Next Article in Journal
Characterization and Typology of Backyard Small Pig Farms in Jipijapa, Ecuador
Next Article in Special Issue
The Aromatase–Estrogen System in the Testes of Non-Mammalian Vertebrates
Previous Article in Journal
Effects of Dietary Polyphenols from Olive Mill Waste Waters on Inflammatory and Apoptotic Effectors in Rabbit Ovary
Previous Article in Special Issue
The Effect of Single and Triple Testicular Biopsy Using Biopty Gun on Spermatogenesis in Pubertal Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Proliferative and Apoptotic Pathways in the Testis of Quail Coturnix coturnix during the Seasonal Reproductive Cycle

by
Sara Falvo
1,†,
Luigi Rosati
2,†,
Maria Maddalena Di Fiore
1,
Federica Di Giacomo Russo
1,
Gabriella Chieffi Baccari
1 and
Alessandra Santillo
1,*
1
Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
2
Dipartimento di Biologia, Università degli Studi di Napoli “Federico II”, 80138 Napoli, Italy
*
Author to whom correspondence should be addressed.
First two authors equally contributed.
Animals 2021, 11(6), 1729; https://doi.org/10.3390/ani11061729
Submission received: 9 May 2021 / Revised: 27 May 2021 / Accepted: 31 May 2021 / Published: 9 June 2021
(This article belongs to the Special Issue New Insights into Animal Spermatogenesis)

Abstract

:

Simple Summary

The quail Coturnix coturnix exhibits an annual cycle of testis size, sexual steroid production, and spermatogenesis. The testicular levels of both 17β-estradiol (E2) and androgens are higher during the reproductive period compared to the non-reproductive period, suggesting that estrogens act in synergy with the androgens for the initiation of spermatogenesis. Therefore, the present study aimed to investigate the estrogen responsive system in quail testis in relation to the reproduction seasons, with a focus on the molecular pathways activated in both active and regressive quail testes. The results indicated that estrogens participated in the activation of mitotic and meiotic events during the reproductive period by activating the ERK1/2 and Akt-1 pathways. In the non-reproductive period, when the E2/ERα levels are low, ERK1/2 and Akt-1 pathways remain inactive and apoptotic events occur. Our results suggest that the activation or inhibition of these molecular pathways plays a crucial role in the physiological switch “on/off” of the testicular activity in male quail during the seasonal reproductive cycle.

Abstract

The quail Coturnix coturnix is a seasonal breeding species, with the annual reproductive cycle of its testes comprising an activation phase and a regression phase. Our previous results have proven that the testicular levels of both 17β-estradiol (E2) and androgens are higher during the reproductive period compared to the non-reproductive period, which led us to hypothesize that estrogens and androgens may act synergistically to initiate spermatogenesis. The present study was, therefore, aimed to investigate the estrogen responsive system in quail testis in relation to the reproduction seasonality, with a focus on the molecular pathways elicited in both active and regressive quail testes. Western blotting and immunohistochemistry analysis revealed that the expression of ERα, which is the predominant form of estrogen receptors in quail testis, was correlated with E2 concentration, suggesting that increased levels of E2-induced ERα could play a key role in the resumption of spermatogenesis during the reproductive period, when both PCNA and SYCP3, the mitotic and meiotic markers, respectively, were also increased. In the reproductive period we also found the activation of the ERK1/2 and Akt-1 kinase pathways and an increase in second messengers cAMP and cGMP levels. In the non-reproductive phase, when the E2/ERα levels were low, the inactivation of ERK1/2 and Akt-1 pathways favored apoptotic events due to an increase in the levels of Bax and cytochrome C, with a consequent regression of the gonad.

1. Introduction

In seasonal breeding vertebrates, steroidogenesis and spermatogenesis undergo fluctuations during the transition between the stages of the annual cycle [1,2,3]. Sex steroid hormones, such as testosterone and estrogens, are the key factors in the hormonal interplay that governs seasonal breeding [1,2,3]. Estrogens, which have historically been considered female hormones, have exhibited potential functions in the male reproductive system as well, such as in the processes of spermatogenesis and spermiogenesis [4]. The estrogen 17β-estradiol (E2) is synthesized by the irreversible conversion of testosterone in the presence of the P450 aromatase enzyme [5]. The action of E2 is mediated by two intracellular estrogen receptors (ERs), namely, ERα and ERβ [6]. The information regarding the ERs in avian testis is scarce. Earlier studies have reported the mRNA expression of testicular ERs in the embryos of chicken [7,8] and duck [9], and the adults of gander [10] and Japanese quail [11,12]. The immunoreactivity of ERs has been detected in the testes of chickens [13,14], Japanese quail [15,16], and domestic goose [17]. Meanwhile, a seasonal pattern of ER expression and E2 concentration has only been demonstrated in male domestic goose [17].
Several studies suggested that estrogen activity within the testis might be mediated by the regulation of the activity of mitogen-activated protein kinases (MAPKs) [18,19], which are known to occupy a focal point in signal transduction [20]. The most extensively studied members of the MAPK family are: (1) the extracellular signal-regulated kinases (ERK1 and ERK2), which are involved in the regulation of spermatogonia proliferation and the meiotic progression of spermatocytes [21,22,23], and (2) the serine/threonine kinase Akt-1, which mediates the growth factor-dependent cell survival in various cell types through the inactivation of several pro-apoptotic molecules [24,25]. In the testes of mammalian as well as non-mammalian seasonal breeders, cell proliferation and apoptosis occur at different time points during the annual cycle [26,27,28]. In several bird species, the transition from the breeding to non-breeding stage leads to significant testicular atrophy, which has been associated with the increased incidence of cell death through apoptosis and the decreased rate of cell proliferation [29,30,31,32]. However, the intracellular signal transduction mechanisms underlying these changes occurring in the avian testicular activity during the reproductive cycle remain unknown so far. In this regard, the quail Coturnix coturnix represents an excellent avian model, as it demonstrates an annual cycle of testis size, sexual steroid production, and spermatogenesis [33,34,35]. A previous study by our research group revealed that in quail testis, the levels of both E2 and androgens were higher during the reproductive period compared to the non-reproductive period, suggesting that estrogens act in synergy with androgens for the initiation of spermatogenesis [35]. Therefore, the present study was aimed to investigate the estrogen responsive system in quail testis in relation to reproduction seasonality. In order to achieve this objective, an analysis of the ERα signaling and the potential participation of ERK1/2 and Akt-1, in both active and regressive quail testes was performed, with particular focus on the proliferative and apoptotic pathways.

2. Materials and Methods

2.1. Animals

Adult males of quails, C. coturnix, (3-month-old) were collected in the surroundings of Naples during January–February (non-reproductive period) and July–August (reproductive period), according to the Ministry of Health guidelines as previously reported [36]. For each period, 10 animals were used. For each animal, the testes were dissected out, weighed and rapidly immersed either in Bouin’s fluid (Sigma-Aldrich, Milan, Italy) and then embedded in paraffin, for histological investigations, or in liquid nitrogen and then stored at −80 °C for biochemical analysis.

2.2. 17β-Estradiol Assay

17β-estradiol levels were determined in the testis of quails from both non-reproductive and reproductive periods using an enzyme immunoassay kit (DiaMetra, Milan, Italy). Testes (n = 5 from each period) were homogenized 1:10 (w/v) with PBS ×1. The homogenate, obtained from each testis, was then mixed vigorously with ethyl ether (1:10 v/v) and the ether phase was withdrawn after centrifugation at 3000× g for 10 min. The upper phase (ethyl ether) was transferred to a glass tube and was left to evaporate on a hot plate at 40–50 °C under a hood. The residue was dissolved in 0.25 mL of 0.05 M sodium phosphate buffer, pH 7.5, containing bovine serum albumin at a concentration of 10 mg/mL, and then utilized for the ELISA assay [37,38,39]. All samples (three replicates for each sample) ran in the same ELISA. The used 17β-estradiol ELISA kit has the following characteristics: sensitivity of 8.68 pg/mL, an intra-assay variability < 9% and an inter-assay variability < 10%.

