Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 402
Filter
1.
Curr Protoc ; 3(11): e920, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37933593

ABSTRACT

Human fertility is declining in Western countries, and it is becoming increasingly clear that male infertility plays a pivotal role in the overall fertility decline. To understand the process that drives successful male germ cell maturation, the study of spermatogenesis of model organisms, such as mice, is essential. Residual bodies (RBs) play an important role in the last stages of spermatogenesis. They are formed at the time when post-meiotic spermatids undergo sequential differentiation steps so that the acrosome and flagellum are developed, the nucleus is markedly condensed, and the cytoplasm is lost. The masses of lost cytoplasm become RBs. Our recent work has shown that RB dynamics are highly sensitive to even small fertility defects. It was also noted that the transcriptome and proteome of RBs changes in response to spermatogenic defects. Thus, RBs represent an excellent and highly sensitive entity for studying male fertility. Previously published protocols for RB purification had some major limitations: they produced an RB fraction that was heavily contaminated with spermatozoa and erythrocytes or required tens of grams of starting material. In addition, most of the available protocols were developed for purification of RBs from rat testes. Here, we present a protocol that allows the isolation of 2.5-3 × 106 RBs from mouse testes with a purity of 98% from only 1 g of starting material. The purified material can be used for various downstream applications to study male fertility, such as transcriptome and proteome analyses, super-resolution microscopy, and electron and cryo-electron microscopy, amongst many others. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: An improved method for purification of the residual bodies from the seminiferous tubules of mice.


Subject(s)
Proteome , Seminiferous Tubules , Rats , Mice , Male , Animals , Humans , Cryoelectron Microscopy , Seminiferous Tubules/physiology , Spermatozoa , Spermatids
2.
Reproduction ; 164(1): 9-18, 2022 05 23.
Article in English | MEDLINE | ID: mdl-35521906

ABSTRACT

Spermatozoa released from Sertoli cells must be transported to the epididymis. However, the mechanism of the luminal flow in seminiferous tubules has remained unclear to date. Therefore, in this study, we investigated luminal flow and movements in the seminiferous tubules by three-dimensional analysis and in vivo imaging. Serial 5-µm-thick mouse testicular sections at 50-µm-intervals were prepared and stained by Periodic Acid-Schiff-hematoxylin. After three-dimensional reconstruction of the seminiferous tubules, the localization of the released spermatozoa and the stages observed in the sections were recorded in each reconstructed tubule. Luminal movements in the seminiferous tubules were observed by in vivo imaging using a fluorescent-reporter mouse and two-photon excitation microscopy system. Spermatozoa without contact to the seminiferous epithelium were not accumulated toward the rete testis. Additionally, such spermatozoa were found on their way not only to the most proximal rete testis but also a more distant rete testis from any stage VIII seminiferous epithelia. In vivo imaging demonstrated that the direction of the flagella of spermatozoa attached to the seminiferous epithelium was repeatedly reversed. The epithelium at the inner curve of the seminiferous tubule was shaken more actively and had fewer spermatozoa attached compared with the epithelium at the outer curve. Our results hence suggest that the luminal flow in the seminiferous tubules is repeatedly reversed and that this physical force helps spermatozoa to be released from Sertoli cells. In brief: Spermatozoa are released from Sertoli cells and flow in the seminiferous tubule to the rete testis. Our results suggest that the luminal flow in the tubules is repeatedly reversed and that this physical force helps spermatozoa release from the Sertoli cells.


Subject(s)
Microfluidics , Seminiferous Tubules , Sertoli Cells , Spermatozoa , Animals , Imaging, Three-Dimensional , Male , Mice , Microfluidics/methods , Microscopy , Rete Testis/physiology , Rheology/methods , Seminiferous Epithelium/diagnostic imaging , Seminiferous Epithelium/physiology , Seminiferous Tubules/diagnostic imaging , Seminiferous Tubules/physiology , Sertoli Cells/physiology , Spermatozoa/physiology , Testis/diagnostic imaging , Testis/physiology
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.
Cell Tissue Res ; 385(1): 191-205, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33783608

ABSTRACT

Phospholipase D6 (PLD6) plays pivotal roles in mitochondrial dynamics and spermatogenesis, but the cellular and subcellular localization of endogenous PLD6 in testis germ cells is poorly defined. We examined the distribution and subcellular localization of PLD6 in mouse testes using validated specific anti-PLD6 antibodies. Ectopically expressed PLD6 protein was detected in the mitochondria of PLD6-transfected cells, but endogenous PLD6 expression in mouse testes was localized to the perinuclear region of pachytene spermatocytes, and more prominently, to the round (Golgi and cap phases) and elongating spermatids (acrosomal phase); these results suggest that PLD6 is localized to the Golgi apparatus. The distribution of PLD6 in the round spermatids partially overlapped with that of the cis-Golgi marker GM130, indicating that the PLD6 expression corresponded to the GM130-positive subdomains of the Golgi apparatus. Correlative light and electron microscopy revealed that PLD6 expression in developing spermatids was localized almost exclusively to several flattened cisternae, and these structures might correspond to the medial Golgi subcompartment; neither the trans-Golgi networks nor the developing acrosomal system expressed PLD6. Further, we observed that PLD6 interacted with tesmin, a testis-specific transcript necessary for successful spermatogenesis in mouse testes. To our knowledge, these results provide the first evidence of PLD6 as a Golgi-localized protein of pachytene spermatocytes and developing spermatids and suggest that its subcompartment-specific distribution within the Golgi apparatus may be related to the specific functions of this organelle during spermatogenesis.


