Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters











Publication year range
1.
Biology (Basel) ; 13(4)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38666890

ABSTRACT

Experimental autoimmune orchitis (EAO) is a well-established rodent model of organ-specific autoimmunity associated with infertility in which the testis immunohistopathology has been extensively studied. In contrast, analysis of testis biopsies from infertile patients associated with inflammation has been more limited. In this work, testicular biopsies from patients with idiopathic non-obstructive azoospermia diagnosed with hypospermatogenesis (HypoSp) [mild: n = 9, and severe: n = 11], with obstructive azoospermia and complete Sp (spermatogenesis) (control group, C, n = 9), and from Sertoli cell-only syndrome (SCOS, n = 9) were analyzed for the presence of immune cells, spermatogonia and Sertoli cell (SCs) alterations, and reproductive hormones levels. These parameters were compared with those obtained in rats with EAO. The presence of increased CD45+ cells in the seminiferous tubules (STs) wall and lumen in severe HypoSp is associated with increased numbers of apoptotic meiotic germ cells and decreased populations of undifferentiated and differentiated spermatogonia. The SCs showed an immature profile with the highest expression of AMH in patients with SCOS and severe HypoSp. In SCOS patients, the amount of SCs/ST and Ki67+ SCs/ST increased and correlated with high serum FSH levels and CD45+ cells. In the severe phase of EAO, immune cell infiltration and apoptosis of meiotic germ cells increased and the number of undifferentiated and differentiated spermatogonia was lowest, as previously reported. Here, we found that orchitis leads to reduced sperm number, viability, and motility. SCs were mature (AMH-) but increased in number, with Ki67+ observed in severely damaged STs and associated with the highest levels of FSH and inflammatory cells. Our findings demonstrate that in a scenario where a chronic inflammatory process is underway, FSH levels, immune cell infiltration, and immature phenotypes of SCs are associated with severe changes in spermatogenesis, leading to azoospermia. Furthermore, AMH and Ki67 expression in SCs is a distinctive marker of severe alterations of STs in human orchitis.

2.
Gene ; 897: 148082, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38101710

ABSTRACT

Transforming growth factor-ß (TGF-ß) and bone morphogenetic protein (BMP) signaling has fundamental roles in the regulation of the stem cell niche for both embryonic and adult stem cells. In zebrafish, male germ stem cell niche is regulated by follicle-stimulating hormone (Fsh) through different members of the TGF-ß superfamily. On the other hand, the specific roles of TGF-ß and BMP signaling pathways are unknown in the zebrafish male germ stem cell niche. Considering this lack of information, the present study aimed to investigate the pharmacological inhibition of TGF-ß (A83-01) and BMP (DMH1) signaling pathways in the presence of recombinant zebrafish Fsh using testicular explants. We also reanalyzed single cell-RNA sequencing (sc-RNA-seq) dataset from adult zebrafish testes to identify the testicular cellular sites of smad expression, and to understand the physiological significance of the changes in smad transcript levels after inhibition of TGF-ß or BMP pathways. Our results showed that A83-01 potentiated the pro-stimulatory effects of Fsh on spermatogonial differentiation leading to an increase in the proportion area occupied by differentiated spermatogonia with concomitant reduction of type A undifferentiated (Aund) spermatogonia. In agreement, expression analysis showed lower mRNA levels for the pluripotency gene pou5f3, and increased expression of dazl (marker of type B spermatogonia and spermatocyte) and igf3 (pro-stimulatory growth factor) following the co-treatment with TGF-ß inhibitor and Fsh. Contrariwise, the inhibition of BMP signaling nullified the pro-stimulatory effects of Fsh, resulting in a reduction of differentiated spermatogonia and increased proportion area occupied by type Aund spermatogonia. Supporting this evidence, BMP signaling inhibition increased the mRNA levels of pluripotency genes nanog and pou5f3, and decreased dazl levels when compared to control. The sc-RNA-seq data unveiled a distinctive pattern of smad expression among testicular cells, primarily observed in spermatogonia (smad 2, 3a, 3b, 8), spermatocytes (smad 2, 3a, 8), Sertoli cells (smad 1, 3a, 3b), and Leydig cells (smad 1, 2). This finding supports the notion that inhibition of TGF-ß and BMP signaling pathways may predominantly impact cellular components within the spermatogonial niche, namely spermatogonia, Sertoli, and Leydig cells. In conclusion, our study demonstrated that TGF-ß and BMP signaling pathways exert antagonistic roles in the zebrafish germ stem cell niche. The members of the TGF-ß subfamily are mainly involved in maintaining the undifferentiated state of spermatogonia, while the BMP subfamily promotes spermatogonial differentiation. Therefore, in the complex regulation of the germ stem cell niche by Fsh, members of the BMP subfamily (pro-differentiation) should be more predominant in the niche than those belonging to the TGF-ß (anti-differentiation). Overall, these findings are not only relevant for understanding the regulation of germ stem cell niche but may also be useful for expanding in vitro the number of undifferentiated spermatogonia more efficiently than using recombinant hormones or growth factors.


