Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 898
Filter
1.
Int. j. morphol ; 41(6): 1596-1602, dic. 2023. ilus
Article in Spanish | LILACS | ID: biblio-1528809

ABSTRACT

El ácido valproico (VPA) es un fármaco antiepiléptico teratógenico que, al ser administrado durante etapas tempranas del embarazo, puede producir alteraciones en el desarrollo embriofetal, las que se manifiestan tanto a nivel del sistema nervioso como del testículo. No obstante, se ha reportado que la administración de vitamina E (VE) podría revertir dichas alteraciones. El objetivo del presente estudio fue determinar el efecto protector de la VE a nivel testicular en fetos y ratones púberes expuestos a VPA durante la fase embrionaria de su desarrollo. Se utilizó un total de 30 ratones hembra adultas gestantes (Mus musculus) cepa BALB/c, las cuales se dividieron en 6 grupos. El estudio contempló el análisis de fetos machos a los 17,5 días post-coital (dpc) y machos juveniles a las 6 semanas post-natal. A los grupos 1 y 4 se les administró 0,3 mL de solución fisiológica (grupos control para 17,5 dpc y 6 semanas postnatal, respectivamente). A los grupos 2 y 5 se les suministró la cantidad de 600 mg/kg de VPA (grupos VPA), en tanto que a los grupos 3 y 6 se les aplicó la misma dosis de VPA complementada con 200 UI de VE (grupos VPA+VE). Se describió la histología normal y patológica del compartimento peritubular del testículo. En los grupos VPA se evidenció una degeneración de la pared peritubular, y atrofia de túbulos seminíferos, así como exfoliación de las células germinales. Por el contrario, en los grupos VPA+VE tales signos no fueron observados y la morfología presentó aspecto normal solo con algunas alteraciones focales. Estos resultados corroboran el hecho que la administración de VE contrarresta en parte, los efectos deletéreos que ocasiona el VPA.


SUMMARY: Valproic acid (VPA) is a teratogenic antiepileptic drug that, when administered during the early stages of pregnancy, can produce alterations in embryo-fetal development, which manifest both at the level of the nervous system and the testicle. However, it has been reported that the administration of vitamin E (VE) could reverse these alterations. The study aimed to determine the protective effect of VE at the testicular level in fetuses and pubertal mice exposed to VPA during the embryonic phase of their development. 30 pregnant adult female mice (Mus musculus) BALB/c strain were used, which were divided into 6 groups. The study included the analysis of male fetuses at 17.5 days post-coital (dpc) and juvenile males at 6 weeks post-natal. Groups 1 and 4 were administered 0.3 mL of physiological solution. Groups 2 and 5 were given 600 mg/kg of VPA (VPA groups), while groups 3 and 6 were given the same dose of VPA supplemented with 200 IU of VE (VPA+VE). The normal and pathological histology of the peritubular compartment of the testis was described. In the VPA groups, degeneration of the peritubular wall, and atrophy of the seminiferous tubules, as well as exfoliation of the germ cells, were evident. On the contrary, in the VPA+VE groups such signs were not observed and the morphology presented a normal appearance with only some focal alterations. These results corroborate the fact that the administration of VE partially counteracts the deleterious effects caused by VPA.


Subject(s)
Animals , Female , Pregnancy , Mice , Testis/drug effects , Vitamin E/administration & dosage , Valproic Acid/toxicity , Prenatal Exposure Delayed Effects , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Testis/cytology , Vitamin E/pharmacology , Mice, Inbred BALB C , Anticonvulsants/toxicity
2.
Sci Rep ; 11(1): 23007, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34837027

ABSTRACT

Spermatogenesis, which is a continuous process from undifferentiated spermatogonia to spermatozoa in the seminiferous tubules, declines with age. To investigate changes in spermatogenesis with aging, we reconstructed the seminiferous tubules of 12 mice aged 12 to 30 months from serial sections and examined age-related and region-specific alterations in the seminiferous epithelium and spermatogenic waves in three dimensions. The basic structure of the seminiferous tubules, including the numbers of tubules, terminating points, branching points, and total tubule length, did not change with age. Age-related alterations in spermatogenesis, primarily assessed by the formation of vacuoles in Sertoli cells, were detected in the seminiferous tubules at 12 months. The proportion of altered tubule segments with impaired spermatogenesis further increased by 24 months, but remained unchanged thereafter. Altered tubule segments were preferentially distributed in tubule areas close to the rete testis and those in the center of the testis. Spermatogenic waves became shorter in length with age. These results provide a basis for examining the decline of spermatogenesis not only with aging, but also in male infertility.