2.3. Immunohistochemistry

Quail testes from reproductive (n = 5) and non-reproductive (n = 5) periods were embedded in paraffin. Each paraffin-embedded reproductive and non-reproductive quail testis was cut (5 μm-thick), and the serial sections mounted on poly-L-lysine slides were processed for immunohistochemistry [40,41]. Briefly, deparaffinized sections were washed in 0.1 M phosphate-buffered saline (PBS; pH 7.6), and then incubated in 2.5% H2O2. To reduce no-specific background the sections were incubated in normal goat serum (Pierce, Rockford, IL, USA) for 1 h at room temperature. Next, sections were treated overnight at 4 °C with the primary antibodies diluted in normal goat serum: (i) rabbit polyclonal antibody ERα (1:400, Abcam, Cambridge, MA, USA), and (ii) mouse monoclonal antibody PCNA (1:300, Sigma-Aldrich, Milan, Italy). After washing in PBS, the sections were incubated for 1 h at room temperature with a biotin-conjugated goat anti-rabbit/mouse secondary antibody (1:2000; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and an avidin-biotin-peroxidase complex (ABC immune peroxidase kit, Pierce, Rockford, IL, USA), using DAB (Sigma Aldrich, Milan, Italy) as chromogen. Sections were counterstained with Mayer’s hematoxylin. For negative controls, the primary antibody was omitted. Immunohistochemical signal was observed using a Zeiss Axioskop microscope; images were acquired by using AxioVision 4.7 software (Zeiss, Oberkochen, Germany). The immunostaining for both ERα and PCNA was carried out at the same time. Three investigators individually blindly rank the intensity of staining using a light microscope; afterwards, one researcher took digital photographs.

2.4. cAMP and cGMP Enzyme Immunoassay

The second messengers, cAMP and cGMP, play a key role in the signal transduction implicated in the regulation of spermatogenesis [42,43]. Frozen testes from reproductive (n = 5) and non-reproductive (n = 5) quails were weighed and homogenized in 10 volumes of cold 5% TCA. Then, the homogenate obtained from each testis was centrifuged at 600× g for 10 min; the supernatant was extracted with 3 volumes of water-saturated ether and aqueous extract was dried. Samples were reconstituted with 0.6 mL of assay buffer 2, a buffer containing proteins, detergents and sodium azide as preservative (A5219, Sigma Aldrich, St. Louis, MO, USA) and then utilized for cAMP (CA201) and cGMP (CG201) enzyme immunoassay (Sigma Aldrich, St. Louis, MO, USA). The acetylated version of both kits was running and the sensitivities were 0.039 pmol/mL for cAMP and 0.088 pmol/mL for cGMP.

2.5. Protein Extraction and Western Blot Analysis

Western blot analysis has been performed to investigate the ERα protein expression as well as ERK1/2 and Akt-1 phosphorylations, in both active and regressive quail testes. To determine the testicular proliferative rate in the quail during both reproductive and non-reproductive periods we investigated the protein expression of proliferating cell nuclear antigen (PCNA), a marker of DNA synthesis [44]. SYCP3, encoding the synaptonemal complex proteins and expressed at the meiotic prophase, has been used as meiotic marker [45,46]. Finally, to study the apoptosis pathway, protein levels of Bax and cytochrome c were assayed [47].
Each testis from reproductive (n = 5) and non-reproductive (n = 5) quails were homogenized directly in lysis buffer containing 50 mM HEPES, 150 mM NaCl, 1 mM EDTA, 1 mM EGTA, 10% glycerol, 1% Triton X-100 (1:2 w/v), 1 mM phenylmethylsulphonyl fluoride (PMSF), 1 μg aprotinin, 0.5 mM sodium orthovanadate, and 20 mM sodium pyrophosphate, pH 7.4 (Sigma Chemical Corporation, St. Louis, MO, USA), then clarified by centrifugation at 14,000× g for 10 min [35]. Protein concentration was determined by the Bradford assay (Bio-Rad, Melville, NY, USA). Fifty micrograms of total protein extracts for each sample (reproductive and non-reproductive testes) were boiled in Laemmli buffer for 5 min at 95 °C before each electrophoresis. Three electrophoresis sodium dodecyl sulfate–PAGE (12% polyacrylamide) were performed at same time. After electrophoresis, proteins were transferred onto a nitrocellulose membrane. The complete transfer was assessed using prestained protein standards (Bio-Rad, Melville, NY, USA). Each membrane was first treated for 1 h with blocking solution (5% no-fat powdered milk in 25 mM Tris, pH 7.4; 200 mM NaCl; 0.5% TritonX-100, TBS/Tween) and then incubated overnight at 4 °C with one of the following primary antibodies: anti-ERα, raised in rabbit, diluited 1:1000 (Abcam, Cambridge, MA, USA), anti-P-ERK1/2, raised in rabbit, diluited 1:1000 (Cell Signaling, Danvers, MA, USA), anti-ERK1/2, raised in rabbit, diluited 1:1000 (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), anti-P-Akt-1, raised in rabbit, diluited 1:1000 (Cell Signaling, Danvers, MA, USA), anti-Akt-1, raised in rabbit, diluited 1:1000 (Cell Signaling, Danvers, MA, USA), anti-PCNA, raised in mouse, diluited 1:1000 (Sigma-Aldrich, Milan, Italy), anti-SYCP3, raised in mouse, diluited 1:250 (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) anti-bax, raised in rabbit, diluited 1:1000 (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), anti-cytochrome complex, raised in rabbit, diluited 1:1000 (Cell Signaling, Danvers, MA, USA). After washing with TBS-tween, membranes were incubated with horseradish-peroxidase anti-rabbit IgG or anti-mouse IgG secondary antibodies for 1 h at room temperature, followed by signal detection using enhanced chemiluminescence (ECL) (Amersham Bioscience, Bath, UK). The number of proteins was quantified using Image J software (National Institutes of Health, Bethesda, Rockville, MD, USA) and normalized with respect to β-actin protein (housekeeping protein), whose expression did not change between two examined periods.

2.6. Statistical Analysis

The values obtained were compared by Student’s t-test for between-group comparisons. The differences were considered statistically significant at p < 0.05 (*) and p < 0.01 (**). All data were expressed as the mean ± standard deviation (SD).

3. Results

3.1. E2 Levels and ERα Protein Expression

E2 concentration and ERα protein expression in quail testis were analyzed during reproductive as well as non-reproductive periods (Figure 1). In particular, E2 levels were observed to be significantly higher in the testes during the reproductive period compared to the values detected in the gonads of non-reproductive quails (130 ± 9 pg/g tissue and 62 ± 7 pg/g tissue, respectively) (Figure 1B).
Western blot analysis revealed that the ERα expression in the testis was significantly higher in the reproductive period compared to the non-reproductive period (Figure 1A,B). These data were supported by the findings of immunohistochemical studies, which revealed an increased signal of ERα in quail testis during the reproductive period (Figure 1C). Specifically, it was observed that during the reproductive period, ERα was localized mainly in Leydig and germ cells, such as spermatocytes I and II, spermatids, and spermatozoa, with the strong positivity detected in spermatocytes I (Figure 1Ca,b). No signal was detected in Sertoli cells and spermatogonia. In the non-reproductive period, a faint positivity was observed only in Sertoli cells (Figure 1Cc). No immunohistochemical signal was detected in the control sections (Figure 1Cd).