Subject(s)
Pyrazoles , Spermatogonia , Thiosemicarbazones , Zebrafish , Animals , Male , Spermatogonia/metabolism , Zebrafish/genetics , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/metabolism , Transforming Growth Factor beta/metabolism , Testis/metabolism , Cell Differentiation/genetics , RNA, Messenger/genetics , Spermatogenesis/genetics
3.
Methods Mol Biol ; 2656: 7-20, 2023.
Article in English | MEDLINE | ID: mdl-37249864

ABSTRACT

Numerous methods have been successfully used to evaluate mammalian spermatogonial biology However, the conventional light microscopy assays present a challenge in precisely identifying spermatogonial phenotypes, which can result in discrepancies between molecular and morphological findings. Such precise association could lead to a more robust interpretation of spermatogonial activity in steady-state spermatogenesis, which may facilitate the translation from basic research to clinical applications. In this chapter, we present two histological processing methods that enable a comprehensive analysis of spermatogonial morphology and function, involving fixation of mammalian testicular tissue in glutaraldehyde and embedding in plastic resin. These techniques have proven to be effective in light microscopy studies.


Subject(s)
Spermatogonia , Testis , Male , Animals , Spermatogenesis , Mammals , Tissue Fixation/methods
4.
BMC Biol ; 21(1): 36, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36797789

ABSTRACT

BACKGROUND: Cellular entry of SARS-CoV-2 has been shown to rely on angiotensin-converting enzyme 2 (ACE2) receptors, whose expression in the testis is among the highest in the body. Additionally, the risk of mortality seems higher among male COVID-19 patients, and though much has been published since the first cases of COVID-19, there remain unanswered questions regarding SARS-CoV-2 impact on testes and potential consequences for reproductive health. We investigated testicular alterations in non-vaccinated deceased COVID-19-patients, the precise location of the virus, its replicative activity, and the immune, vascular, and molecular fluctuations involved in the pathogenesis. RESULTS: We found that SARS-CoV-2 testicular tropism is higher than previously thought and that reliable viral detection in the testis requires sensitive nanosensors or RT-qPCR using a specific methodology. Through an in vitro experiment exposing VERO cells to testicular macerates, we observed viral content in all samples, and the subgenomic RNA's presence reinforced the replicative activity of SARS-CoV-2 in testes of the severe COVID-19 patients. The cellular structures and viral particles, observed by transmission electron microscopy, indicated that macrophages and spermatogonial cells are the main SARS-CoV-2 lodging sites, where new virions form inside the endoplasmic reticulum Golgi intermediate complex. Moreover, we showed infiltrative infected monocytes migrating into the testicular parenchyma. SARS-CoV-2 maintains its replicative and infective abilities long after the patient's infection. Further, we demonstrated high levels of angiotensin II and activated immune cells in the testes of deceased patients. The infected testes show thickening of the tunica propria, germ cell apoptosis, Sertoli cell barrier loss, evident hemorrhage, angiogenesis, Leydig cell inhibition, inflammation, and fibrosis. CONCLUSIONS: Our findings indicate that high angiotensin II levels and activation of mast cells and macrophages may be critical for testicular pathogenesis. Importantly, our findings suggest that patients who become critically ill may exhibit severe alterations and harbor the active virus in the testes.


Subject(s)
COVID-19 , Testis , Viral Tropism , Animals , Humans , Male , Angiotensin II/metabolism , Chlorocebus aethiops , COVID-19/pathology , SARS-CoV-2 , Testis/immunology , Testis/virology , Vero Cells
5.
Andrologia ; 54(4): e14367, 2022 May.
Article in English | MEDLINE | ID: mdl-35034376

ABSTRACT

The reactive oxygen species (ROS) play an important role in various aspects of male reproductive function, for spermatozoa to acquire the ability to fertilize. However, the increase in ROS generation, both due to internal and external factors, can induce oxidative stress, causing alterations in the structure and function of phospholipids and proteins. In the nucleus, ROS attack DNA, causing its fragmentation and activation of apoptosis, thus altering gene and protein expression. Accumulating evidence also reveals that endogenously produced ROS can act as second messengers in regulating cell signalling pathways and in the transduction of signals that are responsible for regulating spermatogonia self-renewal and proliferation. In the epididymis, they actively participate in the formation of disulphide bridges required for the final condensation of chromatin, as well as in the phosphorylation and dephosphorylation of proteins contained in the fibrous sheath of the flagellum, stimulating the activation of progressive motility in epididymal spermatozoa. In this review, the role of small amounts of ROS during spermatogenesis and epididymal sperm maturation was discussed.