Subject(s)
Aging , Seminiferous Tubules/ultrastructure , Spermatogenesis , Testis/ultrastructure , Animals , Male , Mice , Mice, Inbred C57BL , Seminiferous Epithelium/cytology , Seminiferous Epithelium/ultrastructure , Seminiferous Tubules/cytology , Spermatogonia/cytology , Spermatogonia/ultrastructure , Testis/cytology
3.
Reproduction ; 162(2): 141-147, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34085952

ABSTRACT

Sertoli cells proliferate and construct seminiferous tubules during fetal life, then undergo differentiation and maturation in the prepubertal testes. In the adult testes, mature Sertoli cells maintain spermatogonia and support spermatogenesis during the entire lifetime. Although Sertoli-like cells have been derived from iPS cells, they tend to remain immature. To investigate whether Sertoli cells can spontaneously acquire the ability to support spermatogenesis when transferred into the adult testis, we transplanted mouse fetal testicular cells into a Sertoli-depleted adult testis. We found that donor E12.5, E14.5 and E16.5 Sertoli cells colonized adult seminiferous tubules and supported host spermatogenesis 2 months after transplantation, demonstrating that immature fetal Sertoli cells can undergo sufficient maturation in the adult testis to become functional. This technique will be useful to analyze the developmental process of Sertoli cell maturation and to investigate the potential of iPS-derived Sertoli cells to colonize, undergo maturation, and support spermatogenesis within the testis environment.


Subject(s)
Cell Differentiation , Fetus/cytology , Sertoli Cells/cytology , Sertoli Cells/transplantation , Spermatogenesis , Testis/cytology , Animals , Female , Male , Mice , Pregnancy , Seminiferous Tubules/cytology , Seminiferous Tubules/physiology , Sexual Maturation , Testis/physiology
4.
Biomed Pharmacother ; 139: 111514, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33951576

ABSTRACT

Male immune infertility is a kind of disease that damages family life and happiness. The development of novel methods treating male immune infertility is of great significance. This study aimed to investigate the therapeutic effects of Chinese medicine Xiaokang Liuwei Dihuang decoction on immune infertility of male rats and explored the involved mechanisms. Model rats were established by lipopolysaccharide (LPS) injection. Anti-sperm antibody (AsAb) was detected by ELISA assay and testicular cell apoptosis was evaluated by TUNEL staining to verify the successful model establishment and screen suitable doses of Xiaokang Liuwei Dihuang decoction. Thirty rats were then divided into five groups (n = 6 per group): Control, LPS, Xiaokang Liuwei Dihuang decoction (15.12 g/kg, 30.24 g/kg and 45.36 g/kg). Results of HE staining showed that compared with LPS group, Xiaokang Liuwei Dihuang decoction treatments gradually improved the morphology of seminiferous tubules and elevated the number of spermatogenic cells as the doses increased. The sperm number and the levels of testosterone, luteinizing hormone (LH) and follicle stimulating hormone (FSH) in the serum of 15.12 g/kg, 30.24 g/kg and 45.36 g/kg Xiaokang Liuwei Dihuang decoction groups were much higher than those in LPS group. Results of TUNEL staining, ELISA assay and western blot showed that compared with LPS group, the testicular cell apoptosis and the levels of interleukin 1ß (IL-1ß), tumor necrosis factor α (TNF-α), AsAb, malondialdehyde (MDA) and toll-like receptor 2 (TLR2) in the testicular tissue significantly decreased in three Xiaokang Liuwei Dihuang decoction groups. Compared with LPS group, Bax expression in the 30.24 g/kg and 45.36 g/kg Xiaokang Liuwei Dihuang decoction groups was significantly down-regulated as well. In conclusion, Xiaokang Liuwei Dihuang decoction might ameliorate the immune infertility of male rats induced by LPS through regulating the levels of sex hormones, reactive oxygen species, pro-apoptotic and immune factors.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Drugs, Chinese Herbal/therapeutic use , Gonadal Steroid Hormones/metabolism , Infertility, Male/drug therapy , Infertility, Male/immunology , Reactive Oxygen Species/metabolism , Animals , Autoantibodies/analysis , Immunologic Factors/metabolism , Infertility, Male/chemically induced , Lipopolysaccharides , Male , Rats , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Seminiferous Tubules/metabolism , Sperm Count , Spermatogenesis/drug effects , Spermatozoa/immunology , Testis/cytology , Testis/drug effects
5.
Molecules ; 26(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802807

ABSTRACT

Infertility is a potential side effect of radiotherapy and significantly affects the quality of life for adolescent cancer survivors. Very few studies have addressed in pubertal models the mechanistic events that could be targeted to provide protection from gonadotoxicity and data on potential radioprotective treatments in this peculiar period of life are elusive. In this study, we utilized an in vitro model of the mouse pubertal testis to investigate the efficacy of crocetin to counteract ionizing radiation (IR)-induced injury and potential underlying mechanisms. Present experiments provide evidence that exposure of testis fragments from pubertal mice to 2 Gy X-rays induced extensive structural and cellular damage associated with overexpression of PARP1, PCNA, SOD2 and HuR and decreased levels of SIRT1 and catalase. A twenty-four hr exposure to 50 µM crocetin pre- and post-IR significantly reduced testis injury and modulated the response to DNA damage and oxidative stress. Nevertheless, crocetin treatment did not counteract the radiation-induced changes in the expression of SIRT1, p62 and LC3II. These results increase the knowledge of mechanisms underlying radiation damage in pubertal testis and establish the use of crocetin as a fertoprotective agent against IR deleterious effects in pubertal period.