3.2. cAMP and cGMP Levels

The levels of cAMP and cGMP in quail testes were analyzed in both reproductive and non-reproductive periods. Concentrations of both cAMP and cGMP were observed to be higher in the reproductive period compared to the non-reproductive period (Table 1).

3.3. ERK1/2 and Akt-1 Activities

ERK1/2 activity, expressed as phosphorylated-ERK1/2 (P-ERK1/2) levels, was significantly higher in the reproductive period compared to the non-reproductive period (Figure 2A). Similarly, during the reproductive period, the quail testes exhibited a significant increase in the Akt-1 activity (Figure 2B).

3.4. PCNA Protein Expression and Immunolocalization

Western blot analysis revealed that during the reproductive period, the PCNA protein levels in the testis were higher than those in the non-reproductive period (Figure 3A,B). Immunohistochemical analysis also revealed a more intense immunopositivity for PCNA in the reproductive period with respect to the non-reproductive period (Figure 3). In both periods, a strong immunohistochemical signal for PCNA was detected in spermatogonia (Figure 3Ca–c), while no signal was detected in Leydig cells (Figure 3Ca–c). In the reproductive period, a faint signal was also detected in Sertoli cells and spermatocytes I (Figure 3Ca,b). No immunohistochemical signal was detected for the control sections (Figure 3Cd).

3.5. SYCP3 Protein Expression

The expression levels of SYCP3 protein in quail testis during the reproductive period were approximately two-fold higher than those during the non-reproductive period (Figure 4).

3.6. Bax and Cytochrome C Protein Expressions

The present study revealed that the levels of both Bax and cytochrome c proteins were significantly higher in the testis during the non-reproductive period with respect to the reproductive period (Figure 5A–C).

4. Discussion

It is well established that estrogen signaling is required for the maintenance of male reproductive function in vertebrates [17,21,22,48]. The present study is the first to investigate the E2/ERα-activated molecular pathways underlying the testicular activity changes in C. coturnix during its seasonal reproductive cycle. Several studies have reported ERα as the predominant form in quail testis [11,15,49], although there is no study on the seasonal pattern of ERα expression in the current literature. In line with our previous study, the present study demonstrated that quail testis exhibits the highest titers of E2 during the reproductive period, when P450 aromatase, the enzyme that converts testosterone into E2, was expressed at the highest levels [35]. Accordingly, during this period, the levels of both cAMP and cGMP were increased. It is well recognized that both cAMP and cGMP play key roles in spermatogenesis, including the regulation of the blood-testis barrier [43,50]. Furthermore, several studies have demonstrated cAMP to be the main intracellular messenger mediating the expression of P450 aromatase [50], which induces E2 production, along with a consequent upregulation of its receptor. The Western blotting and immunohistochemistry analyses conducted in the present study revealed that the expression of the ERα protein was correlated with E2 concentration. Specifically, higher levels of ERα protein were detected in quail testis during the reproductive period compared to those in the non-reproductive period. ERα immunopositivity was observed in Leydig cells as well as germ cells (spermatocytes I and II, spermatids, and spermatozoa) during the reproductive period, while a weak immunohistochemical signal was detected only in Sertoli cells during the non-reproductive period. These findings suggest that testicular E2 concentration could affect the expression of ERα and that E2/ERα levels have a role in the testicular seasonal activity in C. coturnix. In particular, the increased levels of E2-induced ERα could have a key role in the resumption of spermatogenesis during the reproductive period, when both mitosis and meiosis, as well as the differentiation process of germ cells, are activated. Therefore, we have hypothesized that estrogens might act in synergy with androgens, with the levels of the latter also highest during the reproductive period. In contrast, during the non-reproductive period, when both E2 titers and ERα protein expression were significantly reduced, spermatogenesis was blocked. Seasonal fluctuations have also been described in gander testis [16], although in this species the seasonal expression pattern of ERs is different from those of quail. In fact, in gander testis E2 levels and its receptors showed an opposite trend, with the highest E2 levels in the breeding stage and the highest gene and protein expressions of ERs during the non-breeding stage [16]. The authors hypothesized that excessive doses of estrogens could disrupt testis physiology, therefore a downregulation of ERs protein by E2 as a physiological mechanism decreasing the sensitivity of testicular tissue to estrogens has been proposed for gander testes during the breeding season, when the intratesticular E2 concentration was very high [16]. A positive correlation between ERα protein expression and spermatogenic activity was reported in the testes of immature, mature, as well as aged chickens [13,51]. Furthermore, in quail, a decrease in both oxidative stress-induced plasma estradiol and ERα testicular expression was reported to reduce spermatogenesis [15]. Different from birds, in the amphibian Pelophylax esculentus and reptile Podarcis sicula, the estrogenic pathway is essential during the post-reproductive period. It has been proposed that in these species, estradiol mediates the interruption of the reproductive processes inhibiting androgen biosynthesis [39,52,53,54].
Several studies have suggested that estrogen action in the testis might be mediated by the regulation of the activity of mitogen-activated protein kinases (MAPKs) [18,19]. In the present study, the activation of ERK1/2 and Akt-1 pathways in quail testis was investigated. Both ERK1/2 and Akt-1 pathways are reported to play crucial roles in the testis, including the processes of spermatogonia proliferation and meiotic progression of spermatocytes [23,55,56,57,58,59,60]. In the present study, high levels of P-ERK1/2 were detected in quail testis during the reproductive period, while the non-reproductive period testis exhibited considerably low levels of phosphorylated ERK1/2 protein. Concomitantly, the Akt-1 phosphorylation levels were positively correlated with the P-ERK1/2 levels, resulting high in the reproductive period and low during the non-reproductive phase. These findings suggested that during the reproductive period, both ERK1/2 and Akt-1 pathways could be induced by E2 via binding to ERα; conversely, in the non-reproductive period, when both E2 levels and ERα expression are low, the ERK1/2 and Akt-1 pathways remain inactive. These data are consistent with the other studies, which have reported that E2 regulates ERK1/2 and Akt-1 activation in the male germ cells of the frog Rana esculenta and lizard P. sicula via ERs during the respective annual cycles [21,22,61,62]. A positive correlation between E2/ERs signaling and ERK/Akt-1 pathways has also been reported in mammals [63,64]. In vitro experiments have demonstrated a direct role of E2/ERs signaling in the stimulation of proliferation in both somatic [18,65] and germ cells [19,66,67,68,69] through the activation of ERK1/2 and Akt-1. Interestingly, it was observed in the present study that the changes in the activation status of both ERK1/2 and Akt-1 were well-correlated with the quail spermatogenetic activity, indicating a key role of these proteins in the regulation of testicular epithelium proliferation and meiotic progression of germ cells in quail testis. In this regard, an increase in the testicular protein levels of PCNA, a mitotic marker, was observed during the reproductive period compared to the non-reproductive period. These findings were supported by the immunohistochemical analysis, which revealed an increased immunohistochemical signal for PCNA in quail testis during the reproductive period. In particular, PCNA was observed to be localized in both somatic and germ cells, with a more intense immunopositivity in spermatogonia. Interestingly, several studies have evidenced that PCNA is regulated by ERK1/2 and Akt-1 signaling pathways. Specifically, the inhibition of ERK1/2 and Akt-1 pathways downregulates the expression of PCNA in the Sertoli cells of chicken [69] and piglets [70], thereby suppressing their proliferation. Recently, it has been demonstrated that treatment with excitatory D-amino acids, which are known to promote spermatogenesis, increases ERK1/2 and Akt-1 activation, as well as the PCNA protein levels, in both frog [71] and rat testis [57] as well as in the spermatogonial cell line GC-1 [58,59,60,72,73]. Chieffi et al. [21,22] reported that the E2-induced ERK1/2 activation was associated with the increased PCNA protein expression in the spermatogonia of P. esculentus and P. sicula during their respective reproductive periods. Therefore, the findings of the present study suggest that in quail testis, PCNA protein expression could be enhanced through the E2-induced ERK1/2 and Akt-1 activation during the reproductive period, while in the non-reproductive period, when the ERK1/2 and Akt-1 pathways are inactive, the PCNA protein levels are decreased.
A positive correlation between ERK1/2 and Akt-1 pathway activities and the levels of SYCP3 protein, a meiotic marker, observed in quail testis in the present study indicated a key role of ERK1/2 and Akt-1 phosphorylation in the meiotic progression of germ cells during the reproductive cycle. Similarly, a positive correlation between ERK1/2 activation and the increased number of cells positive for SYCP3 was observed in mouse testis in a previous study [45], which also demonstrated that Akt-1 inhibition abolished SYCP3 induction and the meiotic entry of postnatal mouse male germ cells [56]. Furthermore, inhibition of ERK1/2 was reported to suppress the genic expression of SYCP3 in cultured fetal germ cells [74]. Therefore, the findings of the present study are evidence for the local regulation of quail spermatogenesis via ERK/Akt-1 activation or inhibition during the reproductive cycle.
Studies have reported that reduced PCNA levels in quail testis are associated with testicular regression or atrophy via apoptotic induction [29,44,75,76,77]. Consistent with this, the present study revealed an increase in the expression levels of pro-apoptotic proteins, Bax and cytochrome c, in quail testis during the non-reproductive period; in addition, decreased levels of PCNA and SYCP3 were observed, suggesting that apoptosis might be a factor in the regression of quail testicular activity during the non-reproductive phase of the annual cycle. Interestingly, several studies have reported that apoptosis could be induced by the inhibition of ERK1/2 and Akt-1 signaling pathways [78,79,80]. Therefore, we hypothesized that the inactivation of ERK1/2 and Akt-1 signaling pathways in quail testis during the non-reproductive period could have a key role in the induction of apoptotic events and the consequent regression of the gonad.