Subject(s)
Epididymis , Testis , Epididymis/metabolism , Humans , Male , Reactive Oxygen Species/metabolism , Sperm Maturation/physiology , Spermatozoa/metabolism , Testis/metabolism
6.
Vet. Not. (Online) ; 27(3): 1-23, 1 out. 2021. ilus
Article in Portuguese | VETINDEX | ID: biblio-1502548

ABSTRACT

O transplante de espermatogônias tronco (SSCs, do inglês Spermatogonial Stem Cell) é uma biotecnologia que consiste na transferência de células tronco testiculares de um doador fértil para um receptor cuja espermatogênese endógena foi suprimida. Essa técnica pode ser aplicada para a produção de machos que gerem uma progênie com características genotípicas do doador selecionado. Especialmente na bovinocultura, tanto de leite como de corte, o transplante de SSCs tem o potencial de substituir a inseminação artificial (IA). Pode-se também, colocar SSCs de um mesmo doador (de genética superior) em mais de um receptor o que aumentaria o número de filhos desse doador. Além disso, possui outras aplicações como a restauração da fertilidade em homens após o tratamento de câncer, conservação de espécies ameaçadas de extinção e tratamento de causas específicas de infertilidade. Assim, com esta revisão tem-se como propósito discorrer acerca de uma biotecnologia da reprodução que permitirá a propagação do valor genético de doadores de sêmen considerados de alto valor zootécnico.


Spermatogonial Stem Cell (SSCs) transplantation is a biotechnology that consists in the transfer of testicular stem cells from a fertile donor to a recipient whose endogenous spermatogenesis has been depleted. This technique can be applied to the production of males that generate a progeny with genotypic characteristics of the selected donor. Especially in beef and dairy cattle, SSCs transplantation has the potential to replace artificial insemination (AI). In addition, it has other applications such as restoring fertility in human species after cancer treatment, conserving endangered species and treating specific causes of infertility. Thus, this aim of this review is to discuss the perspectives of reproductive biotechnology that allows the propagation of the genetic material of high pedigree males.


Subject(s)
Male , Animals , Bioengineering/history , Spermatogonia , Stem Cell Transplantation/veterinary , Biotechnology , Insemination, Artificial/trends , Insemination, Artificial/veterinary
7.
Vet. Not. ; 27(3): 1-23, 1 out. 2021. ilus
Article in Portuguese | VETINDEX | ID: vti-32108

ABSTRACT

O transplante de espermatogônias tronco (SSCs, do inglês Spermatogonial Stem Cell) é uma biotecnologia que consiste na transferência de células tronco testiculares de um doador fértil para um receptor cuja espermatogênese endógena foi suprimida. Essa técnica pode ser aplicada para a produção de machos que gerem uma progênie com características genotípicas do doador selecionado. Especialmente na bovinocultura, tanto de leite como de corte, o transplante de SSCs tem o potencial de substituir a inseminação artificial (IA). Pode-se também, colocar SSCs de um mesmo doador (de genética superior) em mais de um receptor o que aumentaria o número de filhos desse doador. Além disso, possui outras aplicações como a restauração da fertilidade em homens após o tratamento de câncer, conservação de espécies ameaçadas de extinção e tratamento de causas específicas de infertilidade. Assim, com esta revisão tem-se como propósito discorrer acerca de uma biotecnologia da reprodução que permitirá a propagação do valor genético de doadores de sêmen considerados de alto valor zootécnico.(AU)


Spermatogonial Stem Cell (SSCs) transplantation is a biotechnology that consists in the transfer of testicular stem cells from a fertile donor to a recipient whose endogenous spermatogenesis has been depleted. This technique can be applied to the production of males that generate a progeny with genotypic characteristics of the selected donor. Especially in beef and dairy cattle, SSCs transplantation has the potential to replace artificial insemination (AI). In addition, it has other applications such as restoring fertility in human species after cancer treatment, conserving endangered species and treating specific causes of infertility. Thus, this aim of this review is to discuss the perspectives of reproductive biotechnology that allows the propagation of the genetic material of high pedigree males.(AU)


Subject(s)
Animals , Male , Bioengineering/history , Stem Cell Transplantation/veterinary , Spermatogonia , Biotechnology , Insemination, Artificial/trends , Insemination, Artificial/veterinary
8.
Front Vet Sci ; 8: 597964, 2021.
Article in English | MEDLINE | ID: mdl-33816583