Subject(s)
Phospholipases/metabolism , Seminiferous Tubules/physiology , Testis/physiology , Animals , Male , Mice
5.
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
6.
In Vitro Cell Dev Biol Anim ; 56(3): 261-270, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32212030

ABSTRACT

Optimization of tissue culture systems able to complete male germ cell maturation to post-meiotic stages is considered as an important matter in reproductive biology. Considering that hypoxia is one of the factors limiting the efficiency of organ culture, the aim of this study was to use isolated seminiferous tubules (STs), having more surface and less thickness, in an organotypic culture system in order to improve oxygen diffusion and reduce hypoxia. The mechanically separated STs embedded in agarose or alginate and 1-3-mm3 testicular tissue fragments of 3 adult mice were separately placed on the flat surface of agarose gel that was half-soaked in the medium. Survival and differentiation of germ cells using PLZF and SCP3 markers, identity of Sertoli cell using GATA4, cell proliferation with the Ki67 marker, and ST integrity using a ST scoring were evaluated up to 36 d at different culture times, each corresponding to the duration of one spermatogenic cycle. We observed a significantly reduced ST integrity in STs embedded in agarose or alginate on day 9 (versus tissue fragments p ≤ 0.05). There was no difference in the number of PLZF-positive cells between groups, but the number of SCP3 (in all-time points) and GATA4-positive cells was significantly higher in the culture of embedded STs. Although embedding STs can be useful for the progress of in vitro spermatogenesis, it makes them sensitive to degeneration. Further improvements are required to modify the air-liquid interface method to maintain ST integrity.


Subject(s)
Alginates/pharmacology , Organ Culture Techniques/methods , Seminiferous Tubules/physiology , Sepharose/pharmacology , Spermatogenesis , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Sertoli Cells/cytology , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Spermatogenesis/drug effects , Spermatogonia/cytology , Spermatogonia/drug effects , Spermatogonia/metabolism , Spermatozoa/cytology , Tissue Survival/drug effects
7.
Hum Reprod Update ; 26(3): 368-391, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32163572

ABSTRACT

BACKGROUND: Although the prognosis of childhood cancer survivors has increased dramatically during recent years, chemotherapy and radiation treatments for cancer and other conditions may lead to permanent infertility in prepubertal boys. Recent developments have shown that spermatogonial stem cell (SSC) transplantation may be a hope for restoring fertility in adult survivors of childhood cancers. For this reason, several centres around the world are collecting and cryopreserving testicular tissue or cells anticipating that, in the near future, some patients will return for SSC transplantation. This review summarizes the current knowledge and utility of SSC transplantation techniques. OBJECTIVE AND RATIONALE: The aim of this narrative review is to provide an overview of the currently used experimental injection techniques for SSC transplantation in animal and human testes. This is crucial in understanding and determining the role of the different techniques necessary for successful transplantation. SEARCH METHODS: A comprehensive review of peer-reviewed publications on this topic was performed using the PubMed and Google Scholar databases. The search was limited to English language work and studies between 1994 (from the first study on SSC transplantation) and April 2019. Key search terms included mouse, rat, boar, ram, dog, sheep, goat, cattle, monkey, human, cadaver, testes, SSC transplantation, injection and technique. OUTCOMES: This review provides an extensive clinical overview of the current research in the field of human SSC transplantation. Rete testis injection with ultrasonography guidance currently seems the most promising injection technique thus far; however, the ability to draw clear conclusions is limited due to long ischemia time of cadaver testis, the relatively decreased volume of the testis, the diminishing size of seminiferous tubules, a lack of intratesticular pressure and leakage into the interstitium during the injection on human cadaver testis. Current evidence does not support improved outcomes from multiple infusions through the rete testes. Overall, further optimization is required to increase the efficiency and safety of the infusion method. WIDER IMPLICATIONS: Identifying a favourable injection method for SSC transplantation will provide insight into the mechanisms of successful assisted human reproduction. Future research could focus on reducing leakage and establishing the optimal infusion cell concentrations and pressure.