Subject(s)
Carotenoids/pharmacology , Fertility/drug effects , Puberty/drug effects , Radiation Injuries/drug therapy , Testis/drug effects , Vitamin A/analogs & derivatives , Animals , Autophagy/drug effects , Autophagy/radiation effects , Carotenoids/therapeutic use , Catalase/metabolism , Cells, Cultured , Down-Regulation , ELAV-Like Protein 1/metabolism , Fertility/radiation effects , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/radiation effects , Immunohistochemistry , In Vitro Techniques , Male , Mice , Microtubule-Associated Proteins/metabolism , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Puberty/radiation effects , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Seminiferous Tubules/radiation effects , Sirtuin 1/metabolism , Superoxide Dismutase/metabolism , Testis/radiation effects , Up-Regulation , Vitamin A/pharmacology , Vitamin A/therapeutic use , X-Rays
6.
J Reprod Immunol ; 145: 103325, 2021 06.
Article in English | MEDLINE | ID: mdl-33930667

ABSTRACT

Contraceptive vaccine (CV) is a valuable, non-invasive, and alternative method for purposeful contraception. Sperm antigens are useful targets for producing CVs due to their specialized expression in sperm. In this study, a recombinant protein containing three main sperm epitopes (IZUMO1, SACA3, and PH-20) was designed and evaluated as CV to control fertility in male mice. The chimeric recombinant protein was expressed and purified in E. coli. Male mice were immunized by 100 µg purified protein and sera were collected to assess IgG antibodies. Evaluating the reproductive performance, immunized male mice mated with normal-fertile female mice and mating rate and the number of newborns was studied. Immunized mice were sacrificed and necropsy and histopathology studies were conducted. The results revealed that the designed chimeric protein stimulated the immune system of the mice effectively. The level of IgG antibody was significantly higher in vaccinated mouse rather than control mouse. Eighty percent of the vaccinated mice became infertile and in the remaining ones, the number of children decreased to 4-6 offspring instead of 10-12 in normal mice. Histopathological studies showed that no organs including heart, brain, lung, liver, kidney and intestine were damaged. However, Normal spermatogenesis has been disrupted and necrotic spermatogonia cells were reported in Seminiferous tubules. We concluded that the designed chimeric protein containing IZUMO1, SACA3, and PH-20 epitopes can stimulate the immune system and cause male contraception without any side effects.


Subject(s)
Contraception, Immunologic/methods , Infertility, Male/immunology , Recombinant Fusion Proteins/immunology , Vaccines, Contraceptive/immunology , Animals , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Disease Models, Animal , Epitopes/administration & dosage , Epitopes/genetics , Epitopes/immunology , Humans , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/immunology , Immunoglobulins/administration & dosage , Immunoglobulins/genetics , Immunoglobulins/immunology , Infertility, Male/pathology , Isoantigens/administration & dosage , Isoantigens/genetics , Isoantigens/immunology , Male , Membrane Proteins/administration & dosage , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Seminal Plasma Proteins/administration & dosage , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/immunology , Seminiferous Tubules/cytology , Seminiferous Tubules/immunology , Seminiferous Tubules/pathology , Spermatogonia/immunology , Spermatogonia/pathology , Vaccines, Contraceptive/administration & dosage , Vaccines, Contraceptive/genetics
7.
Gene ; 783: 145553, 2021 May 30.
Article in English | MEDLINE | ID: mdl-33711407

ABSTRACT

Despite other tissues, the effect of different exercise training protocols (ETPs) on the expression levels of metabolic substrates transmembrane transporters in the testicular tissue, remains completely unexplored. Thus, the effects of low, moderate and high-intensity ETPs on the SCs and germ cells potentials in GLUT-1, GLUT-3 and MCT-4 expression levels was investigated in this study. The animals were assigned into 4 groups, including sedentary control, low-intensity continuous (LICT), moderate-intensity (MICT) and high-intensity (HICT) ETPs-induced groups (n = 6/group). The GLUT-1, GLUT-3 and MCT-4 expressions, cytoplasmic carbohydrate storages of SCs and germ cells, the SCs survival and the spermatogenesis and spermiogenesis rates were assessed. The LICT and MICT did not significantly alter the protein expression levels of GLUT-3 and MCT-4 in the SCs and germ cells, while decreased the GLUT-1 protein content versus the sedentary control animals. In contrast, the HICT remarkably suppressed the GLUT-1 and MCT-4 in both SCs, and germ cells and diminished GLUT-3 in SCs and increased in the germ cells. No significant changes were revealed in the cytoplasmic carbohydrate storage in the LICT and MICT groups, while significantly diminished in the HICT group. The HICT group showed a failed spermatogenesis and spermiogenesis, which were not demonstrated in the sedentary control, LICT and MICT groups. In conclusion, the HICT, by reducing the GLUT-1, GLUT-3 and MCT-4 protein contents in the SCs and reducing the SCs survival, can suppress the glucose transmembrane transport and inhibit the lactate export from SCs, which in turn, ends with failed spermatogenesis and spermiogenesis.


Subject(s)
Monocarboxylic Acid Transporters/biosynthesis , Muscle Proteins/biosynthesis , Physical Conditioning, Animal , Sertoli Cells/metabolism , Spermatozoa/metabolism , Animals , Carbohydrate Metabolism , Excitatory Amino Acid Transporter 1 , Glucose Transporter Type 3 , Male , Metabolic Networks and Pathways , RNA, Messenger/metabolism , Rats, Wistar , Seminiferous Tubules/cytology , Sperm Count
8.
J Toxicol Sci ; 46(3): 115-123, 2021.
Article in English | MEDLINE | ID: mdl-33642517

ABSTRACT

The importance of glucose is well known as an energy source in testes. In order to evaluate the effects of long-lasting hypoglycemia on testes, a novel glucokinase activator, TMG-123, was dosed to rats at 5, 20 and 100 mg/kg for 13 weeks. As a result, plasma glucose levels decreased for several hours with increasing doses over the dose range of 5 to 100 mg/kg. No toxicological findings attributable to the test article were observed in clinical observation, measurements of body weight and food consumption, necropsy, and organ weight measurement. Histopathology showed scattered degeneration of seminiferous tubules in testes, and exfoliation of germ cells related to the degeneration of seminiferous tubules was observed in the lumen of both epididymides in the same animals at the end of the dosing period. Similar histopathological findings were noted at the end of the recovery period. In addition, a fertility study was conducted at the same doses for 13 weeks for males and 5 weeks for females. Sperm analysis showed decreases in the sperm concentration and the motility index and an increase in the incidences of sperm malformations. However, there were no abnormalities in the copulation or fertility rate. These results suggest that long-lasting hypoglycemia in rats is harmful to spermatogenesis and the testicular damage does not recover.


Subject(s)
Enzyme Activators/toxicity , Germ Cells/drug effects , Germ Cells/pathology , Glucokinase/metabolism , Glucokinase/toxicity , Hypoglycemia/chemically induced , Hypoglycemia/pathology , Hypoglycemic Agents/toxicity , Seminiferous Tubules/drug effects , Seminiferous Tubules/pathology , Spermatogenesis/drug effects , Spermatozoa/drug effects , Spermatozoa/pathology , Animals , Copulation/drug effects , Female , Fertility/drug effects , Male , Rats, Sprague-Dawley , Seminiferous Tubules/cytology
9.
Bull Math Biol ; 83(4): 33, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33594605

ABSTRACT

In vertebrates, sperm is generated in testicular tube-like structures called seminiferous tubules. The differentiation stages of spermatogenesis exhibit a dynamic spatiotemporal wavetrain pattern. There are two types of pattern-the vertical type, which is observed in mice, and the helical type, which is observed in humans. The mechanisms of this pattern difference remain little understood. In the present study, we used a three-species reaction-diffusion model to reproduce the wavetrain pattern observed in vivo. We hypothesized that the wavelength of the pattern in mice was larger than that in humans and undertook numerical simulations. We found complex patterns of helical and vertical pattern frequency, which can be understood by pattern selection using boundary conditions. From these theoretical results, we predicted that a small number of vertical patterns should be present in human seminiferous tubules. We then found vertical patterns in histological sections of human tubules, consistent with the theoretical prediction. Finally, we showed that the previously reported irregularity of the human pattern could be reproduced using two factors: a wider unstable wavenumber range and the irregular geometry of human compared with mouse seminiferous tubules. These results show that mathematical modeling is useful for understanding the pattern dynamics of seminiferous tubules in vivo.


Subject(s)
Models, Biological , Seminiferous Tubules , Animals , Cell Differentiation , Computer Simulation , Humans , Male , Mice , Seminiferous Tubules/cytology
10.
Mol Cell Endocrinol ; 525: 111179, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33515640

ABSTRACT

Peritubular stem Leydig cells (SLCs) have been identified from rat testicular seminiferous tubules. However, no stem cells for peritubular myoid cells have been reported in the adult testis so far. In the present study, we tested the hypothesis that the peritubular SLCs are multipotent and able to form either Leydig or myoid cells. Using cultured tubules, we show that in the presence of PDGFAA and luteinizing hormone, SLCs became testosterone-producing Leydig cells, while in the presence of PDGFBB and TGFB, the cells formed α-smooth muscle actin-expressing myoid cells. This multipotency was also confirmed by culture of isolated CD90+ SLCs. These results suggest that these stem cells outside the myoid layer are multipotent and give rise to either Leydig or myoid cells, depending on the inducing factors. These cells may serve as a common precursor population for maintaining homeostasis of both Leydig and myoid cell populations in the adult testis.


Subject(s)
Cell Differentiation , Cell Lineage , Leydig Cells/cytology , Seminiferous Tubules/cytology , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Leydig Cells/drug effects , Male , Platelet-Derived Growth Factor/pharmacology , Rats, Sprague-Dawley , Receptors, Platelet-Derived Growth Factor/metabolism , Thy-1 Antigens/metabolism
11.
Elife ; 102021 01 27.
Article in English | MEDLINE | ID: mdl-33502316

ABSTRACT

Spermatogenesis, the complex process of male germ cell proliferation, differentiation, and maturation, is the basis of male fertility. In the seminiferous tubules of the testes, spermatozoa are constantly generated from spermatogonial stem cells through a stereotyped sequence of mitotic and meiotic divisions. The basic physiological principles, however, that control both maturation and luminal transport of the still immotile spermatozoa within the seminiferous tubules remain poorly, if at all, defined. Here, we show that coordinated contractions of smooth muscle-like testicular peritubular cells provide the propulsive force for luminal sperm transport toward the rete testis. Using a mouse model for in vivo imaging, we describe and quantify spontaneous tubular contractions and show a causal relationship between peritubular Ca2+ waves and peristaltic transport. Moreover, we identify P2 receptor-dependent purinergic signaling pathways as physiological triggers of tubular contractions both in vitro and in vivo. When challenged with extracellular ATP, transport of luminal content inside the seminiferous tubules displays stage-dependent directionality. We thus suggest that paracrine purinergic signaling coordinates peristaltic recurrent contractions of the mouse seminiferous tubules to propel immotile spermatozoa to the rete testis.


As sperm develop in the testis, the immature cells must make their way through a maze of small tubes known as seminiferous tubules. However, at this stage, the cells do not yet move the long tails that normally allow them to 'swim'; it is therefore unclear how they are able to move through the tubules. Now, Fleck, Kenzler et al. have showed that, in mice, muscle-like cells within the walls of seminiferous tubules can create waves of contractions that push sperm along. Further experiments were then conducted on cells grown in the laboratory. This revealed that a signaling molecule called ATP orchestrates the moving process by activating a cascade of molecular events that result in contractions. Fleck, Kenzler et al. then harnessed an advanced microscopy technique to demonstrate that this mechanism occurs in living mice. Together, these results provide a better understanding of how sperm mature, which could potentially be relevant for both male infertility and birth control.


Subject(s)
Adenosine Triphosphate/metabolism , Sperm Transport , Testis/physiology , Animals , Humans , Male , Mice , Seminiferous Tubules/cytology
12.
Reprod Domest Anim ; 56(4): 629-641, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33492695

ABSTRACT

The microenvironment in the seminiferous tubules of buffalo changes with age, which affects the self-renewal and growth of spermatogonial stem cells (SSCs) and the process of spermatogenesis, but the mechanism remains to be elucidated. RNA-seq was performed to compare the transcript profiles of pre-pubertal buffalo (PUB) and adult buffalo (ADU) seminiferous tubules. In total, 17,299 genes from PUB and ADU seminiferous tubules identified through RNA-seq, among which 12,271 were expressed in PUB and ADU seminiferous tubules, 4,027 were expressed in only ADU seminiferous tubules, and 956 were expressed in only PUB seminiferous tubules. Of the 17,299 genes, we identified 13,714 genes that had significant differences in expression levels between PUB and ADU through GO enrichment analysis. Among these genes, 5,342 were significantly upregulated and possibly related to the formation or identity of the surface antigen on SSCs during self-renewal; 7,832 genes were significantly downregulated, indicating that genes in PUB seminiferous tubules do not participate in the biological processes of sperm differentiation or formation in this phase compared with those in ADU seminiferous tubules. Subsequently, through the combination with KEGG analysis, we detected enrichment in a number of genes related to the development of spermatogonial stem cells, providing a reference for study of the development mechanism of buffalo spermatogonial stem cells in the future. In conclusion, our data provide detailed information on the mRNA transcriptomes in PUB and ADU seminiferous tubules, revealing the crucial factors involved in maintaining the microenvironment and providing a reference for further in vitro cultivation of SSCs.