ABSTRACT

This is the first time that testicular tissue (n = 44) and isolated testicular cells (n = 51) were cryopreserved from alpaca testes 24 h postmortem. For this purpose, internally designed freezing media and cryopreservation protocols were used. Testicular tissue fragments (25 mg) and isolated testicular cells were frozen in MTDB (trehalose and black maca), MTD (trehalose), MSDB (sucrose and black maca), and MSD (sucrose) media. Isolated spermatogonial cells were cryopreserved in two ways, before and after proliferation in vitro. After cryopreservation, the percentage of cell viability in Group 1 (>50% of cell viability) by trypan blue did not show differences within each group (p > 0.05) but showed significant differences when comparing fragments with isolated cells (p < 0.05). Spermatogonial stem cells (SSC) were identified by flow cytometry as strong Dolichos biflorus agglutinin (sDBA) and mitochondrial activity of SSC as strongly positive for MitoSense (sMitoSense+) in intact mitochondria cells, weakly positive for MitoSense (wMitoSense+) in early apoptosis, and necrosis with 7-Aminoactinomycin-D positive (7-AAD). After freezing, in Group 1M (≥30% sMitoSense+), the fragments did not show differences between the media (p > 0.05), but in the isolated cells frozen in MSDB medium, 63.68 ± 8.90% (p < 0.05). In Group 2M (<30% sMitoSense+), necrosis (7AAD+) in MSDB medium was 27.03 ± 5.80%, and necrosis in isolated cells was 14.05 ± 9.3% with significant differences between these groups (p < 0.05); in sMitoSense+, the isolated cells (34.40 ± 23%) had a higher percentage than the fragments (12.4 ± 5.2) (p < 0.05). On the other hand, MSDB and MSD media were significantly higher for isolated cells than for fragments in sDBA+ (p < 0.05). On the other hand, the SSC (sDBA+) had significant differences (p < 0.05) between fresh cells 7.43 ± 1.3% (sDBA+) compared with those cryopreserved in MSDB medium 1.46 ± 0.34% (sDBA+). Additionally, the proliferated and cryopreserved SSC 6.29 ± 1.17% (sDBA+) did not show significant differences concerning the fresh cells (p > 0.05). In conclusion, the black maca showed antioxidant properties when it was included in the freezing medium and, therefore, improved the SSC's conservation of the alpaca. Furthermore, the proliferation of isolated cells in vitro produces a higher amount of SSC after thawing them for further preclinical or clinical work.

9.
J Histochem Cytochem ; 68(5): 327-342, 2020 05.
Article in English | MEDLINE | ID: mdl-32228279

ABSTRACT

The aim of this study was to investigate carnitine action against negative effects of etoposide on stem/progenitor spermatogonia and on sperm production. Carnitine (250 mg/kg body weight/day) and etoposide (5 mg/kg body weight/day) were administered from 25-days postpartum to 32-days postpartum. Testes were collected at 32-days postpartum, 64-days postpartum, and 127-days postpartum, and submitted to the immuno-labeling of UTF1, SOX2, and PLZF proteins to identify undifferentiated spermatogonia populations. At 127-days postpartum, sperm were collected for analysis. Carnitine+etoposide group showed a higher numerical density of spermatogonia labeled for all studied proteins at 64-days postpartum (critical age) compared to the etoposide group. Moreover, there was an improvement of spermatic parameters and sperm DNA integrity in rats of the carnitine+etoposide group in comparison with rats of the etoposide group. The results suggest that carnitine improves the self-renewal of undifferentiated spermatogonia and promotes a partial protection on them, alleviating the etoposide harmful late effects and leading to an enhancement of the sperm parameters in adulthood.


Subject(s)
Carnitine/pharmacology , Cell Self Renewal/drug effects , Etoposide/toxicity , Spermatogonia/cytology , Spermatogonia/drug effects , Animals , DNA Damage , Dose-Response Relationship, Drug , Male , Organ Size/drug effects , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Rats , SOXB1 Transcription Factors/metabolism , Seminiferous Epithelium/drug effects , Seminiferous Epithelium/growth & development , Spermatogenesis/drug effects , Spermatogonia/metabolism , Testis/drug effects , Testis/growth & development , Transcription Factors/metabolism
10.
Reprod Biol ; 19(4): 329-339, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31757605

ABSTRACT

During an inflammatory process of the testis, the network of somatic, immune, and germ cell interactions is altered leading to organ dysfunction. In testicular biopsies of infertile men, spermatogenesis impairment is associated with reduced spermatogonia proliferation, increased number of immune cells, and content of pro-inflammatory cytokines. TNFα-TNFR and nitric oxide (NO)-NO synthase systems are up-regulated in models of testicular damage and in human testis with maturation arrest. The purpose of this study was to test the hypothesis that TNFα-TNFR system and NO alter the function of spermatogonia in the inflamed testis. We studied the effect of TNFα and NO on GC-1 spermatogonia cell cycle progression and death by flow cytometry. GC-1 cells expressed TNFR1 and TNFR2 (immunofluorescence). TNFα (10 and 50 ng/ml) and DETA-Nonoate (0.5 and 2 mM), a NO releaser, increased the percentage of cells in S-phase of the cell cycle and reduced the percentage in G1, inducing also cell apoptosis. TNFα effect was not mediated by oxidative stress unlike NO, since the presence of N-acetyl-l-cysteine (2.5 and 5.0 mM) prevented NO induced cell cycle arrest and death. GC-1 spermatogonia overpass NO induced cell cycle arrest but no TNFα, since after removal of NO, spermatogonia progressed through the cell cycle. We propose TNFα and NO might contribute to impairment of spermatogenesis by preventing adequate functioning of the spermatogonia population. Our results showed that TNFα and NO impaired spermatogonia cell cycle, inducing GC-1 arrest in the S phase.