Subject(s)
Adult Germline Stem Cells/transplantation , Fertility Preservation/methods , Spermatogenesis/physiology , Spermatogonia/transplantation , Stem Cell Transplantation/methods , Animals , Cattle , Child , Cryopreservation , Dogs , Humans , Male , Mice , Models, Animal , Neoplasms/therapy , Rats , Seminiferous Tubules/physiology , Sheep , Spermatogonia/cytology , Swine
8.
J Struct Biol ; 209(3): 107449, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31931124

ABSTRACT

Optical clearing techniques provide unprecedented opportunities to study large tissue samples at histological resolution, eliminating the need for physical sectioning while preserving the three-dimensional structure of intact biological systems. There is significant potential for applying optical clearing to reproductive tissues. In testicular biology, for example, the study of spermatogenesis and the use of spermatogonial stem cells offer high-impact applications in fertility medicine and reproductive biotechnology. The objective of our study is to apply optical clearing, immunofluorescence, and confocal microscopy to testicular tissue in order to reconstruct its three-dimensional microstructure in intact samples. We used Triton-X/DMSO clearing in combination with refractive index matching to achieve optical transparency of fixed mouse testes. An antibody against smooth muscle actin was used to label peritubular myoid cells of seminiferous tubules while an antibody against ubiquitin C-terminal hydrolase was used to label Sertoli cells and spermatogonia in the seminiferous epithelium. Specimens were then imaged using confocal fluorescence microscopy. We were able to successfully clear testicular tissue and utilize immunofluorescent probes. Additionally, we successfully visualized the histological compartments of testicular tissue in three-dimensional reconstructions. Optical clearing combined with immunofluorescence and confocal imaging offers a powerful new method to analyze the cytoarchitecture of testicular tissue at histological resolution while maintaining the macro-scale perspective of the intact system. Considering the importance of the murine model, our developed method represents a significant contribution to the field of male reproductive biology, enabling the study of testicular function.


Subject(s)
Imaging, Three-Dimensional , Microscopy, Confocal , Seminiferous Tubules/ultrastructure , Testis/ultrastructure , Animals , Male , Mice , Microscopy, Fluorescence/methods , Seminiferous Tubules/physiology , Sertoli Cells/physiology , Sertoli Cells/ultrastructure , Spermatogenesis , Spermatogonia/physiology , Spermatogonia/ultrastructure , Testis/physiology
9.
Theriogenology ; 142: 363-367, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31711695

ABSTRACT

The seminiferous epithelium goes through multiple changes which enables the differentiation of a spermatogonia in a fully mature spermatozoon. The timing of these changes is species-specific and influences the duration of the reproductive cycles. Bats are among wild mammals whose coordination between male and female reproductive cycles are imperative, since most females show seasonal preferences, even in the Tropics. This seasonal variation demands constant sperm production ready for spermiation in order to guarantee its genetic dispersion and reproduction success. Despite their abundance, little is known about the duration of reproductive cycles in Neotropical bat species, a relevant information for the species management and for conservational strategies regarding anthropogenic and climate influences on bats reproduction. In this study, we aimed at characterizing the stages of the seminiferous epithelium cycle (SEC) of the fruit bat Artibeus lituratus and to determine its duration based on the immunohistochemical analysis of the bromodeoxyuridine (BrDU) activity. SEC stages were characterized according to the tubular morphology method and the frequency of each stage was estimated. After intratesticular injections of BrDU, the animals were euthanized at different times, and the estimation of SEC duration was performed by observing the most advanced germ cells in the seminiferous epithelium. The most advanced stained cells after 2 days of BrdU injection were the primary spermatocytes in pachytene, transitioning from stages 1-2 of the SEC. Within 2 days, we found a progression of 30.42% of the SEC, and an entire cycle lasted 6.58 days on average. Considering that 4.5 seminiferous epithelium cycles are necessary for the whole spermatogenic processes to be completed, the total length of spermatogenesis in A. lituratus was estimated at 29.61 days. Our findings support a pattern of bimodal seasonal polyestry for this species, with rapid spermatogenic cycles.


Subject(s)
Cell Differentiation , Chiroptera/physiology , Seminiferous Epithelium/cytology , Seminiferous Epithelium/physiology , Spermatogenesis/physiology , Animals , Male , Reproduction/physiology , Seminiferous Tubules/cytology , Seminiferous Tubules/physiology , Sperm Maturation , Spermatocytes/cytology , Spermatocytes/physiology , Spermatogonia/cytology , Spermatogonia/physiology , Time Factors
10.
Microsc Microanal ; 26(1): 148-156, 2020 02.
Article in English | MEDLINE | ID: mdl-31753050

