad de que el aceite de soya actúe como disruptor endócrino, amerita más atención y abre la posibilidad para el desarrollo de nuevos métodos de control de plagas.

Palabras clave: disruptores endócrinos; tamoxifeno; aceite de soya; diferenciación sexual del hipotálamo; morfología testicular

Introduction

During development, in the “critical period” of brain sexual differentiation, the brains of male rats are exposed to higher levels of testosterone of testicular origin, which increases at the end of gestation, primarily two hours after birth (Anne et al., 2011). In neurons, testosterone is converted to estrogens by the P450 aromatase enzyme. Estrogens may act in two different stages: an “organization” phase during the prenatal and early postnatal periods, and an “activation” phase in adulthood (Phoenix et al., 1959). The organizational effects of estradiol induce sexual differentiation of the hypothalamus (Lauber et al., 1997) and actively regulate the neural circuits (Barraclough, 1966; Simerly et al., 2002), which are responsible for the tonic secretion of gonadotropins (Finkelstein et al., 1991; Bagatell et al., 1994) and reproductive behaviors characteristic of males (Lauber et al., 1997 and Simerly, 2002). Estrogens are also involved in spermatogenesis and sperm maturation in the epididymis (Balasinor et al., 2001).

Exposure to exogenous endocrine-disrupting chemicals (EDCs, of natural or synthetic origin) that mimic or block the effects of endogenous sex steroid hormones (Wilson and Davies, 2007) can send mixed messages to the body, causing developmental abnormalities with irreversible effects. The role of estrogens in sexual reproduction has been confirmed by the use of antiestrogens such as tamoxifen (Tx; Z-1-[4-(2-dimethylamino-ethoxy)-phenyl]-1, 2-diphenyl-1-butane) (Barraclough, 1967; Dörner and Staudt, 1968; Döhler et al., 1984), a synthetic drug commonly used in breast cancer treatment. Its mechanism of action remains unclear, but it is known that it exerts several biological effects. It may act as a complete estrogen agonist or antagonist depending on the dose, target organ, sex, and species (Gill-Sharma et al., 1993; Furr and Jordan, 1984; Macnab et al., 1984). In addition to Tx, there are other chemicals that can alter the hormonal milieu, affecting growth, sexual development and behavior.

In male mice, Tx is detrimental for testicular and accessory gland development after neonatal administration (24 hours post-birth; 20 or 100 µg/d for 5 days; Iguchi and Hirokawa, 1986) and renders male mice infertile (Taguchi, 1987). Tx impedes the interaction of estradiol with its receptor in the hypothalamus, in addition to inhibiting the aromatase enzyme, leading to cerebral feminization (Döhler et al., 1985). In adults, a reversible effect of Tx on the hypothalamus-hypophysis axis has been reported (Gill-Sharma et al., 1993); Balasinor et al. (2002) observed a dose-dependent reduction in fertility due to embryo loss in females mated to male rats treated with Tx.

In our group, Herrera et al. (2013) found that a single subcutaneous (sc) dose of Tx (200 µg) in rats one hour after birth modified the expression of certain hypothalamic genes. To advance on those studies, an experiment was designed to examine the expression of 11 specific genes in the hypothalamus of male rats within the first six hours after treatment at one hour after birth with Tx in SO, SO or control. Some of the animals were weighed at weekly intervals and sacrificed as adults at 90 d of age, when both testicles were removed, weighed and subjected to histological analysis. Observations of the changes in testicular morphology in those animals are presented in this communication.

Materials and methods

Animals

The animals were obtained from and housed in the Production and Animal Experimentation Laboratory Unit (UPEAL) of the Universidad Autónoma Metropolitana, campus Xochimilco (UAMX), in a controlled environment (21°C) with 12-hour light-dark inverted cycles (lights off at 4:00 AM) as well as food (Rodent Diet 5001, LabDiet) and water ad libitum.

Nine Wistar rats (6 females and 3 males) were used as breeders. The males were 90 day-old rats that weighed more than 400 g. Their reproductive capacity was evaluated based on their copulatory behavior, the presence of a seminal plug in females after copulation, and pregnancy confirmation. The females were 60-day-old rats that weighed more than 200 g. Their estrous cycle was monitored daily through vaginal cytology for at least 3 cycles to confirm their endocrine-reproductive normality. Estrous rats were placed in a cage with a male to obtain a first pregnancy and to corroborate their reproductive capacity. After the first litter, a second round of selection was conducted based on the observed maternal behavior. Finally, only rats that had estrous cycles, gestation, and normal litters, as well as adequate maternal behavior, were selected.