Subject(s)
Inflammation/physiopathology , Nitric Oxide/physiology , Spermatogonia/physiology , Tumor Necrosis Factor-alpha/physiology , Apoptosis , Cell Cycle , Cell Line , Humans , Male , Oxidative Stress , Receptors, Tumor Necrosis Factor/metabolism , Spermatogenesis
11.
Toxicol Mech Methods ; 29(7): 488-498, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31050326

ABSTRACT

The increased incidence of testicular disorders in young men and the possible influence of environmental chemicals, such as dibutyl phthalate (DBP) and acrylamide (AA), requires experimental models for identifying modes of action. Most published reproductive toxicologic studies use RNA samples from the total testis to evaluate testicular gene expression; however, analyses of isolated cell types could provide a more specific tool. Among testicular germ cells, spermatogonia are critical since they represent the onset of spermatogenesis. This study aimed, (1) to establish a technique for spermatogonia isolation; (2) to apply this isolation technique to verify possible gene expression alterations (Pou5f1, Kitlg, Mki-67, Bak1 and Spry4) in prepubertal post-natal day, (PND24) and pubertal (PND45) testes after in utero and postnatal exposure to DBP or AA. The technique was efficient for isolation of a majority of spermatogonia. In utero DBP exposure led to reduced litter body weight at birth, reduced anogenital distance of male pups on PND4, and increased frequency of male nipple retention on PND14 compared to controls. DBP-exposed relative testes weights were reduced only at PND24 compared to control but they did not differ at PND45. DBP-exposed animals showed reduced expression levels of Pou5f1 and Mki67 on PND24, and reduced expression of Pou5f1 and Spry4 on PND45. AA exposure reduced expression of Pou5f1, Mki67, and Spry4 at PND45 although not significantly. Our results suggest that DBP acts by reducing cell proliferation and impairing differentiation in prepubertal and pubertal testes.


Subject(s)
Acrylamide/toxicity , Dibutyl Phthalate/toxicity , Environmental Pollutants/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Spermatogonia/drug effects , Testis/drug effects , Animals , Body Weight , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Gene Expression/drug effects , Male , Organ Size/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Prenatal Exposure Delayed Effects/pathology , Rats, Sprague-Dawley , Spermatogonia/pathology , Testis/embryology , Testis/growth & development , Testis/pathology
12.
Gen Comp Endocrinol ; 273: 249-259, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30391241

ABSTRACT

Spermatogenesis is a process driven by stem cell, where germ cell cycle is under the control of a specific genotype species. Considering that Jundiá (Rhamdia quelen) is a Neotropical catfish with great economical importance and useful experimental model, little information is available on basic aspects of its reproductive biology, especially on spermatogenesis. As a result, this study aimed to characterize the male germ cells, estimate the duration of spermatogenesis and evaluate the expression of selected stem cell genes in Jundiá testis. Similar to other fish species, our results showed a remarkable decrease of germ cell nuclear volume during Jundiá spermatogenesis, particularly from type A undifferentiated to late type B spermatogonia and from diplotene to late spermatids. Using a S-phase marker, bromodeoxyuridine (BrdU), the combined duration of meiotic and spermiogenic phases in this species was estimated in approximately 7 days. This is considered very short when compared to mammals, where spermatogenesis last from 30 to 74 days. Selected stem cell genes were partially sequenced and characterized in Jundiá testis. Expression analysis showed higher plzf and pou5f3 mRNA levels in the cell fractions enriched by type A undifferentiated spermatogonia. These results were further confirmed by in situ hybridization that showed strong signal of plzf and pou5f3 mRNA in type A undifferentiated spermatogonia. Altogether, these information will expand our knowledge of the reproductive biology of this species, contributing to improve its production and management, and also for biotechnological applications, such as germ cell transplantation.


Subject(s)
Biomarkers/metabolism , Catfishes/metabolism , Spermatogenesis , Spermatogonia/cytology , Stem Cells/metabolism , Tropical Climate , Animals , Catfishes/genetics , Gene Expression Regulation, Developmental , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproduction , Spermatids/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Tissue Distribution
13.
Mol Hum Reprod ; 25(3): 124-136, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30590698

ABSTRACT

Molecular mechanisms responsible for the initiation of primate spermatogenesis remain poorly characterized. Previously, 48 h stimulation of the testes of three juvenile rhesus monkeys with pulsatile LH and FSH resulted in down-regulation of a cohort of genes recognized to favor spermatogonia stem cell renewal. This change in genetic landscape occurred in concert with amplification of Sertoli cell proliferation and the commitment of undifferentiated spermatogonia to differentiate. In this report, the non-protein coding small RNA transcriptomes of the same testes were characterized using RNA sequencing: 537 mature micro-RNAs (miRNAs), 322 small nucleolar RNAs (snoRNAs) and 49 small nuclear RNAs (snRNAs) were identified. Pathway analysis of the 20 most highly expressed miRNAs suggested that these transcripts contribute to limiting the proliferation of the primate Sertoli cell during juvenile development. Gonadotrophin treatment resulted in differential expression of 35 miRNAs, 12 snoRNAs and four snRNA transcripts. Ten differentially expressed miRNAs were derived from the imprinted delta-like homolog 1-iodothyronine deiodinase 3 (DLK1-DIO3) locus that is linked to stem cell fate decisions. Four gonadotrophin-regulated expressed miRNAs were predicted to trigger a local increase in thyroid hormone activity within the juvenile testis. The latter finding leads us to predict that, in primates, a gonadotrophin-induced selective increase in testicular thyroid hormone activity, together with the established increase in androgen levels, at the onset of puberty is necessary for the normal timing of Sertoli cell maturation, and therefore initiation of spermatogenesis. Further examination of this hypothesis requires that peripubertal changes in thyroid hormone activity of the testis of a representative higher primate be determined empirically.