ABSTRACT

The seminiferous tubule (ST) is the location of spermatogenesis, where mature spermatozoa are produced with the assistance of Sertoli cells. The role of extracellular vesicles in the direct communication between Sertoli-germ cells in the ST is still not fully understood. In this study, we reported multivesicular bodies (MVBs) and their source of CD63-enriched exosomes by light and ultrastructure microscopy during the reproductive phases of turtles. Strong CD63 immunopositivity was detected at the basal region in the early and luminal regions of the ST during late spermatogenesis by immunohistochemistry (IHC), immunofluorescence (IF), and western blot (WB) analysis. Labeling of CD63 was detected in the Sertoli cell cytoplasmic processes that surround the developing germ cells during early spermatogenesis and in the lumen of the ST with elongated spermatids during late spermatogenesis. Furthermore, ultrastructure analysis confirmed the existence of numerous MVBs in the Sertoli cell prolongations that surround the round and primary spermatogonia during acrosome biogenesis and with the embedded heads of spermatids in the cytoplasm of Sertoli cells. Additionally, in spermatids, Chrysanthemum flower centers (CFCs) generated isolated membranes involved in MVBs and autophagosome formation, and their fusion to form amphiosomes was also observed. Additionally, autophagy inhibition by 3-methyladenine (after 24 h) increased CD63 protein signals during late spermatogenesis, as detected by IF and WB. Collectively, our study found MVBs and CD63 rich exosomes within the Sertoli cells and their response to autophagy inhibition in the ST during the spermatogenesis in the turtle.


Subject(s)
Exosomes/ultrastructure , Multivesicular Bodies/ultrastructure , Seminiferous Tubules/physiology , Seminiferous Tubules/ultrastructure , Spermatogenesis , Tetraspanin 30/analysis , Turtles/physiology , Animals , Blotting, Western , Exosomes/chemistry , Immunohistochemistry , Male , Microscopy, Electron , Microscopy, Fluorescence , Multivesicular Bodies/chemistry
11.
Andrology ; 7(6): 827-839, 2019 11.
Article in English | MEDLINE | ID: mdl-31250567

ABSTRACT

BACKGROUND: Ageing in men is believed to be associated with fertility decline and elevated risk of congenital disorders for the offspring. The previous studies also reported reduced germ and Sertoli cell numbers in older men. However, it is not clear whether ageing in men with normal spermatogenesis affects the testis and germ cell population dynamics in a way sufficient for transmitting adverse age effects to the offspring. OBJECTIVES: We examined men with normal spermatogenesis at different ages concerning effects on persisting testicular cell types, that is the germ line and Sertoli cells, as these cell populations are prone to be exposed to age effects. MATERIAL AND METHODS: Ageing was assessed in testicular biopsies of 32 patients assigned to three age groups: (i) 28.8 ± 2.7 years; (ii) 48.1 ± 1 years; and (iii) 70.9 ± 6.2 years, n = 8 each, with normal spermatogenesis according to the Bergmann-Kliesch score, and in a group of meiotic arrest patients (29.9 ± 3.8 years, n = 8) to decipher potential links between different germ cell types. Besides morphometry of seminiferous tubules and Sertoli cell nuclei, we investigated spermatogenic output/efficiency, and dynamics of spermatogonial populations via immunohistochemistry for MAGE A4, PCNA, CREM and quantified A-pale/A-dark spermatogonia. RESULTS: We found a constant spermatogenic output (CREM-positive round spermatids) in all age groups studied. In men beyond their mid-40s (group 2), we detected increased nuclear and nucleolar size in Sertoli cells, indirectly indicating an elevated protein turnover. From the 7th decade (group 3) of life onwards, testes showed increased proliferation of undifferentiated spermatogonia, decreased spermatogenic efficiency and elevated numbers of proliferating A-dark spermatogonia. DISCUSSION AND CONCLUSION: Maintaining normal sperm output seems to be an intrinsic determinant of spermatogenesis. Ageing appears to affect this output and might provoke compensatory proliferation increase in A spermatogonia which, in turn, might hamper germ cell integrity.


Subject(s)
Seminiferous Tubules/physiology , Sertoli Cells/physiology , Spermatogenesis/physiology , Spermatogonia/physiology , Spermatozoa/physiology , Adult , Aged , Aging/physiology , Congenital Abnormalities/epidemiology , Genetic Diseases, Inborn/epidemiology , Humans , Male , Middle Aged
12.
Cell Mol Life Sci ; 76(14): 2681-2695, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30980107

ABSTRACT

Spermatogenesis is fundamental to the establishment and maintenance of male reproduction, whereas its abnormality results in male infertility. Somatic cells, including Leydig cells, myoid cells, and Sertoli cells, constitute the microenvironment or the niche of testis, which is essential for regulating normal spermatogenesis. Leydig cells are an important component of the testicular stroma, while peritubular myoid cells are one of the major cell types of seminiferous tubules. Here we addressed the roles and mechanisms of Leydig cells and myoid cells in the regulation of spermatogenesis. Specifically, we summarized the biological features of Leydig cells and peritubular myoid cells, and we introduced the process of testosterone production and its major regulation. We also discussed other hormones, cytokines, growth factors, transcription factors and receptors associated with Leydig cells and myoid cells in mediating spermatogenesis. Furthermore, we highlighted the issues that are worthy of further studies in the regulation of spermatogenesis by Leydig cells and peritubular myoid cells. This review would provide novel insights into molecular mechanisms of the somatic cells in controlling spermatogenesis, and it could offer new targets for developing therapeutic approaches of male infertility.