Two weeks after the first delivery, new estrous cycles were monitored again. At estrous, one female and one male were placed in a cage again for 3 hours. The beginning of pregnancy was verified by the presence of sperm through vaginal cytology and/or the formation of a seminal plug. Pregnant rats were housed in cages under the conditions previously described. Because our main goal was to analyze the effects of early administration (1 hour after birth) of Tx and its vehicle (SO) on testicular microscopic morphology in adult rats, strict control of the moment of fertilization and constant monitoring of mothers were necessary, primarily at the end of gestation, to identify the exact time of birth.

The time of birth of each animal was considered hour zero. Newborn males were separated and divided randomly into 3 groups of 5 animals each. An hour after birth, treatment was administered: one group was treated subcutaneously with 200 μg of Tx (Sigma-Aldrich- Sigma-Aldrich Corporate Offices, 3050 Spruce St. St. Louis, MO 63103, U.S.A.) diluted in commercial SO (20 μL), another group was treated subcutaneously only with SO (20 μL), and the control group received no treatment. Tamoxifen was used to induce brain feminization. Soybean oil alone was given to avoid confounding the effects of treatment due to the vehicle. Treated rats were returned to their mothers until weaning (21 days) and then fed in the same manner as the other animals.

Collection and processing of samples

All rats were weighed weekly until 90 days post-treatment and sacrificed by cervical dislocation. Immediately, testicles were removed in a block, dissected under a stereoscopic microscope, weighed on an analytical balance, perfused and fixed in Bouin solution for 8 hours. Subsequently, they were dehydrated in a decreasing ethanol series, cleared in xylene, and embedded in paraffin to obtain cross-section slides (5 microns) on a rotary microtome (Leica RM2125 RT). Finally, hematoxylin-eosin staining was performed for morphological evaluation, and the testicle cross-sections were observed under a microscope.

Morphological evaluation

Only round tubules were digitized for analysis in 20X and 40X magnification fields. Ten tubules were analyzed for each testicle (20 tubules for each rat). Measurements of diameter, area, height of the germinal epithelium, and interstitial space were made from the scanned sections with the LSM5 software (Carl Zeiss, Germany) using the marking tools. The photographed cross-sections were used to identify the tubular stages and to count cells using the tubular morphology method (Berndtson, 1977; Hess and França, 2007), which is based on the shape of the spermatid nuclei, the occurrence of meiotic divisions, and the position of spermatids in the seminiferous epithelium (Farias et al., 2014). All slides were analyzed by 3 veterinary morphologists, and the individual results were averaged for this report.

Statistical analysis

A Shapiro Wilks test was performed to determine the normality of the data for the different parameters studied. Depending on the normality of distribution, data were analyzed with a Kruskal-Wallis test (morphology and cell counts) or one-way ANOVA (body weight and testicular weight). The results are expressed as the mean values ± SE (standard error) and mean values ± SD (standard deviation), respectively. Values analyzed were derived from 5 animals per group for body weight and testicular weight and from 10 tubules randomly taken from each testicle. PAST software (Paleontological Statistics Package for Education and Data Analysis, Huang et al., 2013) was used for all statistical calculations.

Results and discussion

The results for testicular weight and body weight at 90 days post-treatment are shown in Table 1. The average testicular weight was significantly lower in animals treated with Tx than in the control group or the SO-treated group (F: 47.19; degrees of freedom between groups: 2; degrees of freedom within groups: 12; P < 0.001). The average body weight at 90 days was lowest for the animals treated with Tx, followed by the group treated with SO, whereas the animals in the control group were the heaviest (F: 16.67; degrees of freedom between groups: 2; degrees of freedom within groups: 12; P < 0.001). Only the animals treated with Tx showed a significant reduction in tube diameter (H: 12.88; Hc: 13.00; P < 0.005), tubular area (H: 9.85; Hc 9.85; P < 0.05), and height of the germinal epithelium (H: 27.04; Hc: 27.05; P < 0.001) [Table 2]. In comparison to the control group, the intertubular space was largest in the animals treated with Tx, followed by those treated with SO.