Subject(s)
MicroRNAs/metabolism , Testis/metabolism , Thyroid Hormones/metabolism , Animals , Follicle Stimulating Hormone/metabolism , Luteinizing Hormone/metabolism , Macaca mulatta , Male , MicroRNAs/genetics , Sequence Analysis, RNA , Signal Transduction/genetics , Signal Transduction/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology , Transcriptome/genetics
14.
Reprod Domest Anim ; 52 Suppl 2: 170-176, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27774720

ABSTRACT

Spermatogenesis is a process in which differentiated cells are produced and the adult stem cell population-known as spermatogonial stem cells (SSCs)-is continuously replenished. However, the molecular mechanisms underlying these processes are not fully understood in the canine species. We addressed this in this study by analysing the expression of specific markers in spermatogonia of seminiferous tubules of canine testes. SSCs at different stages of reproductive development (prepubertal and adult) were examined by immunohistochemistry and flow cytometry. Glial cell-derived neurotrophic factor family receptor alpha-1 (GFRA1), deleted in azoospermia-like (DAZL) and promyelocytic leukaemia zinc finger (PLZF) were expressed in SSCs, while stimulated by retinoic acid gene 8 (STRA8) was detected only in undifferentiated spermatogonia in prepubertal testis and differentiated spermatogonia and spermatocytes in adult canine. Octamer-binding transcription factor 4 (OCT4) showed an expression pattern, and the levels did not differ between the groups examined. However, C-kit expression varied as a function of reproductive developmental stage. Our results demonstrate that these proteins play critical roles in the self-renewal and differentiation of SSCs and can serve as markers to identify canine spermatogonia at specific stages of development.


Subject(s)
Dogs/physiology , Proteins/analysis , Spermatogenesis/physiology , Spermatogonia/chemistry , Adult Germline Stem Cells/chemistry , Animals , Biomarkers/analysis , Deleted in Azoospermia 1 Protein , Flow Cytometry/veterinary , Glial Cell Line-Derived Neurotrophic Factor Receptors/analysis , Immunohistochemistry/veterinary , Kruppel-Like Transcription Factors/analysis , Male , RNA-Binding Proteins/analysis , Seminiferous Tubules/cytology , Sexual Maturation , Spermatogonia/growth & development
15.
Anat Rec (Hoboken) ; 300(3): 589-599, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27770506

ABSTRACT

Prochilodus lineatus is an important representative of the order Characiformes and a species that offers great advantages to fish farming. Therefore, detailed knowledge of its reproductive biology can be applied to various fields of production and biotechnology. In this study, we have identified testicular germ cells during spermatogenesis and have evaluated the volumetric proportion of the testes occupied by structures of the tubular and intertubular compartments. In addition, the individual volume of type A spermatogonia was measured and used to estimate the mean number of these cells per testis. Gonads of adult P. lineatus males were extracted and fixed. Light and transmission electron microscopy were applied to fragments of three testicular regions. Histological, stereological, and morphometric analyses were performed. The stereological data suggest that components of the tubular and intertubular compartments of the P. lineatus testes present a uniform distribution in all three regions and therefore reflect regions with similar distributions of cell types. In addition, P. lineatus testes showed ∼0.6% of type A spermatogonia, as well as a predominance of cysts of primary spermatocytes and spermatids during the reproductive phase evaluated. The results from this study provide a better understanding of the morphology and structure of the testis and of the characterization of the type A spermatogonia in P. lineatus. The nuclear diameter of germ cells also decreases significantly during spermatogenesis. The data presented herein are the first of its kind for the order Characiformes and may be useful for future biotechnology studies on fish reproduction. Anat Rec, 300:589-599, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Characiformes/anatomy & histology , Spermatogonia/cytology , Testis/anatomy & histology , Animals , Male , Microscopy, Electron, Transmission , Spermatogenesis , Testis/cytology
16.
Einstein (Säo Paulo) ; 13(4): 637-639, Oct.-Dec. 2015.
Article in Portuguese | LILACS | ID: lil-770506

ABSTRACT

ABSTRACT Spermatogonial stem cells, which exist in the testicles since birth, are progenitors cells of male gametes. These cells are critical for the process of spermatogenesis, and not able to produce mature sperm cells before puberty due to their dependency of hormonal stimuli. This characteristic of the reproductive system limits the preservation of fertility only to males who are able to produce an ejaculate. This fact puts some light on the increase in survival rates of childhood cancer over the past decades because of improvements in the diagnosis and effective treatment in pediatric cancer patients. Therefore, we highlight one of the most important challenges concerning male fertility preservation that is the toxic effect of cancer therapy on reproductive function, especially the spermatogenesis. Currently, the experimental alternative for fertility preservation of prepubertal boys is the testicular tissue cryopreservationfor, for future isolation and spermatogonial stem cells transplantation, in order to restore the spermatogenesis. We present a brief review on isolation, characterization and culture conditions for the in vitro proliferation of spermatogonial stem cells, as well as the future perspectives as an alternative for fertility preservation in prepubertal boys. The possibility of restoring male fertility constitutes a research tool with an huge potential in basic and applied science. The development of these techniques may be a hope for the future of fertility preservation in cases that no other options exist, e.g, pediatric cancer patients.


RESUMO As espermatogônias-tronco, presentes nos testículos desde o nascimento, são as células progenitoras dos gametas masculinos, e, desse modo, críticas para o processo de espermatogênese. Antes da puberdade, essas células não são capazes de produzir espermatozoides maduros, o que só ocorrerá após o estímulo hormonal. Essa característica do sistema reprodutivo limita a possibilidade de preservação da fertilidade apenas para homens capazes de produzir um ejaculado. Tal fato coloca em evidência o aumento nas taxas de sobrevivência de crianças com câncer nas últimas décadas, devido principalmente à melhora no diagnóstico e ao tratamento dos pacientes pediátricos. Dessa forma, destaca-se um dos mais importantes desafios relativos à preservação da fertilidade masculina, que é o efeito tóxico das terapias anticâncer para o sistema reprodutivo, especialmente a espermatogênese. Tendo isso em vista, a alternativa experimental atualmente estudada para a preservação da fertilidade de pacientes pré-púberes é a criopreservação de tecido testicular para futuro isolamento e transplante de espermatogônias-tronco, a fim de restabelecer a espermatogênese. Apresentamos aqui uma breve revisão sobre isolamento, caracterização e condições de cultivo para a proliferação de espermatogônias-tronco, bem como as futuras perspectivas, como alternativa para preservação da fertilidade de meninos pré-púberes. A possibilidade de restabelecer a fertilidade masculina é uma ferramenta de pesquisa com potencial enorme de uso na pesquisa básica e aplicada. O desenvolvimento dessas técnicas pode fornecer uma esperança futura de preservação de fertilidade nos casos em que não há nenhuma outra opção, como para os pacientes pediátricos de câncer.


Subject(s)
Child , Humans , Male , Adult Stem Cells/transplantation , Fertility Preservation/methods , Infertility, Male/therapy , Stem Cell Transplantation , Biomarkers , Cryopreservation/methods , Puberty , Primary Cell Culture/methods , Stem Cell Transplantation/trends
17.
Anim Reprod Sci ; 142(1-2): 63-70, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24100067

ABSTRACT

The feral pig (Sus scrofa sp) also known as Monteiro pig, originated from a domestic pig breed that was introduced into Pantanal region in Brazil in the eighteenth century. Although the feral pig has commercial potential, there are few reports in the literature concerning the reproductive biology of this species. Therefore, the aim of this study was to further describe the feral pig testis parenchyma as well as characterize the stages of the seminiferous epithelium cycle by tubular morphology method, and to evaluate the number of differentiated spermatogonia generations in this species. Eight sexually mature feral pigs were analyzed. Fragments of testes were embedded in plastic resin and used to prepare slides for morphometrical studies. It was concluded that the feral pig has six generations of differentiated spermatogonials (A1, A2, A3, A4, In, B) and that the cellular composition in the eight stages of the seminiferous epithelium cycle of these animals were very similar to those reported in species of suidae and tayssuidae already studied.


Subject(s)
Spermatogenesis/physiology , Swine/anatomy & histology , Swine/physiology , Testis/anatomy & histology , Testis/physiology , Animals , Brazil , Epithelium/physiology , Male
18.
Anim. Reprod. (Online) ; 7(3): 187-196, July/September 2010. ilus
Article in English | VETINDEX | ID: biblio-1461637

ABSTRACT

The sexual plasticity of fish gonads declines after the sex-differentiation period; however, the plasticity of the germ cells themselves after this stage remains poorly understood. We characterized the sexual plasticity of gonial germ cells by transplanting them into sexually undifferentiated embryonic gonads in rainbow trout (Oncorhynchus mykiss). Spermatogonia or oogonia isolated from the meiotic gonads of vasa-green fluorescent protein (Gfp) gene transgenic trout were transplanted into the peritoneal cavity of newly hatched embryos of both sexes, and the behavior of the GFPlabeled donor cells was observed. The transplanted spermatogonia and oogonia migrated towards the recipient gonadal anlagen, and were subsequently incorporated into them. We also confirmed that the donor-derived gonial germ cells resumed gametogenesis in the recipient somatic microenvironment synchronously with the endogenous germ cells. Surprisingly, the donor-derived spermatogonia started to proliferate and differentiate into oocytes in female recipients. At 2 years post-transplantation, the eggs from mature female recipients were artificially inseminated with sperm from intact male rainbow trout. Normal, live offspring with the donor-derived haplotype were obtained. In addition, oogonia-derived sperm were produced in the male recipients. These donor-derived sperm were shown to be fully functional, as live offspring carrying GFP-labeled germ cells with the donor haplotype were obtained in the first filial (F1) generation. These findings indicate that rainbow trout pre-meiotic germ cells, which are likely to be spermatogonial or oogonial stem cells, possess a high level of sexual plasticity, and that the sexual differentiation of germ cells is controlled solely by the somatic microenvironment, rather than being cell autonomous.


Subject(s)
Animals , Sex Differentiation/physiology , Spermatogonia/growth & development , Oogonia/growth & development , Cell Transplantation/methods , Cell Transplantation/veterinary , Oncorhynchus mykiss/growth & development , Transplantation, Heterologous/adverse effects
19.
Anim. Reprod. ; 7(3): 187-196, July/September 2010. ilus
Article in English | VETINDEX | ID: vti-5921

ABSTRACT

The sexual plasticity of fish gonads declines after the sex-differentiation period; however, the plasticity of the germ cells themselves after this stage remains poorly understood. We characterized the sexual plasticity of gonial germ cells by transplanting them into sexually undifferentiated embryonic gonads in rainbow trout (Oncorhynchus mykiss). Spermatogonia or oogonia isolated from the meiotic gonads of vasa-green fluorescent protein (Gfp) gene transgenic trout were transplanted into the peritoneal cavity of newly hatched embryos of both sexes, and the behavior of the GFPlabeled donor cells was observed. The transplanted spermatogonia and oogonia migrated towards the recipient gonadal anlagen, and were subsequently incorporated into them. We also confirmed that the donor-derived gonial germ cells resumed gametogenesis in the recipient somatic microenvironment synchronously with the endogenous germ cells. Surprisingly, the donor-derived spermatogonia started to proliferate and differentiate into oocytes in female recipients. At 2 years post-transplantation, the eggs from mature female recipients were artificially inseminated with sperm from intact male rainbow trout. Normal, live offspring with the donor-derived haplotype were obtained. In addition, oogonia-derived sperm were produced in the male recipients. These donor-derived sperm were shown to be fully functional, as live offspring carrying GFP-labeled germ cells with the donor haplotype were obtained in the first filial (F1) generation. These findings indicate that rainbow trout pre-meiotic germ cells, which are likely to be spermatogonial or oogonial stem cells, possess a high level of sexual plasticity, and that the sexual differentiation of germ cells is controlled solely by the somatic microenvironment, rather than being cell autonomous.(AU)


Subject(s)
Animals , Sex Differentiation/physiology , Cell Transplantation/methods , Cell Transplantation/veterinary , Oogonia/growth & development , Spermatogonia/growth & development , Oncorhynchus mykiss/growth & development , Transplantation, Heterologous/adverse effects
20.
Int. braz. j. urol ; 34(3): 319-328, May-June 2008. graf, tab
Article in English | LILACS | ID: lil-489591

ABSTRACT

INTRODUCTION: Infertility is the primary concern for boys with uni- or bilateral undescended testes. An early and seemingly successful orchiopexy does not improve fertility in a substantial number of cryptorchid males. We confirmed that LH-RH analogue (LH-RHa) treatment induces an increase in and maturation of the germ cells; however, it was uncertain if treatment would improve the chance of fertility later in life. MATERIALS AND METHODS: Thirty unilateral cryptorchid boys, with an average age of 3 years at the time of surgery, were included in the study. Testicular biopsy showed that they had impaired testicular maturation and were therefore at high risk for infertility. Fifteen of the 30 unilateral cryptorchid boys were treated with 10 µg LH-RHa (Buserelin) nasal spray, administered on alternate days for a period of 6 months, following orchiopexy. The control group consisted of 15 cryptorchid boys who had been treated by Schoemakers type of orchiopexy, alone. After puberty, the ejaculates of both groups were analyzed. RESULTS: All males in the untreated group were severely oligospermic, with 20 percent being azoospermic. In contrast, 86 percent of the treated ex-cryptorchid males had a sperm concentration within the normal range; this was significantly different from the sperm concentration found in the untreated group (p = 0.000008). CONCLUSION: For the first time, we demonstrate that infertility in cryptorchidism can be successfully corrected when suitably treated with a LH-RHa. Sperm parameters normalized following therapy in the majority of cryptorchid males who, untreated, would have remained infertile. This innovative hormonal treatment will have a profound effect on the current recommended surgical treatment of boys with undescended testes.


Subject(s)
Child , Child, Preschool , Humans , Infant , Male , Buserelin/administration & dosage , Cryptorchidism/drug therapy , Gonadotropin-Releasing Hormone/analogs & derivatives , Infertility, Male/prevention & control , Sperm Count , Administration, Intranasal , Biopsy , Cryptorchidism/complications , Cryptorchidism/surgery , Oligospermia/prevention & control , Spermatogonia , Testis/pathology , Testis/surgery , Urologic Surgical Procedures, Male
SELECTION OF CITATIONS
SEARCH DETAIL