Subject(s)
Infertility, Male/physiopathology , Leydig Cells/cytology , Seminiferous Tubules/cytology , Sertoli Cells/cytology , Spermatogenesis , Humans , Leydig Cells/physiology , Male , Seminiferous Tubules/physiology , Sertoli Cells/physiology
13.
Vet Med Sci ; 5(2): 151-156, 2019 05.
Article in English | MEDLINE | ID: mdl-30816002

ABSTRACT

Whether paired organs are equivalent has to be analyzed by assessing both structural and functional aspects. The present study compared the paired left and right testis from Ghezel sheep originating from Northwestern Iran (Azerbaijan) and Northeastern Turkey. Twenty-five pairs of testes were collected from mature Ghezel rams from Tabriz slaughterhouse. The two glands were compared for their size, weight and activity rate. Weight, length, width and thickness were measured. Then, paraffinized blocks were prepared by routine histological techniques and sections were stained by the haematoxylin-eosin method. Five 6 µm sections were prepared from each paraffinized block and 50 seminiferous tubules (STs) were analysed in each section for tubular differential index (TDI) and spermiogenesis index (SPI). TDI and SPI were compared between the paired left and right testes. Weight, length and thickness of left testes were significantly higher than in right testes (P < 0.05). Moreover, TDI and SPI were found to be higher in left testes than the right (P < 0.05).


Subject(s)
Sheep, Domestic/anatomy & histology , Spermatogenesis , Testis/anatomy & histology , Animals , Iran , Male , Organ Size , Seminiferous Tubules/physiology , Turkey
14.
Hum Reprod ; 34(3): 403-413, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30753464

ABSTRACT

STUDY QUESTION: Can full spermatogenesis be achieved after xenotransplantation of prepubertal primate testis tissue to the mouse, in testis or subcutaneously? SUMMARY ANSWER: Intratesticular xenotransplantation supported the differentiation of immature germ cells from marmoset (Callithrix jacchus) into spermatids and spermatozoa at 4 and 9 months post-transplantation, while in subcutaneous transplants, spermatogenic arrest was observed at 4 months and none of the transplants survived at 9 months. WHAT IS KNOWN ALREADY: Auto-transplantation of cryopreserved immature testis tissue (ITT) could be a potential fertility restoration strategy for patients with complete loss of germ cells due to chemo- and/or radiotherapy at a young age. Before ITT transplantation can be used for clinical application, it is a prerequisite to demonstrate the feasibility of the technique and identify the conditions required for establishing spermatogenesis in primate ITT transplants. Although xenotransplantation of ITT from several species has resulted in complete spermatogenesis, in human and marmoset, ITT has not been successful. STUDY DESIGN, SIZE, DURATION: In this study, we used marmoset as a pre-clinical animal model. ITT was obtained from two 6-month-old co-twin marmosets. A total of 147 testis tissue pieces (~0.8-1.0 mm3 each) were transplanted into the testicular parenchyma (intratesticular; n = 40) or under the dorsal skin (ectopic; n = 107) of 4-week-old immunodeficient Swiss Nu/Nu mice (n = 20). Each mouse received one single marmoset testis tissue piece in each testis and 4-6 pieces subcutaneously. Xenotransplants were retrieved at 4 and 9 months post-transplantation and evaluations were performed with regards to transplant survival, spermatogonial quantity and germ cell differentiation. PARTICIPANTS/MATERIALS, SETTING, METHODS: Transplant survival was histologically evaluated by haematoxylin-periodic acid Schiff (H/PAS) staining. Spermatogonia were identified by MAGE-A4 via immunohistochemistry. Germ cell differentiation was assessed by morphological identification of different germ cell types on H/PAS stained sections. Meiotically active germ cells were identified by BOLL expression. CREM immunohistochemistry was performed to confirm the presence of post-meiotic germ cells and ACROSIN was used to determine the presence of round, elongating and elongated spermatids. MAIN RESULTS AND THE ROLE OF CHANCE: Four months post-transplantation, 50% of the intratesticular transplants and 21% of the ectopic transplants were recovered (P = 0.019). The number of spermatogonia per tubule did not show any variation. In 33% of the recovered intratesticular transplants, complete spermatogenesis was established. Overall, 78% of the intratesticular transplants showed post-meiotic differentiation (round spermatids, elongating/elongated spermatids and spermatozoa). However, during the same period, spermatocytes (early meiotic germ cells) were the most advanced germ cell type present in the ectopic transplants. Nine months post-transplantation, 50% of the intratesticular transplants survived, whilst none of the ectopic transplants was recovered (P < 0.0001). Transplants contained more spermatogonia per tubule (P = 0.018) than at 4 months. Complete spermatogenesis was observed in all recovered transplants (100%), indicating a progressive spermatogenic development in intratesticular transplants between the two time-points. Nine months post-transplantation, transplants contained more seminiferous tubules with post-meiotic germ cells (37 vs. 5%; P < 0.001) and fewer tubules without germ cells (2 vs. 8%; P = 0.014) compared to 4 months post-transplantation. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Although xenotransplantation of marmoset ITT was successful, it does not fully reflect all aspects of a future clinical setting. Furthermore, due to ethical restrictions, we were not able to prove the functionality of the spermatozoa produced in the marmoset transplants. WIDER IMPLICATIONS OF THE FINDINGS: In this pre-clinical study, we demonstrated that testicular parenchyma provides the required microenvironment for germ cell differentiation and long-term survival of immature marmoset testis tissue, likely due to the favourable temperature regulation, growth factors and hormonal support. These results encourage the design of new experiments on human ITT xenotransplantation and show that intratesticular transplantation is likely to be superior to ectopic transplantation for fertility restoration following gonadotoxic treatment in childhood. STUDY FUNDING/COMPETING INTEREST(S): This project was funded by the ITN Marie Curie Programme 'Growsperm' (EU-FP7-PEOPLE-2013-ITN 603568) and the scientific Fund Willy Gepts from the UZ Brussel (ADSI677). D.V.S. is a post-doctoral fellow of the Fonds Wetenschappelijk Onderzoek (FWO; 12M2815N). No conflict of interest is declared.