5. Conclusions

In conclusion, our results indicate that estrogens might be acting in synergy with the androgens in quail testis during the reproductive period. In particular, high levels of E2/ERα induce mitotic and meiotic events in germ cells through the activation of ERK1/2 and Akt-1 pathways. On the contrary, during the non-reproductive period, when the E2/ERα levels are low, ERK1/2 and Akt-1 pathways are inactive and apoptotic events occur. Therefore, it is suggested that the activation or inhibition of these molecular pathways might have crucial roles in the physiological switch “on/off” of the testicular activity in male quails during their seasonal reproductive cycle.

Author Contributions

Conceptualization, A.S., M.M.D.F. and L.R.; Investigation, S.F., F.D.G.R. and L.R.; data curation, A.S., L.R. and S.F.; writing—original draft preparation, A.S. and S.F.; writing—review and editing, A.S., G.C.B., M.M.D.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by University of Campania “Luigi Vanvitelli” grant number B68D19001880005 Project VALERE 2019). The APC was funded by University of Campania “Luigi Vanvitelli”.

Institutional Review Board Statement

The study was conducted according to the ethical provisions imposed by the European Union and permitted by the National Committee of the Italian Ministry of Health on in vivo experimentation before 2013. All treatments were performed before 2013, the samples were collected and stored at the Department of Biology and analyzed to obtain present results.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

Authors declare no conflicts of interest or competing interest.