Subject(s)
Adult Germline Stem Cells/physiology , Buffaloes/physiology , Gene Expression Profiling/veterinary , Sexual Maturation/physiology , Animals , Gene Expression Regulation, Developmental , Male , RNA, Messenger , Seminiferous Tubules/cytology , Seminiferous Tubules/physiology
13.
J Morphol ; 282(4): 500-510, 2021 04.
Article in English | MEDLINE | ID: mdl-33459394

ABSTRACT

The elimination of the spermatid cytoplasm during spermiogenesis enables the sperm to acquire a streamlined architecture, which allows for unhindered swimming. While this process has been well described in vertebrates, it has rarely been reported in invertebrates. In this study, we observed the process of cytoplasm elimination during spermiogenesis in Octopus tankahkeei (Mollusca, Cephalopoda) using light microscopy, transmission electron microscopy, and immunofluorescence. In the early spermatid, the cell is circular, and the nucleus is centrally located. With spermatid development, the cell becomes polarized. The nucleus gradually elongates and moves toward the end of the cell where the tail is forming. As a result, the cytoplasm moves past the nucleus at the anterior region of the future sperm head (the foreside of the acrosome). Following this, during the late stage of spermiogenesis, the cytoplasm condenses and collects on the foreside of the acrosome until finally the residual body is discarded from the top of the sperm head. This represents a distinct directionality for the development of cytoplasmic polarity and discarding of residual body compared with that reported for vertebrates (in which the cytoplasm of the elongating spermatids is polarized toward the caudal region). The fact that the cytoplasm also becomes concentrated suggests that water pumps may be involved in the elimination of water from the cytoplasm before the residual body is discarded. Furthermore, we found that microtubules, forming a manchette-like structure, are involved not only in reshaping of the nucleus but also in the transport of mitochondria and vesicles to the foreside of the acrosome, subsequently allowing them to be discarded with the residual body. This study broadens our understanding of the development of polarization and elimination of cytoplasm from spermatids in animals.


Subject(s)
Cytoplasm/metabolism , Octopodiformes/physiology , Spermatids/growth & development , Spermatogenesis , Animals , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Male , Microtubules/metabolism , Microtubules/ultrastructure , Octopodiformes/ultrastructure , Seminiferous Tubules/cytology , Spermatids/cytology , Spermatids/ultrastructure , Spermatozoa/cytology , Spermatozoa/ultrastructure , Testis/cytology
14.
Chemosphere ; 262: 127855, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32799149

ABSTRACT

Studies have shown that phthalates are capable of affecting the development and functions of male reproductive system. The effect of phthalates on Leydig cell functions is well documented. However, little is known about their potential effects on the functions of stem Leydig cells (SLC). In the present study, we have examined the effects of mono-(2-ethylhexyl) phthalate (MEHP) on SLC functions in vitro by culturing seminiferous tubules and isolated SLCs. The results indicate that MEHP can significantly inhibit the proliferation and differentiation of SLCs in both the organ and cell culture systems. Interestingly, the minimal effective concentration that is able to affect SLC function was lower in the tubule culture system (1 µM) than in the isolated cells (10 µM), suggesting a possible involvement of the niche cells. Also, MEHP appeared to affect both the efficiency of SLCs to form Leydig cells and a selected group of Leydig cell-specific genes, including Lhcgr, Scarb1, Hsd3b1, Cyp17a1, Star, Srd5a1, Akr1c14, Insl3, Hao2 and Pah. Since SLCs are multipotent, we also tested the effect of MEHP on the differentiation of SLCs to adipocytes. Though MEHP by itself can not specify SLCs into adipocyte lineage, it indeed significantly increased the adipogenic activity of SLCs if used with an adipocyte inducing medium by up-regulation of multiple adipogenic-related genes, including Pparg and Cebpa. Overall, the results indicate that MEHP inhibits SLCs differentiating into Leydig lineage while stimulates the differentiating potential of SLCs to adipocytes.


Subject(s)
Leydig Cells/drug effects , Phthalic Acids/toxicity , Adipocytes , Animals , Cell Differentiation/drug effects , Diethylhexyl Phthalate/pharmacology , Male , Seminiferous Tubules/cytology , Steroid 17-alpha-Hydroxylase , Testosterone/pharmacology
15.
Gene ; 764: 145080, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-32858178

ABSTRACT

Spermatocyte (spc) formation from spermatogonia (spg) differentiation is the first step of spermatogenesis which produces prodigious spermatozoa for a lifetime. After decades of studies, several factors involved in the functioning of a mouse were discovered both inside and outside spg. Considering the peculiar expression and working pattern of each factor, this review divides the whole conversion of spg to spc into four consecutive development processes with a focus on extracellular cues and downstream transcription network in each one. Potential coordination among Dmrt1, Sohlh1/2 and BMP families mediates Ngn3 upregulation, which marks progenitor spg, with other changes. After that, retinoic acid (RA), as a master regulator, promotes A1 spg formation with its helpers and Sall4. A1-to-B spg transition is under the control of Kitl and impulsive RA signaling together with early and late transcription factors Stra8 and Dmrt6. Finally, RA and its responsive effectors conduct the entry into meiosis. The systematic transcription network from outside to inside still needs research to supplement or settle the controversials in each process. As a step further ahead, this review provides possible drug targets for infertility therapy by cross-linking humans and mouse model.