Table 1: Mean ± SD for testicular weight and body weight at 90 days. 

Testicular weights and body weights of 90-day-old rats. Distinct superscripts within each line indicate statistically significant differences (P < 0.05).

Table 2: Testicle morphometry of 90-day-old male rats treated with tamoxifen and/or soybean oil 1 h post-birth (mean ± SE). 

Testicular alterations induced by Tx and/or SO treatments. Distinct superscripts within each line indicate statistically significant differences (P < 0.05).

The average results for cell count per tubule are shown in Table 3. The numbers of spermatogonia (H: 6,953; Hc: 6,959; P < 0.05) and Sertoli cells (H: 9.89; Hc: 10.31; P < 0.05) were significantly lower in the Tx and SO experimental groups in comparison to the control group. No significant differences in the number of Leydig cells were observed (H: 1.33; Hc: 1.33; P < 0.5). Compared to the control group, the males treated with SO showed the greatest reduction in the number of spermatocytes (H: 12.21; Hc: 12.22 P < 0.05) and elongated spermatids (H: 11.62; Hc: 11.64; P <0.05), followed by the Tx-treated males.

Table 3: Testicular cell counts of 90-day-old male rats treated with tamoxifen and/or soybean oil 1 h post-birth (mean ± SE). 

Testicular alterations induced by Tx and/or SO treatments. Distinct superscripts within each line indicate statistically significant differences (P < 0.05).

As shown in Figure 1, testicles in the control group (A-B) showed evidence of seminiferous tubules with normal spermatogenesis. The SO-treated group (C-D) showed vacuolization in the seminiferous tubules, destruction of the germinal epithelium, and a reduction in the amount of germ and Sertoli cells; additionally, increases in the intertubular spaces and ungrouped Leydig cells were observed. These alterations were also noted in animals treated with Tx, which in addition showed a significant reduction in tubular diameter and tubular area as well as a larger intertubular space than either the control or SO groups.

Figure 1: Representative hematoxylin-eosin-stained histological sections of testicles collected from the control group (A y B), rats treated with SO (C y D) and rats treated with Tx (E y F). A, C and E, 200X. B, D and F, 400X. T: Tubule. LC: Leydig cells. Histological changes induced by Tx and/or SO treatments can be observed. Black arrows point to the largest intertubular space. White arrows in E and F indicate the ungrouping of the LC. Asterisks indicate the vacuoles in the seminiferous tubules. 

The results revealed that during the critical period of sexual differentiation of the hypothalamus, a single administration of Tx and SO reduced body weight, testicular weight, and altered the gonadal histomorphology and spermatogenesis in adult rats. In our experiment, although no measurements of circulating hormone concentrations were performed, the effect on weight gain over 90 days in treated animals is an indication of insufficient testosterone stimulation, despite the lack of effects on the number of Leydig cells due to treatment. It must be noted that the edematous environment and the dispersion of cells in the intertubular space in treated animals, as well as the possible lack of proper gonadotropic stimulation, could reduce testosterone production. The disruption of normal gonadotropin secretion (and therefore testosterone) due to inadequate hypothalamic control caused by the treatments could explain the low numbers of Sertoli cells, which constitute the structural basis for the blood-testis barrier (Yu-hua et al., 2015) and provide support for spermatogenesis. The diminished presence of FSH and testosterone in the seminiferous tubules could have affected Sertoli cell function and estrogen production, which is congruous with the observed reduction in germ cell numbers, most likely caused by increased apoptosis in an environment with vacuolization and tubular edema.