References

  1. Andò, S.; Ciarcia, G.; Panno, M.L.; Imbrogno, E.; Tarantino, G.; Buffone, M.; Beraldi, E.; Angelini, F.; Botte, V. Sex steroids levels in the plasma and testis during the reproductive cycle of lizard Podarcis s. sicula. Raf. Gen. Comp. Endocrinol. 1992, 85, 1–7. [Google Scholar] [CrossRef]
  2. Deviche, P.; Hurley, L.L.; Fokidis, H.B. Avian testicular structure, function, and regulation. In Hormones and Reproduction in Vertebrates: Birds; Norris, D.O., Lopez, K.H., Eds.; Academic Press: San Diego, CA, USA, 2011; pp. 27–70. [Google Scholar]
  3. Rastogi, R.K.; Pinelli, C.; Polese, G.; D’Aniello, B.; Chieffi Baccari, G. Hormones and reproductive cycles in anuran amphibians. In Hormones and Reproduction of Vertebrates; Norris, D.O., Lopez, K.H., Eds.; Elsevier: Boulder, CO, USA, 2011; Volume 2, pp. 171–186. [Google Scholar]
  4. Hess, R.A.; Cooke, P.S. Estrogen in the male: A historical perspective. Biol. Reprod. 2018, 99, 27–44. [Google Scholar] [CrossRef] [PubMed]
  5. Cooke, P.S.; Nanjappa, M.K.; Ko, C.; Prins, G.S.; Hess, R.A. Estrogens in male physiology. Physiol. Rev. 2017, 97, 995–1043. [Google Scholar] [CrossRef]
  6. Osz, J.; Brelivet, Y.; Peluso-Iltis, C.; Cura, V.; Eiler, S.; Ruff, M.; Bourguet, W.; Rochel, N.; Moras, D. Structural basis for a molecular allosteric control mechanism of cofactor binding to nuclear receptors. Proc. Natl. Acad. Sci. USA 2012, 109, E588–E594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Andrews, J.E.; Smith, C.A.; Sinclair, A.H. Sites of estrogen receptor and aromatase expression in the chicken embryo. Gen. Comp. Endocrinol. 1997, 108, 182–190. [Google Scholar] [CrossRef] [PubMed]
  8. Nakabayashi, O.; Kikuchi, H.; Kikuchi, T.; Mizuno, S. Differential expression of genes for aromatase and estrogen receptor during the gonadal development in chicken embryos. J. Mol. Endocrinol. 1998, 20, 193–202. [Google Scholar] [CrossRef]
  9. Koba, N.; Ohfuji, T.; Ha, Y.; Mizushima, S.; Tsukada, A.; Saito, N.; Shimada, K. Profiles of mRNA expression of FOXL2, P450arom, DMRT1, AMH, P450 c17, SF1, ERa and AR, in relation to gonadal sex differentiation in duck embryo. J. Poult. Sci. 2008, 45, 132–138. [Google Scholar] [CrossRef] [Green Version]
  10. Opalka, M.; Leska, A.; Kamińska, B.; Dusza, L. Oestrogen receptor α and β mRNA expression in testis of ganders fed diets containing different levels of phytoestrogens. J. Anim. Feed. Sci. 2008, 17, 600–607. [Google Scholar] [CrossRef]
  11. Ichikawa, K.; Yamamoto, I.; Tsukada, A.; Saito, N.; Shimada, K. cDNA cloning and mRNA expression of estrogen receptor a in Japanese quail. J. Poult. Sci. 2003, 40, 121–129. [Google Scholar] [CrossRef] [Green Version]
  12. Lakaye, B.; Foidart, A.; Grisar, T.; Balthazart, J. Partial cloning and distribution of estrogen receptor beta in the avian brain. Neuroreporter 1998, 9, 2743–2748. [Google Scholar] [CrossRef]
  13. Gonzàlez-Moràn, M.G. Changes in the immunohistochemical localization of estrogen receptor alpha and in the stereological parameters of the testes of mature and aged chickens (Gallus domesticus). Biochem. Biophys. Res. Commun. 2019, 510, 309–314. [Google Scholar] [CrossRef]
  14. Oliveira, A.G.; Dornas, R.A.P.; Mahecha, G.A.B.; Oliveira, C.A. Occurrence and cellular distribution of estrogen receptors ERα and ERβ in the testis and epididymal region of roosters. Gen. Comp. Endocrinol. 2011, 170, 597–603. [Google Scholar] [CrossRef] [Green Version]
  15. Baghel, K.; Niranjan, M.K.; Srivastava, R. Water and food restriction decreases immunoreactivity of oestrogen receptor alpha and antioxidant activity in testes of sexually mature Coturnix coturnix japonica. J. Anim. Physiol. Anim. Nutr. 2020, 104, 1738–1747. [Google Scholar] [CrossRef]
  16. Baghel, K.; Srivastava, R. Photoperiod dependent expression of estrogen receptor alpha in testes of Japanese quail: Involvement of Withania somnifera in apoptosis amelioration. Biochem. Biophys. Res. Commun. 2021, 534, 957–965. [Google Scholar] [CrossRef]
  17. Leska, A.; Kiezun, J.; Kaminska, B.; Dusza, L. Estradiol concentration and the expression of estrogen receptors in the testes of the domestic goose (Anser anser f.domestica) during the annual reproductive cycle. Domest. Anim. Endocrinol. 2015, 51, 96–104. [Google Scholar] [CrossRef] [PubMed]
  18. Lucas, T.F.; Royer, C.; Siu, E.R.; Lazari, M.F.; Porto, C.S. Expression and signaling of G protein-coupled estrogen receptor 1 (GPER) in rat sertoli cells. Biol. Reprod. 2010, 83, 307–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Sirianni, R.; Chimento, A.; Ruggiero, C.; De Luca, A.; Lappano, R.; Ando, S.; Maggiolini, M.; Pezzi, V. The novel estrogen receptor, G protein-coupled receptor 30, mediates the proliferative effects induced by 17beta-estradiol on mouse spermatogonial GC-1 cell line. Endocrinology 2008, 149, 5043–5051. [Google Scholar] [CrossRef] [Green Version]
  20. Cobb, M.H. MAP kinase pathways. Prog. Biophys. Mol. Biol. 1999, 71, 479–500. [Google Scholar] [CrossRef]
  21. Chieffi, P.; Colucci-D’Amato, G.L.; Staibano, S.; Franco, R.; Tramontano, D. Estradiol-induced mitogen-activated protein kinase (extracellular signal-regulated kinase 1 and 2) activity in the frog (Rana esculenta) testis. J. Endocrinol. 2000, 167, 77–84. [Google Scholar] [CrossRef] [Green Version]
  22. Chieffi, P.; Colucci D’amato, L.; Guarino, F.; Salvatore, G.; Angelini, F. 17b-estradiol induces spermatogonial proliferation through mitogen-activated protein kinase (extracellular signal-regulated kinase 1/2) activity in the lizard (Podarcis s. sicula). Mol. Reprod. Dev. 2002, 61, 218–225. [Google Scholar] [CrossRef]
  23. Sette, C.; Barchi, M.; Bianchini, A.; Conti, M.; Rossi, P.; Geremia, R. Activation of the mitogen-activated protein kinase ERK1 during meiotic progression of mouse pachytene spermatocytes. J. Biol. Chem. 1999, 274, 33571–33579. [Google Scholar] [CrossRef] [Green Version]
  24. Datta, S.R.; Brunet, A.; Greenberg, M.E. Cellular survival: A play in three Akts. Genes. Dev. 1999, 13, 2905–2927. [Google Scholar] [CrossRef] [PubMed]
  25. Kandel, E.S.; Hay, N. The regulation and activities of the multifunctional serine/threonine kinase Akt/PKB. Exp. Cell. Res. 1999, 253, 210–229. [Google Scholar] [CrossRef] [PubMed]
  26. Blottner, S.; Hingst, O.; Meyer, H.H.D. Inverse relationship between testicular proliferation and apoptosis in mammalian seasonal breeders. Theriogenology 1995, 44, 320–328. [Google Scholar] [CrossRef]
  27. Young, K.A.; Nelson, R.J. Mediation of seasonal testicular regression by apoptosis. Reproduction 2001, 122, 677–685. [Google Scholar] [CrossRef] [PubMed]
  28. Scaia, M.F.; Czuchlej, S.C.; Cervino, N.; Ceballos, N.R. Apoptosis, Proliferation and Presence of Estradiol Receptors in the Testes and Bidder’s Organ of the Toad Rhinella Arenarum (Amphibia, Anura). J. Morphol. 2016, 277, 412–423. [Google Scholar] [CrossRef]
  29. Banerjee, S.; Chaturvedi, C.M. Apoptotic mechanism behind the testicular atrophy in photorefractory and scotosensitive quail: Involvement of GnIH induced p-53 dependent Bax- Caspase-3 mediated pathway. J. Photochem. Photobiol. B. Biol. 2017, 176, 124–135. [Google Scholar] [CrossRef]
  30. Islam, M.N.; Tsukahara, N.; Sugita, S. Apoptosis-mediated seasonal testicular regression in the Japanese Jungle crow (Corvus macrorhynchos). Theriogenology 2012, 77, 1854–1865. [Google Scholar] [CrossRef]