Subject(s)
Spermatogenesis , Testis/physiology , Testis/transplantation , Animals , Callithrix , Cell Differentiation , Cryopreservation , Germ Cells/cytology , Male , Mice , Seminiferous Tubules/physiology , Sertoli Cells/physiology , Spermatids/physiology , Spermatogonia/physiology , Spermatozoa/physiology , Transplantation, Heterologous
15.
PLoS Genet ; 15(2): e1007909, 2019 02.
Article in English | MEDLINE | ID: mdl-30735494

ABSTRACT

Gonad differentiation is a crucial step conditioning the future fertility of individuals and most of the master genes involved in this process have been investigated in detail. However, transcriptomic analyses of developing gonads from different animal models have revealed that hundreds of genes present sexually dimorphic expression patterns. DMXL2 was one of these genes and its function in mammalian gonads was unknown. We therefore investigated the phenotypes of total and gonad-specific Dmxl2 knockout mouse lines. The total loss-of-function of Dmxl2 was lethal in neonates, with death occurring within 12 hours of birth. Dmxl2-knockout neonates were weak and did not feed. They also presented defects of olfactory information transmission and severe hypoglycemia, suggesting that their premature death might be due to global neuronal and/or metabolic deficiencies. Dmxl2 expression in the gonads increased after birth, during follicle formation in females and spermatogenesis in males. DMXL2 was detected in both the supporting and germinal cells of both sexes. As Dmxl2 loss-of-function was lethal, only limited investigations of the gonads of Dmxl2 KO pups were possible. They revealed no major defects at birth. The gonadal function of Dmxl2 was then assessed by conditional deletions of the gene in gonadal supporting cells, germinal cells, or both. Conditional Dmxl2 ablation in the gonads did not impair fertility in males or females. By contrast, male mice with Dmxl2 deletions, either throughout the testes or exclusively in germ cells, presented a subtle testicular phenotype during the first wave of spermatogenesis that was clearly detectable at puberty. Indeed, Dmxl2 loss-of-function throughout the testes or in germ cells only, led to sperm counts more than 60% lower than normal and defective seminiferous tubule architecture. Transcriptomic and immunohistochemichal analyses on these abnormal testes revealed a deregulation of Sertoli cell phagocytic activity related to germ cell apoptosis augmentation. In conclusion, we show that Dmxl2 exerts its principal function in the testes at the onset of puberty, although its absence does not compromise male fertility in mice.


Subject(s)
Nerve Tissue Proteins/genetics , Spermatogenesis/genetics , Spermatozoa/physiology , Animals , Apoptosis/genetics , Female , Fertility/genetics , Germ Cells/physiology , Gonads/physiology , Infertility, Female/genetics , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Seminiferous Tubules/physiology , Sertoli Cells/physiology , Testis/physiology
16.
Anat Sci Int ; 94(1): 1-10, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30128978

ABSTRACT

We developed a technique to analyze the high-resolution three-dimensional (3D) structure of seminiferous tubules. It consists of segmentation of tubules in serial paraffin sections of the testis by marking the basement membrane with periodic acid-Schiff or a fluorescent anti-laminin antibody followed by 3D reconstruction of tubules with high-performance software. Using this method, we analyzed testes from mice at different ages and accurately elucidated the 3D structure of seminiferous tubules, including the number and length of tubules as well as the numbers of connections with the rete testis, branching points, and blind ends. We also developed a technique to identify the precise spermatogenic stage and cellular composition of the seminiferous epithelium. It consists of the combination of lectin histochemistry for acrosomes and immunohistochemistry for specific cell markers visualized with fluorescence. Using this method, we examined seminiferous tubules from normal mice and counted the number of each cell type at each stage, and thereby established a quantitative standard for the cellular composition of the seminiferous epithelium. We then investigated seminiferous epithelia from genetically modified infertile mice deficient in certain cell adhesion molecules and revealed characteristic abnormalities in the cellular composition. We also analyzed the distribution and direction of spermatogenic waves along the length of adult seminiferous tubules as well as the site of the first onset of spermatogenesis in postnatal seminiferous tubules. These methods will be useful for investigating the structure and function of seminiferous tubules in mice and humans under normal and pathological conditions.