The mammalian testis is characterized by the synthesis of steroid hormones and sperm production (Carreau et al., 2006). Normal testicular development and spermatogenesis maintenance are primarily controlled by gonadotropins and testosterone (Carreau et al., 1999). Their effects are modulated by local factors, among which estrogens are essential for the maintenance of the reproductive tract in males (Carreau et al., 2003). Estrogens are synthesized in Leydig cells (Abney, 1999), Sertoli cells, and round and elongated spermatids (Hess et al., 2001; Saunders et al., 2005; Lambard et al., 2005). In these cells, the existence of specific estrogen alpha (ERα) and beta (ERβ) receptors suggests paracrine and autocrine effects of estradiol (Carreau et al., 2002; Scobie et al., 2002). Estrogens act as survival factors in germ cells (Carreau et al., 2003). It has been demonstrated that the P450 aromatase enzyme is required for the maturation of sperm and fluid absorption mechanisms in the efferent ducts (Lambard et al., 2005). In the absence of estrogen receptors, fluid absorption in these ducts, in the rete testis and in the seminiferous tubules is reduced (Hess et al., 2000) and accumulates in the testicles, causing damage to the germinal epithelium and therefore causing alterations in sperm development and fertility (Hess et al., 2001), as was observed in our work. This suggests that the treatments influenced the mechanisms involved to provide the estrogenic action necessary for normal testicular function.

Under physiological conditions, estrogens play an important role in the normal development of the structure and function of the testicles and the male reproductive tract. In the testicle, estrogens depend on the availability of androgens for their synthesis; however, under experimental conditions, the neonatal administration of exogenous estrogenic substances orally, in feed or water, reduces sperm counts, plasma testosterone and the number of Sertoli cells (Goyal et al., 2003; Sharpe et al., 2003 and Atanossova et al., 2005). In post-pubertal rats, Assinder et al. (2007) reported that weaned male rats fed for 24 days on a diet high in phytoestrogens demonstrated increased apoptosis of germ cells, a reduction in spermatic counts and a lumenal increase in the seminiferous tubules, without apparent involvement of the hypothalamus-hypophysis-testicle axis, but rather involving the paracrine and autocrine effects of estrogens in the testicle.

Soy and its derivatives are known sources of isoflavones, a particular class of phytoestrogens that may interact in the signaling pathways of endogenous estrogen (Cederroth et al., 2012). Previous studies have shown that during critical periods of development, exposure to phytoestrogen induces adverse morphological and physiological effects on male sexual differentiation in mice (Roberts et al., 2000; Wisniewski et al., 2003). Levy et al. (1995) reported that treatment with genistein (derived from soy) reduces body weight (in males and females) and the anus-genital distance at birth (in males) and delays the onset of puberty. These results were confirmed by Wisniewski et al. (2005), who also observed phenotypic and behavioral disorders in male mice of mothers fed with 5 or 300 mg of genistein/kg of body weight during pregnancy and lactation. At 21 days after birth, the offspring exhibited permanent demasculinization: a reduction in sperm cells, testosterone and aggressive behavior and an increase in defensive behavior. In our study, the observation of vacuolation, decreased germinal epithelium height, and increased intertubular space constituted important findings. Other researchers, such as Perez-Rivero et al. (2009 and 2014), have previously reported these changes in dogs and vampire bats fed with rich phytoestrogen (coumestrol) diets. These observations are compatible with those in our experiment but are associated with a single sc injection of SO.

Tamoxifen administration during the perinatal stage blocks the interaction of estradiol with its receptor in the hypothalamus and inhibits the aromatase enzyme (Döhler et al., 1985), which is necessary for the masculinization of the neural circuits that modulate the behavior and the gonadotropin secretion typical of males in adulthood. Tx induces oxidative stress and apoptosis in albino mouse spermatozoa, morphological abnormalities (DNA adduct formation; Padmalatha and Vijayalaxmi, 2001) and seminiferous tubule distortion with germ cell detachment. In adult male rats, Tx (200, 400 or 800 mg/kg of body weight) caused multinucleated giant cells (D’Souza, 2003) and reduced fertility (Balasinor et al., 2001; Gill-Sharma, 1993).

It is known that Tx may act as an estrogen antagonist or agonist on the reproductive axis of the rat (Bellido et al., 2003), and SO can be a source of phytoestrogens. In this study, both substances were administered in the first hour after the birth of male rat pups, which is close to the critical time for hypothalamus sex differentiation; thus, the morphological changes observed in the testicles could be attributable to the effects of the treatments on hypothalamic nuclei controlling tonic gonadotropin secretion, with an additional effect on testicular steroidogenesis. The role of testosterone in testicular function and spermatogenesis is well documented. However, it is worth mentioning that in models of soy phytoestrogen administration in adult rodents, the disruption of testicular function can be independent of the circulating concentrations of gonadotrophins and testosterone.

Conclusions

This communication documents the irreversible changes on the testicular function and histomorphology of adult rats caused by a single, acute treatment of tamoxifen and/or soybean oil at one hour after birth. These changes may be associated with disruption of the normal course of sex differentiation of the hypothalamus during the known critical period for this process.

Funding

This project was partially financed by CONACYT grant 105961-M, ANUIES-ECOS-CONACYT grant M10-A02, and CONACYT scholarship 201677.

Referencias

1 ) Abney TO. 1999. The potential roles of estrogens in regulating Leydig cell development and function: A review. Steroids, 64:610-617. DOI: 10.1016/S0039-128X(99)00041-0. [ Links ]

2 ) Anne T, Konkle AT, M, Margaret M. McCarthy M.M. 2011. Developmental time course of estradiol, testosterone, and dihydrotestosterone levels in discrete regions of male and female rat brain. Endocrinology, 152:223-235. DOI: 10.1210/en.2010-0607. [ Links ]

3 ) Assinder S, Davis R, Fenwick M, Glover A. 2007. Adult-only exposure of male rats to a diet of high phytoestrogens content increase apoptosis of meiotic and post-meiotic germ cells. Reproduction, 133:11-19. DOI: 10.1530/rep.1.01211. [ Links ]

4 ) Atanassova NN, Walker M, McKinnell C, Fisher JS, Sharpe RM. 2005. Evidence that androgens and oestrogens, as well as folliclestimulating hormone, can alter Sertoli cell number in the neonatal rat. Journal of Endocrinology, 184:107-117. DOI: 10.1677/joe.1.05884. [ Links ]

5 ) Bagatell CJ, Dahl KD, Bremmer WJ. 1994. The direct pituitary effect of testosterone to inhibit gonadotropin secretion in men is partially mediated by aromatization to estradiol. Journal of Andrology, 15:15-21. DOI: 10.1002/j.1939-4640.1994.tb01674.x. [ Links ]

6 ) Balasinor N, Gill-Sharma MK, Parte P, D´Souza S, Kedia N, Juneja HS. 2001. Effect of paternal administration of an antiestrogen, tamoxifen on embryo development in rats. Molecular and Cellular Endocrinology, 190:159-66. DOI:10.1016/S0303-7207(01)00723-7. [ Links ]

7 ) Balasinor N, Gill-Sharma MK, Parte P, D´Souza S, Kedia N, et al., 2002. Effect of paternal administration of an antiestrogen, tamoxifen on embryo development in rats. Molecular and Cellular Endocrinology, 190:159-66. DOI:10.1016/S0303-7207(01)00723-7. http://dx.doi.org/10.1016/S0303-7207(01)00723-7. [ Links ]

8 ) Barraclough A. 1967. Modifications in reproductive function after exposure to hormones during the prenatal and early posnatal period. In: Martini L, Ganong WF (eds). Neuroendocrinology. United States of America, New York and London Academic Press. DOI: 10.1016/B978-1-4832-3229-4.50010-5. [ Links ]

9 ) Barraclough CA. 1966. Modifications in the CNS regulation of reproduction after exposure of prepubertal rats to steroid hormones. Recent Progress in Hormone Research Journal, 22:503-539. DOI: 10.1016/B978-1-4831-9825-5.50016-6. [ Links ]

10 ) Bellido C, Martín de las Mulas J, Tena-Sempere M, Aguilar R, Alonso R, Sánchez-Criado JE. 2003. Tamoxifen induces gonadotropin-releasing hormone self-priming through an estrogen-dependent progesterone receptor expression in the gonadotrope of the rat. Neuroendocrinology , 77(6):425-435. DOI: 10.1159/000071314. [ Links ]

11 ) Berndston WE. 1977. Methods for quantifying mammalian spermatogenesis: A review. Journal of Animal Science, 44:818-833. DOI: 10.2134/jas1977.445818x. [ Links ]

12 ) Carreau S, Bourguiba S, Lambard S, Galeraud DI, Genissel C, Levallet J. 2002. Reproductive system: Aromatase and estrogens. Molecular and Cellular Endocrinology, 193:137-143. DOI: 10.1016/S0303-7207(02)00107-7. [ Links ]

13 ) Carreau S, Delalande C, Silandre D, Bourguiba S, Lambard S. 2006. Aromatase and estrogen receptors in male reproduction. Molecular and Cellular Endocrinology, 246:65-68. DOI: 10.1016/j.mce.2005.11.021. [ Links ]

14 ) Carreau S, Genissel C, Bilinska B, Levallet J. 1999. Sources of oestrogen in the testis and the reproductive tract of the male. International Journal of Andrology, 22:211-213. DOI: 10.1046/j.1365-2605.1999.00172.x. [ Links ]

15 ) Carreau S, Lambard S, Delalande C, Denis GI, Bilinska B, Bourguiva S. 2003. Aromatase expression and role of estrogens in male gonad: A review. Reproductive Biology and Endocrinology, 35:1-6. DOI: 10.1186/1477-7827-1-35. [ Links ]

16 ) Cederroth CR, Zimmermann C, Nef SS. 2012. Soy, phytoestrogens and their impact on reproductive health. Molecular and Cellular Endocrinology, 355:192- 200. DOI: 10.1016/j.mce.2011.05.049. [ Links ]

17 ) Döhler K, Srivastava S, Shryne J, Jarzab B, Sipos A, Gorski R. 1984. Differentiation of the sexually dimorphic nucleus in the preoptic area of the rat brain is inhibited by posnatal treatment with an estrogen antagonist. Neuroendocrinology , 38:297-301. DOI: 10.1159/000123907. [ Links ]

18 ) Döhler KD, Hines M, Coquelin A, Davis F, Shryne JE, Sickmöler PM, et al. 1985. Pre- and posnatal influence of an estrogen antagonist and an androgen antagonist on differentiation of the preoptic area in male and female rats. Neuroendocrinology , 42:443-448. DOI: 10.1159/000124484. [ Links ]

19 ) Dörner G, Staudt J. 1968. Structural changes in the preoptic anterior hypothalamic area of the male rat. Following neonatal castration and androgen treatment. Neuroendocrinology , 3:136-140. DOI: 10.1159/000121703. [ Links ]

20 ) D’ Souza UJA. 2003. Tamoxifen induced multinucleated cells (symplasts) and distortion of seminiferous tubules in rat testis. Asian Journal of Andrology, 5(3):217-220 Asian Journal of Andrology, 5(3):217-220 http://www.asiaandro.com/archive/1008-682x/5/217.htm [access: 15 jun 2016]. [ Links ]

21 ) Farias TO, Notini AA, Talamoni SA, Godinho HP. 2014. Testis morphometry and stages of the seminiferous epithelium cycle in an epididymal sperm-storing neotropical vespertilionid, Myotis levis (Chiroptera). Journal of Veterinary Medicine, 44:361-369. DOI: 10.1111/ahe.12148. [ Links ]

22 ) Finkelstein JS, O’Dea LSL, Whitcomb RW, Crowley WF. 1991. Sex steroid control of gonadotropin secretion in the human male. II Effects of estradiol administered in normal and gonadotropin-releasing hormone-deficient men. Journal of Clinical Endocrinology and Metabolism, 70:621-628. DOI: 10.1210/jcem-73-3-621. [ Links ]

23 ) Furr BJA, Jordan VC. 1984. The pharmacology and clinical uses of tamoxifen. Pharmacology and Therapeutics, 25:127-205. DOI: 10.1016/0163-7258(84)90043-3. [ Links ]

24 ) Gill-Sharma MK, Gopalkrishnan K, Balasinor N, Parte P, Jayaraman S, Juneja HS. 1993. Effects of tamoxifen on the fertility of male rats. Journal of Reproduction and Fertility, 99:395-402. DOI: 10.1530/jrf.0.0990395. [ Links ]

25 ) Goyal HO, Robateau A, Braden TD, Williams CS, Srivastava KK, Ali K. 2003. Neonatal estrogen exposure of male rats alters reproductive functions at adulthood. Biology of Reproduction, 68:2081-2091. DOI: 10.1095/biolreprod.102.010637. [ Links ]

26 ) Herrera GH, Rosado GA, Vergara OM, Salcedo VM, Miliar GA, Heuze de IY et al. 2013. Genetic expression associated to cell cycle, apoptosis, synaptogenesis and cell differentiation during sex differentiation in rats. Revista Mexicana de Ciencias Pecuarias, 4(3):289-304. http://www.scielo.org.mx/pdf/rmcp/v4n3/v4n3a3.pdf [access: 15 jun 2016]. [ Links ]

27 ) Hess RA, Bunick D, Bahr J. 2001. Oestrogen its receptor and function in male reproductive tract- a review. Molecular and Cellular Endocrinology, 178:29-38. DOI: 10.1016/S0303-7207(01)00412-9. [ Links ]

28 ) Hess RA, Bunick D, Lubahn BD, Zhou Q, Bouma J. 2000. Morphologic changes in efferent ductules and epididymis in estrogen receptor-a knockout mice. Journal of Andrology , 21(1):107-121. DOI:10.1002/j.1939-4640.2000.tb03282.x. http://dx.doi.org/10.1002/j.1939-4640.2000.tb03282.x. [ Links ]

29 ) Hess, RA., Renato de França. 2008. Spermatogenesis and cycle of the seminiferous epithelium. Advances in Experimental Medicine and Biology, 636:1-15. DOI:10.1007/978-0-387-09597-4_1. http://dx.doi.org/10.1007/978- 0-387-09597-4_1. [ Links ]

30 ) Huang B, Harper DAT, Hammer O. 2013. Introduction to PAST, a comprehensive statistics software package for paleontological data analysis [PDF]. Acta Paleontologica Sinica, 52(2):181. http://palaeo-electronica.org/2001_1/past/past.pdf [access: 15 jun 2016]. [ Links ]

31 ) Iguchi T, Hirokawa M. 1986. Changes in the male genital organs of mice exposed neonatally to tamoxifen. Proceedings of the Japan Academy, 62:157-160. DOI: 10.2183/pjab.62.157. [ Links ]

32 ) Kosif R, Yilmaz F, Evrendilek AG, Diramali M. 2010. Histopathological effects of Aloe barbadensis and soybean oil on rat liver. International Journal of Morphology, 28(4):1101-1106. DOI: 10.4067/S0717-95022010000400019. [ Links ]

33 ) Lambard S, Silandre D, Delalande C, Galeraud DI, Bourguiba S, Carreau S. 2005. Aromatase in testis: Expression and role in male reproduction. Journal of Steroid Biochemistry & Molecular Biology, 95:63-69. DOI: 10.1016/j.jsbmb.2005.04.020. [ Links ]

34 ) Lauber ME, Sarasin A, Lichtensteiger W. 1997. Sex differences and androgen-dependent regulation of aromatase (CYP19) mRNA expression in the developing and adult rat brain. Journal of Steroid Biochemistry and Molecular Biology, 61:359-364. DOI: 10.1016/S0960-0760(97)80033-7. [ Links ]

35 ) Levy JR, Faber KA, Ayyash L, Hughes CL Jr. 1995. The effect of prenatal exposure to the phytoestrogen genistein on sexual differentiation in rats. Proceedings of the Society for Experimental Biology and Medicine, 208(1):60-66. DOI: 10.3181/00379727-208-43832. [ Links ]

36 ) Macnab MW, Tallarida RJ, Joseph R. 1984. An evaluation of tamoxifen as a partial agonist by classical receptor theory - an explanation of the dual action of tamoxifen. European Journal of Pharmacology, 103:321-326. DOI: 10.1016/0014-2999(84)90493-X. [ Links ]

37 ) Padmalatha RS, Vijayalaxmi KK. 2001. Tamoxifen citrate induced sperm shape abnormalities in the in vivo mouse. Mutation Research, 492:1-6. DOI: 10.1016/S1383-5718(00)00160-1. [ Links ]

38 ) Pérez-Rivero JJ, Pérez MM, Aguilar SA. 2014. Histometric analysis of vampire bat (Desmodus rotundus) testicles treated with coumestrol by oral route. Journal of Applied Animal Research, 42(2):208-212. DOI: 10.1080/09712119.2013.827578. [ Links ]

39 ) Perez-Rivero JJ, Martinez MJJ, Perez MM, Aguilar SA, Garcia SMD Serrano H. 2009. Phytoestrogen treatment induces testis alterations in dogs. Potential use in population control. Veterinary Research Communications, 33:87-95. DOI: 10.1007/s11259-008-9077-3. [ Links ]

40 ) Phoenix CH, Goy RW, Gerall AA, Young WC. 1959. Organizing action of prenatally administered testosterone propionate on the tissues mediating mating behavior in the female guinea pig. Endocrinology , 65(3):369-382. DOI: 10.1016/j.yhbeh.2009.01.004. [ Links ]

41 ) Roberts D, Veeramachaneni DN, Schlaff WD, Awoniyi CA. 2000. Effects of chronic dietary exposure to genistein, a phytoestrogen, during various stages of development on reproductive hormones and spermatogenesis in rats. Endocrine, 13(3):281-286. DOI: 10.1385/ENDO:13:3:281. [ Links ]

42 ) Saunders PTK. 2005. Does estrogen receptor b play a significant role in human reproduction? TRENDS in Endocrinology and Metabolism, 16(5):222-227. DOI: 10.1016/j.tem.2005.05.006. [ Links ]

43 ) Scobie GA, Macpherson S, Millar MR, Groome NP, Romana P, Saunders PTK. 2002. Human estrogen receptors: Differential expression of ERalpha and beta and the identification of ERbeta variants. Steroids , 67:985-992. DOI: 10.1016/S0039-128X(02)00047-8. [ Links ]

44 ) Sharpe RM, Rivas A, Walker M, McKinnell C, Fisher JS. 2003. Effect of neonatal treatment of rats with potent or weak (environmental) oestrogens, or with a GnRH antagonist, on Leydig cell development and function through puberty into adulthood. International Journal of Andrology, 26:26-36. DOI: 10.1046/j.1365-2605.2003.00385.x. [ Links ]

45 ) Simerly R. 2002. Wired for reproduction: Organization and development of sexually dimorphic circuits in the mammalian forebrain. Annual Review of Neuroscience, 25:507-536. DOI: 10.1146/annurev.neuro.25.112701.142745. [ Links ]

46 ) Taguchi N. 1987. Reproductive tract lesions in the male mice treated neonatally with tamoxifen. Biology of Reproduction, 37:113-116. DOI: 10.1095/biolreprod37.1.113. [ Links ]

47 ) Wilson CA, Davies DC. 2007. The control of sexual differentiation of the reproductive system and brain. Reproduction , 133:331-359. DOI: 10.1530/REP-06-0078. [ Links ]

48 ) Wisniewski AB, Cernetich A, Gearhart JP, Klein SL. 2005. Perinatal exposure to genistein alters reproductive development and aggressive behaviour in male mice. Physiology and Behavior; 84:327-334. DOI: 10.1016/j.physbeh.2004.12.008. [ Links ]

49 ) Wisniewski AB, Klein SL, Lakshmanan Y, Gearhart JP. 2003. Exposure to genistein during gestation and lactation demasculinizes the reproductive system in rats. Journal of Urology, 169(4):1582-1586. DOI: 10.1097/01. ju.0000046780.23389.e0. [ Links ]

50 ) Yu-hua L, Fei D, Fen Y, Xiao-Yu Z, Hong-jie P, Yang L et al. 2015. Pubertal exposure to bisphenol A affects the reproduction of male mice and sex ratio of offspring. Journal of Reproduction & Contraception, 26(1):14-21. DOI: 10.7669/j.issn.1001-7844.2015.01.0014. [ Links ]

Acknowledgements

The authors thank Beatriz Marquez Cruz and Pablo Sanchez Peña from the Laboratory of Pathology (General Hospital Zones 47 and 1A, respectively, Mexican Institute of Social Security) for their technical assistance. The Internal Committee for the Care and Use of Laboratory Animals of UAMX approved the procedures performed on the animals.

Received: February 10, 2016; Accepted: June 08, 2016

*Autor para correspondencia: Correo electrónico: mvergara@correo.xoc.uam.mx

The authors declare no conflicts of interest.

Alicia González-González, experimental development and analysis of results. Everardo González-Padilla, in the discussion and analysis of results. Francisco Fierro, in the analysis of results. Mónica Salas-Rojas, in the morphometric analysis. Ma. De Lourdes Juarez-Mosqueda, in sample processing. Juan José Perez-Rivero, in morphometric and statistical analysis. Clara Ortega-Camarillo and Marcela Vergara Onofre, in experimental design and supply of materials.

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License