Subject(s)
Gene Expression Regulation, Developmental , Spermatocytes/physiology , Spermatogenesis/genetics , Spermatogonia/physiology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Self Renewal/genetics , Humans , Male , Mice , Seminiferous Tubules/cytology , Seminiferous Tubules/growth & development , Transcription Factors/metabolism , Transcription, Genetic , Tretinoin/metabolism , Up-Regulation
16.
J Vis Exp ; (164)2020 10 07.
Article in English | MEDLINE | ID: mdl-33104058

ABSTRACT

Spermatogenesis is a unique differentiation process that ultimately gives rise to one of the most distinct cell types of the body, the sperm. Differentiation of germ cells takes place in the cytoplasmic pockets of somatic Sertoli cells that host 4 to 5 generations of germ cells simultaneously and coordinate and synchronize their development. Therefore, the composition of germ cell types within a cross-section is constant, and these cell associations are also known as stages (I-XII) of the seminiferous epithelial cycle. Importantly, stages can also be identified from intact seminiferous tubules based on their differential light absorption/scatter characteristics revealed by transillumination, and the fact that the stages follow each other along the tubule in a numerical order. This article describes a transillumination-assisted microdissection method for the isolation of seminiferous tubule segments representing specific stages of mouse seminiferous epithelial cycle. The light absorption pattern of seminiferous tubules is first inspected under a dissection microscope, and then tubule segments representing specific stages are cut and used for downstream applications. Here we describe immunostaining protocols for stage-specific squash preparations and for intact tubule segments. This method allows a researcher to focus on biological events taking place at specific phases of spermatogenesis, thus providing a unique tool for developmental, toxicological, and cytological studies of spermatogenesis and underlying molecular mechanisms.


Subject(s)
Epithelial Cells/cytology , Seminiferous Tubules/cytology , Staining and Labeling , Transillumination , Acrosome/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Nucleus/metabolism , Macrophages/metabolism , Male , Mice , Microdissection , Sertoli Cells/cytology , Spermatogenesis , Spermatozoa/cytology
17.
Tissue Cell ; 67: 101450, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33091765

ABSTRACT

It has been established that excess germ cells in normal and in pathological conditions are removed from testicular tissue by the mechanism of apoptosis. Studies on germ cell apoptosis in avian species are grossly lacking, and there are only a few reports on induced germ cell degenerations in the testis tissue of birds. This study was designed to investigate the process of apoptosis of germ cells in the Japanese quail (Coturnix coturnix japonica). Germ cell degenerations were investigated in birds of all age groups, namely pre-pubertal, pubertal, adult, and aged. Apoptosis of germ cells in the quails, as shown by hematoxylin & eosin (H&E), TdT dUTP Nick End Labeling (TUNEL) assay and electron microscopy, was similar to that observed in previous studies of germ cells and somatic cells of mammalian species. The observed morphological features of these apoptotic cells ranged from irregular plasma and nuclear membranes in the early stage of apoptosis to rupture of the nuclear membrane, condensation of nuclear material, as well as fragments of apoptotic bodies, in later stages of apoptosis. In the TUNEL-positive cell counts, there was a significant difference between the mean cell counts for the four age groups (P < 0.05). Post hoc analysis revealed a highly significant difference in the aged group relative to the pubertal and adult age groups, while the cell counts of the pre-pubertal group were significantly higher than those of the pubertal group. However, there was no significant difference between cell counts of the pre-pubertal and the adult, and between the pre-pubertal and the aged groups.


Subject(s)
Apoptosis , Coturnix/physiology , Germ Cells/cytology , Testis/cytology , Aging/physiology , Animals , Germ Cells/ultrastructure , In Situ Nick-End Labeling , Male , Seminiferous Tubules/cytology , Seminiferous Tubules/ultrastructure
18.
Anim Reprod Sci ; 219: 106534, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32828409

ABSTRACT

There are age-related changes in testicular anatomy and physiology whereby there are modifications of sperm production and reproductive hormone functions. Effects of age on testicular microanatomy are well documented in humans, while there is limited understanding of these changes in dogs. The aim of this study was to evaluate age-related changes of seminiferous tubule morphology, interstitial fibrosis and spermatogenesis in dogs. Dogs (n = 32) were divided into four age groups: peripubertal (n = eight), relatively younger (n = seven), reproductively mature (n = seven) and relatively older (n = ten). Picrosirius Red stained sections were used for morphometrical analysis of testicular tissues, while the characteristics of seminiferous epithelium were assessed using a modified Johnsen scoring system for haematoxylin and eosin stained sections. Seminiferous epithelium and seminiferous tubule area increased from peripuberty to reproductive maturity, indicating there were changes during sexual maturation and subsequently there were decreases with further aging. There was a similar age-related trend for changes in seminiferous epithelium height with values being greatest in reproductively mature dogs; while there were no age-related differences in tubular diameter. Collagen content in the testicular interstitium gradually decreased from peripuberty to the age when dogs were reproductively mature and there were subsequent increases in relatively older dogs, thus, there was an association between the extent of testicular fibrosis and senescence. There was a decrease in spermatogenetic functions from relatively younger to older ages. Further investigations are warranted to establish mechanisms responsible for age-related changes of testicular morphology and related clinical implications.