  31. Jenkins, L.K.; Ross, W.L.; Young, K.A. Increases in apoptosis and declines in Bcl-XL protein characterize testicular regression in American crows (Corvus brachyrhynchos). Reprod. Fertil. Dev. 2007, 19, 461–469. [Google Scholar] [CrossRef] [Green Version]
  32. Young, K.A.; Ball, G.F.; Nelson, R.J. Photoperiod-induced testicular apoptosis in European starlings (Sturnus vulgaris). Biol. Reprod. 2001, 64, 706–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Baraldi-Artoni, S.M.; Orsi, A.M.; Carvaljo, T.L.L.; Lopes, R.A. The Annual Testicular Cycle of the Domestic Quail (Coturnix coturnix japonica). Anat. Histol. Embryol. 1997, 26, 337–339. [Google Scholar] [CrossRef] [PubMed]
  34. Baraldi-Artoni, S.M.; Orsi, A.M.; Carvaljo, T.L.L.; Vicentin, C.A.; Stefanini, M.A. Seasonal morphology of domestic quail (Coturnix coturnix japonica) testis. Adv. Anat. Embryol. Cell. Biol. 1999, 28, 217–220. [Google Scholar] [CrossRef]
  35. Rosati, L.; Di Fiore, M.M.; Prisco, P.; Di Giacomo Russo, F.; Venditti, M.; Andreuccetti, P.; Chieffi Baccari, G.; Santillo, A. Seasonal expression and cellular distribution of star and steroidogenic enzymes in quail testis. J. Exp. Zool. B Mol. Dev. Evol. 2019, 332, 198–209. [Google Scholar] [CrossRef] [PubMed]
  36. Prisco, M.; Rosati, L.; Agnese, M.; Aceto, S.; Andreuccetti, P.; Valiante, S. Pituitary adenylate cyclase-activating polypeptide in the testis of the quail Coturnix coturnix: Expression, localization, and phylogenetic analysis. Evol. Dev. 2019, 21, 145–156. [Google Scholar] [CrossRef] [PubMed]
  37. Falvo, S.; Di Fiore, M.M.; Burrone, L.; Chieffi Baccari, G.; Longobardi, S.; Santillo, A. Androgen and oestrogen modulation by D- aspartate in rat epididymis. Reprod. Fertil. Dev. 2016, 28, 1865–1872. [Google Scholar] [CrossRef]
  38. Rosati, L.; Prisco, M.; Di Fiore, M.M.; Santillo, A.; Sciarrillo, R.; Valiante, S.; Laforgia, V.; Coraggio, F.; Andreuccetti, P.; Agnese, M. Sex steroid hormone secretion in the wall lizard Podarcis sicula testis: The involvement of VIP. J. Exp. Zool. A Ecol. Genet. Physiol. 2015, 323, 714–721. [Google Scholar] [CrossRef] [PubMed]
  39. Rosati, L.; Prisco, M.; Di Fiore, M.M.; Santillo, A.; Valiante, S.; Andreuccetti, P.; Agnese, M. Role of PACAP on testosterone and 17β-estradiol production in the testis of wall lizard Podarcis sicula. Comp. Biochem. Physiol. Part A Mol. Integr. Physiol. 2016, 191, 180–186. [Google Scholar] [CrossRef] [PubMed]
  40. Rosati, L.; Santillo, A.; Di Fiore, M.M.; Andreuccetti, P.; Prisco, M. Testicular steroidogenic enzymes in the lizard Podarcis sicula during the spermatogenic cycle. C. R. Biol. 2017, 340, 492–498. [Google Scholar] [CrossRef]
  41. Rosati, L.; Agnese, M.; Verderame, M.; Aniello, F.; Venditti, M.; Mita, D.G.; Andreuccetti, P.; Prisco, M. Morphological and molecular responses in ovaries of Mytilus galloprovincialis collected in two different sites of the Naples Bay. J. Exp. Zool. A Ecol. Integr. Physiol. 2019, 331, 52–60. [Google Scholar] [CrossRef] [Green Version]
  42. Di Fiore, M.M.; Lamanna, C. , Assisi, L., Botte, V. Opposing effects of D-aspartic acid and nitric oxide on tuning of testosterone production in mallard testis during the reproductive cycle. Reprod. Biol. Endocrinol. 2008, 6, 28. [Google Scholar] [CrossRef] [Green Version]
  43. Lee, N.P.Y.; Cheng, C.Y. Nitric oxide/nitric oxide synthase, spermatogenesis, and tight junction dynamics. Biol. Reprod. 2004, 70, 267–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Banerjee, S.; Chaturvedi, C.M. Specific neural phase relation of serotonin and dopamine modulate the testicular activity in Japanese quail. J. Cell. Physiol. 2019, 234, 2866–2879. [Google Scholar] [CrossRef]
  45. Grimaldi, P.; Orlando, P.; Di Siena, S.; Lolicato, F.; Petrosino, S.M.; Bisogno, T.; Geremia, R.; De Petrocellis, L.; Di Marzo, V. The endocannabinoid system and pivotal role of the CB2 receptor in mouse spermatogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 11131–11136. [Google Scholar] [CrossRef] [Green Version]
  46. Santillo, A.; Giacco, A.; Falvo, S.; Di Giacomo Russo, F.; Senese, R.; Di Fiore, M.M.; Chieffi Baccari, G.; Lanni, A.; de Lange, P. Mild exercise rescues steroidogenesis and spermatogenesis in rats submitted to food withdrawal. Front. Endocrinol. 2020, 11, 302. [Google Scholar] [CrossRef] [PubMed]
  47. Shukla, K.K.; Mahdi, A.A.; Rajender, S. Apoptosis, spermatogenesis and male infertility. Front. Biosci. 2012, 4, 746–754. [Google Scholar] [CrossRef]
  48. Carreau, S.; Bouraima-Lelong, H.; Delalande, C. Estrogens—New players in spermatogenesis. Reprod. Biol. 2011, 11, 174–193. [Google Scholar] [CrossRef]
  49. Mattsson, A.; Brunström, B. Effects of selective and combined activation of estrogen receptor alpha and beta on reproductive organ development and sexual behaviour in Japanese quail (Coturnix japonica). PLoS ONE 2017, 12, e0180548. [Google Scholar] [CrossRef] [Green Version]
  50. Stocco, C. Aromatase expression in the ovary: Hormonal and molecular regulation. Steroids 2008, 73, 473–487. [Google Scholar] [CrossRef] [Green Version]
  51. Gonzàlez-Moràn, M.G.; Guerra-Araiza, C.; Campos, M.G.; Camacho-Arroyo, I. Histological and sex steroid hormone receptor changes in testes of immature, mature, and aged chickens. Domest. Anim. Endocrinol. 2008, 35, 371–379. [Google Scholar] [CrossRef]
  52. Polzonetti-Magni, A.; Botte, V.; Bellini-Cardellini, L.; Gobbetti, A.; Crasto, A. Plasma sex hormones and post-reproductive period in the green frog, Rana esculenta complex. Gen. Comp. Endocrinol. 1984, 54, 372–377. [Google Scholar] [CrossRef]
  53. Santillo, A.; Falvo, S.; Chieffi Baccari, G.; Di Fiore, M.M. Seasonal changes in gene expression of steroidogenic enzymes, androgen and estrogen receptors in frog testis. Acta Zool. 2017, 98, 221–227. [Google Scholar] [CrossRef]
  54. Santillo, A.; Falvo, S.; Di Fiore, M.M.; Chieffi Baccari, G. Seasonal changes and sexual dimorphism in gene expression of StAR protein, steroidogenic enzymes and sex hormone receptors in the frog brain. Gen. Comp. Endocrinol. 2017, 246, 226–232. [Google Scholar] [CrossRef]
  55. Hasegawa, K.; Namekawa, S.H.; Saga, Y. MEK/ERK signaling directly and indirectly contributes to the cyclical self-renewal of spermatogonial stem cells. Stem Cells. 2013, 31, 2517–2527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Pellegrini, M.; Di Siena, S.; Claps, G.; Di Cesare, S.; Dolci, S.; Rossi, P.; Geremia, R.; Grimaldi, P. Microgravity promotes differentiation and meiotic entry of postnatal mouse male germ cells. PLoS ONE 2010, 5, e9064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Santillo, A.; Falvo, S.; Chieffi, P.; Burrone, L.; Chieffi Baccari, G.; Longobardi, S.; Di Fiore, M.M. D-aspartate affects NMDA receptor-extracellular signal-regulated kinase pathway and upregulates androgen receptor expression in the rat testis. Theriogenology 2014, 81, 744–751. [Google Scholar] [CrossRef]
  58. Santillo, A.; Falvo, S.; Chieffi, P.; Di Fiore, M.M.; Senese, R.; Chieffi Baccari, G. D-Aspartate Induces Proliferative Pathways in Spermatogonial GC-1 Cells. J. Cell. Physiol. 2016, 231, 490–495. [Google Scholar] [CrossRef]
  59. Santillo, A.; Falvo, S.; Di Fiore, M.M.; Di Giacomo Russo, F.; Chieffi, P.; Usiello, A.; Pinelli, C.; Chieffi Baccari, G. AMPA receptor expression in mouse testis and spermatogonial GC-1 cells: A study on its regulation by excitatory amino acids. J. Cell. Biochem. 2019, 120, 11044–11055. [Google Scholar] [CrossRef]