Subject(s)
Basement Membrane/anatomy & histology , Mice/anatomy & histology , Models, Anatomic , Rete Testis/anatomy & histology , Seminiferous Tubules/anatomy & histology , Animals , Male , Seminiferous Tubules/physiology , Spermatogenesis
17.
J Reprod Dev ; 65(1): 37-46, 2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30416150

ABSTRACT

Polypyrimidine tract-binding protein 1 (PTBP1) is a highly conserved RNA-binding protein that is a well-known regulator of alternative splicing. Testicular tissue is one of the richest tissues with respect to the number of alternative splicing mRNA isoforms, but the molecular role(s) of PTBP1 in the regulation of these isoforms during spermatogenesis is still unclear. Here, we developed a germ cell-specific Ptbp1 conditional knockout (cKO) mouse model by using the Cre-loxP system to investigate the role of PTBP1 in spermatogenesis. Testis weight in Ptbp1 cKO mice was comparable to that in age-matched controls until 3 weeks of age; at ≥ 2 months old, testis weight was significantly lighter in cKO mice than in age-matched controls. Sperm count in Ptbp1 cKO mice at 2 months old was comparable to that in controls, whereas sperm count significantly decreased at 6 months old. Seminiferous tubules that exhibited degeneration in spermatogenic function were more evident in the 2-month-old Ptbp1 cKO mice than in controls. In addition, the early neonatal proliferation of spermatogonia, during postnatal days 1-5, was significantly retarded in Ptbp1 cKO mice compared with that in controls. An in vitro spermatogonia culture model (germline stem cells) revealed that hydroxytamoxifen-induced deletion of PTBP1 from germline stem cells caused severe proliferation arrest accompanied by an increase of apoptotic cell death. These data suggest that PTBP1 contributes to spermatogenesis through regulation of spermatogonia proliferation.


Subject(s)
Cell Proliferation/physiology , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Polypyrimidine Tract-Binding Protein/physiology , Spermatogenesis/physiology , Spermatogonia/cytology , Alternative Splicing/physiology , Animals , Apoptosis , Gene Expression , Heterogeneous-Nuclear Ribonucleoproteins/deficiency , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Male , Mice, Knockout , Organ Size , Polypyrimidine Tract-Binding Protein/deficiency , Polypyrimidine Tract-Binding Protein/genetics , Seminiferous Tubules/physiology , Sperm Count , Spermatocytes/metabolism , Spermatogonia/metabolism , Testis/cytology , Testis/growth & development
18.
Acta Cir Bras ; 33(10): 904-913, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30484500

ABSTRACT

PURPOSE: To quantify, through stereological and morphometric analysis, spermatogenesis in rats undergoing the natural aging process. METHODS: Seventy-two male Wistar rats were divided into 6 equal groups according to age at the time of killing: 3, 6, 9, 12, 18, and 24 months. All the rats were subjected orchiectomy and collection of testicular parenchymal fragments for histological and morphometric analysis. The numerical density of spermatids was calculated using a stereological study, and morphometric analysis was conducted to measure the height of the germinal epithelium and the area of the seminiferous tubules. RESULTS: We found that the 18 and 24 months groups showed a significant reduction in the number of round spermatids. However, the height of the germinal epithelium was not significantly different between the groups. The area of seminiferous tubules was also significantly reduced in the elderly rats compared to that in the young ones. CONCLUSION: Aging of rats showed a significant reduction in the number of round spermatids and the area of the seminiferous tubules, more pronounced in the rats at 18 and 24 months of life.