Subject(s)
Aging/physiology , Dogs , Seminiferous Tubules/cytology , Spermatogenesis/physiology , Testicular Diseases/pathology , Age Factors , Animals , Cell Shape , Dog Diseases/pathology , Fibrosis/pathology , Fibrosis/veterinary , Male , Seminiferous Epithelium/pathology , Seminiferous Epithelium/ultrastructure , Seminiferous Tubules/pathology , Seminiferous Tubules/ultrastructure , Sexual Maturation/physiology
19.
Reproduction ; 160(2): 259-268, 2020 08.
Article in English | MEDLINE | ID: mdl-32449695

ABSTRACT

Testicular peritubular cells (TPCs) are smooth muscle-like cells, which form a compartment surrounding the seminiferous tubules. Previous studies employing isolated human testicular peritubular cells (HTPCs) indicated that their roles in the testis go beyond sperm transport and include paracrine and immunological contributions. Peritubular cells from a non-human primate (MKTPCs), the common marmoset monkey, Callithrix jacchus, share a high degree of homology with HTPCs. However, like their human counterparts these cells age in vitro and replicative senescence limits in-depth functional or mechanistic studies. Therefore, a stable cellular model was established. MKTPCs of a young adult animal were immortalized by piggyBac transposition of human telomerase (hTERT), that is, without the expression of viral oncogenes. Immortalized MKTPCs (iMKTPCs) grew without discernable changes for more than 50 passages. An initial characterization revealed typical genes expressed by peritubular cells (androgen receptor (AR), smooth-muscle actin (ACTA2), calponin (CNN1)). A proteome analysis of the primary MKTPCs and the derived immortalized cell line confirmed that the cells almost completely retained their phenotype. To test whether they respond in a similar way as HTPCs, iMKTPCs were challenged with forskolin (FSK) and ATP. As HTPCs, they showed increased expression level of the StAR protein (StAR) after FSK stimulation, indicating steroidogenic capacity. ATP increased the expression of pro-inflammatory factors (e.g. IL1B; CCL7), as it is the case in HTPCs. Finally, we confirmed that iMKTPCs can efficiently be transfected. Therefore, they represent a highly relevant translational model, which allows mechanistic studies for further exploration of the roles of testicular peritubular cells.


Subject(s)
Cellular Senescence , Models, Animal , Phosphoproteins/metabolism , Proteome/metabolism , Seminiferous Tubules/metabolism , Testis/metabolism , Animals , Callithrix , Male , Proteome/analysis , Seminiferous Tubules/cytology , Testis/cytology
20.
Cell Rep ; 31(2): 107513, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32294451

ABSTRACT

Sertoli cells are supporting cells of the testicular seminiferous tubules, which provide a nurturing environment for spermatogenesis. Adult Sertoli cells are polarized so that they can simultaneously support earlier-stage spermatogenic cells (e.g., spermatogonia) basally and later-stage cells (e.g., spermatids) apically. To test the consequences of disrupting cell polarity in Sertoli cells, we perform a Sertoli-specific conditional deletion of Rac1, which encodes a Rho GTPase required for apicobasal cell polarity. Rac1 conditional knockout adults exhibit spermatogenic arrest at the round spermatid stage, with severe disruption of Sertoli cell polarity, and show increased germline and Sertoli cell apoptosis. Thus, Sertoli Rac1 function is critical for the progression of spermatogenesis but, surprisingly, is dispensable for fetal testicular development, adult maintenance of undifferentiated spermatogonia, and meiotic entry. Our data indicate that Sertoli Rac1 function is required only for certain aspects of spermatogenesis and reveal that there are distinct requirements for cell polarity during cellular differentiation.


Subject(s)
Neuropeptides/metabolism , Sertoli Cells/metabolism , Testis/cytology , rac1 GTP-Binding Protein/metabolism , Animals , Cell Differentiation/physiology , Cell Polarity/physiology , Male , Mice , Mice, Inbred C57BL , Neuropeptides/genetics , Seminiferous Tubules/cytology , Sertoli Cells/cytology , Sertoli Cells/pathology , Spermatids/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology , Spermatogonia/physiology , Testis/growth & development , rac1 GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...