  60. Santillo, A.; Venditti, M.; Minucci, S.; Chieffi Baccari, G.; Falvo, S.; Rosati, L.; Di Fiore, M.M. D-Asp upregulates PREP and GluA2/3 expressions and induces p-ERK1/2 and p-Akt in rat testis. Reproduction 2019, 158, 357–367. [Google Scholar] [CrossRef]
  61. Russo, M.; Troncone, G.; Guarino, F.M.; Angelini, F.; Chieffi, P. Estrogen-Induced Akt-1 Activity in the Lizard (Podarcis s. sicula) Testis. Mol. Reprod. Dev. 2005, 71, 52–57. [Google Scholar] [CrossRef]
  62. Stabile, V.; Russo, M.; Chieffi, P. 17b-Estradiol induces Akt-1 through estrogen receptor-b in the frog (Rana esculenta) male germ cells. Reproduction 2006, 132, 477–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yao, Y.; Chang, X.; Wang, D.; Ma, H.; Wang, H.; Zhang, H.; Li, C.; Wang, J. Roles of ERK1/2 and PI3K/AKT signaling pathways in mitochondria-mediated apoptosis in testes of hypothyroid rats. Toxicol. Res. 2018, 7, 1214–1224. [Google Scholar] [CrossRef] [Green Version]
  64. Zhang, Z.; Duan, L.; Du, X.; Ma, H.; Park, I.; Lee, C.; Zhang, J.; Shi, J. The proliferative effect of estradiol on human prostate stromal cells is mediated through activation of ERK. Prostate 2008, 68, 508–516. [Google Scholar] [CrossRef]
  65. Meroni, S.B.; Galardo, M.N.; Rindone, G.; Gorga, A.; Riera, M.F.; Cigorraga, S.B. Molecular mechanisms and signaling pathways involved in sertoli cell proliferation. Front. Endocrinol. 2019, 10, 224. [Google Scholar] [CrossRef] [PubMed]
  66. Vicini, E.; Loiarro, M.; Di Agostino, S.; Corallini, S.; Capolunghi, F.; Carsetti, R.; Chieffi, P.; Geremia, R.; Stefanini, M.; Sette, C. 17-beta-estradiol elicits genomic and non-genomic responses in mouse male germ cells. J. Cell. Physiol. 2006, 206, 238–245. [Google Scholar] [CrossRef]
  67. La Sala, G.; Farini, D.; De Felici, M. Rapid estrogen signalling in mouse primordial germ cells. Exp. Cell. Res. 2010, 316, 1716–1727. [Google Scholar] [CrossRef]
  68. Boscia, F.; Passaro, C.; Gigantino, V.; Perdona, S.; Franco, R.; Portella, G.; Chieffi, S.; Chieffi, P. High levels of GPR30 protein in human testicular carcinoma in situ and seminomas correlate with low levels of estrogen receptor-Beta and indicate a switch in estrogen responsiveness. J. Cell. Physiol. 2015, 230, 1290–1297. [Google Scholar] [CrossRef] [PubMed]
  69. Xu, K.; Wang, J.; Liu, H.; Zhao, J.; Lu, W. Melatonin promotes the proliferation of chicken sertoli cells by activating the ERK/inhibin alpha subunit signaling pathway. Molecules. 2020, 25, 1230. [Google Scholar] [CrossRef] [Green Version]
  70. Sun, Y.; Yang, W.; Luo, H.; Wang, X.; Chen, Z.; Zhang, J.; Wang, Y.; Li, X. Thyroid hormone inhibits the proliferation of piglet Sertoli cell via PI3K signaling pathway. Theriogenology 2015, 83, 86–94. [Google Scholar] [CrossRef]
  71. Santillo, A.; Chieffi Baccari, G.; Falvo, S.; Di Giacomo Russo, F.; Venditti, M.; Di Fiore, M.M. Effects of D-Aspartate on sex hormone-dependent tissues in Pelophylax esculentus. In Amphibians: Biology, Ecology and Conservation; Cannon, L., Ed.; Nova Science Publishers: New York, NY, USA, 2018; pp. 21–37. [Google Scholar]
  72. Di Fiore, M.M.; Boni, R.; Santillo, A.; Falvo, S.; Gallo, A.; Esposito, S.; Chieffi Baccari, G. D-aspartic acid in vertebrate reproduction: animal models and experimental designs. Biomolecules 2019, 9, 445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Venditti, M.; Santillo, A.; Falvo, S.; Di Fiore, M.M. , Chieffi Baccari, G.; Minucci, S. d-aspartate upregulates daam1 protein levels in the rat testis and induces its localization in spermatogonia nucleus. Biomolecules 2020, 10, 677. [Google Scholar] [CrossRef]
  74. Kim, S.M.; Yokoyama, T.; Ng, D.; Ulu, F.; Yamazaki, Y. Retinoic acid-stimulated ERK1/2 pathway regulates meiotic initiation in cultured fetal germ cells. PLoS ONE 2019, 14, e0224628. [Google Scholar] [CrossRef] [PubMed]
  75. Banerjee, S.; Tsutsui, K.; Chaturvedi, C.M. Apoptosis mediated testicular alteration in Japanese quail (Coturnix coturnix japonica) in response to temporal phase relation of serotonergic and dopaminergic oscillations. J. Exp. Biol. 2016, 219, 1476–1487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Banerjee, S.; Chaturvedi, C.M. Testicular atrophy and reproductive quiescence in photorefractory and scotosensitive quail: Involvement of hypothalamic deep brain photoreceptors and GnRH-GnIH system. J. Photochem. Photobiol. B Biol. 2017, 175, 254–268. [Google Scholar] [CrossRef] [PubMed]
  77. Banerjee, S.; Chaturvedi, C.M. Simulated photoperiod influences testicular activity in quail via modulating local GnRHR-GnIHR, GH-R, Cnx-43 and 14-3-3. J. Photochem. Photobiol. B Biol. 2018, 178, 412–423. [Google Scholar] [CrossRef] [PubMed]
  78. Liu, J.; Li, Q.; Liu, Z.; Lin, L.; Zhang, X.; Cao, M.; Jiang, J. Harmine induces cell cycle arrest and mitochondrial pathway-mediated cellular apoptosis in SW620 cells via inhibition of the Akt and ERK signaling pathways. Oncol. Rep. 2016, 35, 3363–3370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Wang, A.S.; Xu, C.W.; Xie, H.Y.; Yao, A.J.; Shen, Y.Z.; Wan, J.J.; Zhang, H.Q.; Fu, J.F.; Chen, Z.M.; Zou, Z.Q. DHA induces mitochondria-mediated 3T3-L1 adipocyte apoptosis by down-regulation of Akt and ERK. J. Funct. Foods. 2016, 21517–21524. [Google Scholar] [CrossRef]
  80. Li, L.; Wang, X.; Sharvan, R.; Gao, J.; Qu, S. Berberine could inhibit thyroid carcinoma cells by inducing mitochondrial apoptosis, G0/G1 cell cycle arrest and suppressing migration via PI3K-AKT and MAPK signaling pathways. Biomed. Pharmacother. 2017, 951225–951231. [Google Scholar] [CrossRef]
Figure 1. (A) Western blot detection of ERα in quail testis during reproductive and non-reproductive periods. A specific band of 66 kDa was detected. (B) The amount of protein was quantified using the ImageJ program and normalized with respect to β-actin protein. The values presented represent the means ± S.D. of the values obtained for five animals (two bands were shown). * p < 0.05. RP, reproductive period; NRP, non-reproductive period. (C) Immunohistochemical localization (brown areas) of ERα. (a,b) Reproductive period. The signal is evident in Leydig cells (**), spermatocytes I (Spc I) and II (Spc II), spermatids (Spt), and spermatozoa (Spz). No signal is evident in Sertoli cells (arrow) and spermatogonia (Spg). (c) Non-reproductive period. The signal is evident in Sertoli cells (arrow). No positivity was observed in Leydig cells (**) and spermatogonia (Spg). (d) Control section. Scale bars correspond to 5 µm in (a,c,d) and 10 µm in (b).
Figure 1. (A) Western blot detection of ERα in quail testis during reproductive and non-reproductive periods. A specific band of 66 kDa was detected. (B) The amount of protein was quantified using the ImageJ program and normalized with respect to β-actin protein. The values presented represent the means ± S.D. of the values obtained for five animals (two bands were shown). * p < 0.05. RP, reproductive period; NRP, non-reproductive period. (C) Immunohistochemical localization (brown areas) of ERα. (a,b) Reproductive period. The signal is evident in Leydig cells (**), spermatocytes I (Spc I) and II (Spc II), spermatids (Spt), and spermatozoa (Spz). No signal is evident in Sertoli cells (arrow) and spermatogonia (Spg). (c) Non-reproductive period. The signal is evident in Sertoli cells (arrow). No positivity was observed in Leydig cells (**) and spermatogonia (Spg). (d) Control section. Scale bars correspond to 5 µm in (a,c,d) and 10 µm in (b).