Subject(s)
Aging/physiology , Seminiferous Tubules/anatomy & histology , Spermatids/physiology , Spermatogenesis/physiology , Animals , Disease Models, Animal , Male , Orchiectomy , Rats , Rats, Wistar , Seminiferous Tubules/physiology , Seminiferous Tubules/surgery , Sperm Count
19.
Bull Exp Biol Med ; 166(2): 268-273, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30488204

ABSTRACT

In experiments on white outbred male rats, a freshly removed (20 experiments) or cryopreserved (10 experiments) testicle from newborn rats (1-2 days after birth) was transplanted under the renal capsule after bilateral orchiectomy. In all experiments with transplantation of freshly removed testicle, it was engrafted. In 3 months, histological examination revealed the formation of mature seminiferous tubules, but spermatogenesis was blocked at the stage of spermatogonia; groups of proliferating Leydig cells in the loose connective tissue between the tubules were also seen. In 6 and 12 months, the status of the seminiferous tubules remained unchanged, but structures typical of the epididymis and developing vas deferens were revealed. The number of proliferating Leydig cells increased. The initially low testosterone concentration in the blood of castrated males increased significantly as soon as in 1 month after transplantation and grew up to 3 months, remaining at a level ~50% of normal. Engraftment of cryopreserved neonatal testicular tissue was observed in 60% cases, however, engrafted tissue, similar to the fresh one, retained the ability for organogenesis with the formation of mature seminiferous tubules, epididymis, and groups of proliferating Leydig cells. The dynamics of blood testosterone concentration in rats with cryopreserved and fresh transplantation was similar. Subcapsular transplantation did not adversely affect the kidneys, which was seen from normal histological structure of the kidneys and creatinine and urea concentrations in the blood.


Subject(s)
Cryopreservation/methods , Graft Survival , Orchiectomy , Organogenesis , Testis/transplantation , Testosterone/biosynthesis , Animals , Animals, Newborn , Animals, Outbred Strains , Creatinine/blood , Cryoprotective Agents/pharmacology , Dimethyl Sulfoxide/pharmacology , Epididymis/cytology , Epididymis/physiology , Kidney , Kidney Function Tests , Leydig Cells/cytology , Leydig Cells/physiology , Male , Rats , Regenerative Medicine/methods , Seminiferous Tubules/cytology , Seminiferous Tubules/physiology , Testis/cytology , Testis/growth & development , Testis/surgery , Testosterone/blood , Transplantation, Heterotopic , Transplantation, Homologous , Urea/blood
20.
Hum Reprod ; 33(12): 2249-2255, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30358843

ABSTRACT

STUDY QUESTION: Can transplanted primate testicular cells form seminiferous tubules de novo, supporting complete spermatogenesis? SUMMARY ANSWER: Cryopreserved testicular cells from a prepubertal monkey can reorganize in an adult monkey recipient testis forming de novo seminiferous tubular cords supporting complete spermatogenesis. WHAT IS KNOWN ALREADY: De novo morphogenesis of testicular tissue using aggregated cells from non-primate species grafted either subcutaneously or in the testis can support spermatogenesis. STUDY DESIGN, SIZE, DURATION: Two postpubertal rhesus monkeys (Macaca mulatta) were given testicular irradiation. One monkey was given GnRH-antagonist treatment from 8 to 16 weeks after irradiation, while the other received sham injections. At 16 weeks, cryopreserved testicular cells from two different prepubertal monkeys [43 × 106 viable (Trypan-blue excluding) cells in 260 µl, and 80 × 106 viable cells in 400 µl] were transplanted via ultrasound-guided injections to one of the rete testis in each recipient, and immune suppression was given. The contralateral testis was sham transplanted. Testes were analyzed 9 months after transplantation. PARTICIPANTS/MATERIALS, SETTING, METHODS: Spermatogenic recovery was assessed by testicular volume, weight, histology and immunofluorescence. Microsatellite genotyping of regions of testicular sections obtained by LCM determined whether the cells were derived from the host or transplanted cells. MAIN RESULTS AND THE ROLE OF CHANCE: Transplanted testis of the GnRH-antagonist-treated recipient, but not the sham-treated recipient, contained numerous irregularly shaped seminiferous tubular cords, 89% of which had differentiating germ cells, including sperm in a few of them. The percentages of donor genotype in different regions of this testis were as follows: normal tubule, 0%; inflammatory, 0%; abnormal tubule region, 67%; whole interior of abnormal tubules, >99%; adluminal region of the abnormal tubules, 92%. Thus, these abnormal tubules, including the enclosed germ cells, were derived de novo from the donor testicular cells. LARGE SCALE DATA: Not applicable. LIMITATIONS, REASONS FOR CAUTION: The de novo tubules were observed in only one out of the two monkeys transplanted with prepubertal donor testicular cells. WIDER IMPLICATIONS OF THE FINDINGS: These findings may represent a promising strategy for restoration of fertility in male childhood cancer survivors. The approach could be particularly useful in those exposed to therapeutic agents that are detrimental to the normal development of the tubule somatic cells affecting the ability of the endogenous tubules to support spermatogenesis, even from transplanted spermatogonial stem cells. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by research grants P01 HD075795 from Eunice Kennedy Shriver National Institute of Child Health & Human Development (NICHD/NIH) to K.E.O and Cancer Center Support Grant P30 CA016672 from NCI/NIH to The University of Texas MD Anderson Cancer Center. The authors declare that they have no competing interests.


Subject(s)
Seminiferous Tubules/physiology , Spermatogenesis/physiology , Testis/cytology , Testis/transplantation , Animals , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacology , Macaca mulatta , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...