Animals 11 01729 g001
Figure 2. Expressions of P-ERK1/2 and P-Akt-1 in quail testis during reproductive and non-reproductive periods. (A) Detection of P-ERK1/2 protein using Western blot analysis. A specific band of 42 kDa was detected. The amount of phosphorylated ERK1/2 was quantified using the ImageJ program and normalized with respect to ERK1/2. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown in the upper panel). ** p < 0.01. (B) Western blot analysis for Akt-1 protein in quail testis during reproductive and non-reproductive periods. A specific band of 60 kDa was detected. The amount of phosphorylated Akt-1 was quantified using the ImageJ program and normalized with respect to Akt-1. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown). ** p < 0.01. RP, reproductive period; NRP, non-reproductive period.
Figure 2. Expressions of P-ERK1/2 and P-Akt-1 in quail testis during reproductive and non-reproductive periods. (A) Detection of P-ERK1/2 protein using Western blot analysis. A specific band of 42 kDa was detected. The amount of phosphorylated ERK1/2 was quantified using the ImageJ program and normalized with respect to ERK1/2. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown in the upper panel). ** p < 0.01. (B) Western blot analysis for Akt-1 protein in quail testis during reproductive and non-reproductive periods. A specific band of 60 kDa was detected. The amount of phosphorylated Akt-1 was quantified using the ImageJ program and normalized with respect to Akt-1. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown). ** p < 0.01. RP, reproductive period; NRP, non-reproductive period.
Animals 11 01729 g002
Figure 3. (A) Western blot analysis of testicular PCNA expression in quail during reproductive and non-reproductive periods. A specific band of 36 kDa was detected. (B) The amount of PCNA was quantified using the ImageJ program and normalized with respect to β-actin. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown). * p < 0.05. RP, reproductive period; NRP, non-reproductive period. (C) Immunohistochemical localization (brown areas) of PCNA. (a,b) Reproductive period. The signal is evident in spermatogonia (Spg), spermatocytes I (Spc I), and Sertoli (arrow) cells. No signal is evident in Leydig cells (**), spermatocytes II (Spc II), spermatids (Spt), and spermatozoa (Spz). (c) Non-reproductive period. The signal is evident only in spermatogonia (Spg). No positivity was detected in Leydig cells (**) and Sertoli cells (arrow). (d) Control section. Scale bars correspond to 5 µm in (a,c,d) and 10 µm in (b).
Figure 3. (A) Western blot analysis of testicular PCNA expression in quail during reproductive and non-reproductive periods. A specific band of 36 kDa was detected. (B) The amount of PCNA was quantified using the ImageJ program and normalized with respect to β-actin. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown). * p < 0.05. RP, reproductive period; NRP, non-reproductive period. (C) Immunohistochemical localization (brown areas) of PCNA. (a,b) Reproductive period. The signal is evident in spermatogonia (Spg), spermatocytes I (Spc I), and Sertoli (arrow) cells. No signal is evident in Leydig cells (**), spermatocytes II (Spc II), spermatids (Spt), and spermatozoa (Spz). (c) Non-reproductive period. The signal is evident only in spermatogonia (Spg). No positivity was detected in Leydig cells (**) and Sertoli cells (arrow). (d) Control section. Scale bars correspond to 5 µm in (a,c,d) and 10 µm in (b).
Animals 11 01729 g003
Figure 4. Western blot detections for SYCP3 in quail testis during reproductive and non-reproductive periods. A specific band of 33 kDa was detected. The amount of protein was quantified using the ImageJ program and normalized with respect to β-actin protein. The values presented represent the means ± S.D. of the values obtained for five animals (two bands were shown). ** p < 0.01 for reproductive period vs. non-reproductive period. RP, reproductive period; NRP, non-reproductive period.
Figure 4. Western blot detections for SYCP3 in quail testis during reproductive and non-reproductive periods. A specific band of 33 kDa was detected. The amount of protein was quantified using the ImageJ program and normalized with respect to β-actin protein. The values presented represent the means ± S.D. of the values obtained for five animals (two bands were shown). ** p < 0.01 for reproductive period vs. non-reproductive period. RP, reproductive period; NRP, non-reproductive period.
Animals 11 01729 g004
Figure 5. Bax and cytochrome c protein levels in quail testes during reproductive and non-reproductive periods. (A) Western blot analysis for Bax (23 kDa) and cytochrome c (14 kDa) proteins in quail testis during reproductive and non-reproductive periods. The amounts of Bax (B) and cytochrome c (C) were quantified using the ImageJ program and normalized with respect to β-actin. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown). * p < 0.05 for reproductive period vs. non-reproductive period. RP, reproductive period; NRP, non-reproductive period.
Figure 5. Bax and cytochrome c protein levels in quail testes during reproductive and non-reproductive periods. (A) Western blot analysis for Bax (23 kDa) and cytochrome c (14 kDa) proteins in quail testis during reproductive and non-reproductive periods. The amounts of Bax (B) and cytochrome c (C) were quantified using the ImageJ program and normalized with respect to β-actin. The values presented represent the means ± S.D. of the values obtained for five samples (two bands were shown). * p < 0.05 for reproductive period vs. non-reproductive period. RP, reproductive period; NRP, non-reproductive period.
Animals 11 01729 g005
Table 1. cAMP and cGMP levels in reproductive and non-reproductive testes of quail C. coturnix.
Table 1. cAMP and cGMP levels in reproductive and non-reproductive testes of quail C. coturnix.
Reproductive Non-Reproductive
cAMP (pmol/g tissue)45.4 ± 3.6 *33.6 ± 2.7
cGMP (pmol/g tissue)31.7 ± 1.2 **14.5 ± 1.9
** p < 0.01 and * p < 0.05, reproductive vs. non-reproductive period.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Falvo, S.; Rosati, L.; Di Fiore, M.M.; Di Giacomo Russo, F.; Chieffi Baccari, G.; Santillo, A. Proliferative and Apoptotic Pathways in the Testis of Quail Coturnix coturnix during the Seasonal Reproductive Cycle. Animals 2021, 11, 1729. https://doi.org/10.3390/ani11061729

AMA Style

Falvo S, Rosati L, Di Fiore MM, Di Giacomo Russo F, Chieffi Baccari G, Santillo A. Proliferative and Apoptotic Pathways in the Testis of Quail Coturnix coturnix during the Seasonal Reproductive Cycle. Animals. 2021; 11(6):1729. https://doi.org/10.3390/ani11061729

Chicago/Turabian Style

Falvo, Sara, Luigi Rosati, Maria Maddalena Di Fiore, Federica Di Giacomo Russo, Gabriella Chieffi Baccari, and Alessandra Santillo. 2021. "Proliferative and Apoptotic Pathways in the Testis of Quail Coturnix coturnix during the Seasonal Reproductive Cycle" Animals 11, no. 6: 1729. https://doi.org/10.3390/ani11061729

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop