Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 2111, 2023 04 17.
Article in English | MEDLINE | ID: mdl-37069147

ABSTRACT

In sexual reproduction, sperm contribute half the genomic material required for creation of offspring yet core molecular mechanisms essential for their formation are undefined. Here, the α-arrestin molecule arrestin-domain containing 5 (ARRDC5) is identified as an essential regulator of mammalian spermatogenesis. Multispecies testicular tissue transcriptome profiling indicates that expression of Arrdc5 is testis enriched, if not specific, in mice, pigs, cattle, and humans. Knockout of Arrdc5 in mice leads to male specific sterility due to production of low numbers of sperm that are immotile and malformed. Spermiogenesis, the final phase of spermatogenesis when round spermatids transform to spermatozoa, is defective in testes of Arrdc5 deficient mice. Also, epididymal sperm in Arrdc5 knockouts are unable to capacitate and fertilize oocytes. These findings establish ARRDC5 as an essential regulator of mammalian spermatogenesis. Considering the role of arrestin molecules as modulators of cellular signaling and ubiquitination, ARRDC5 is a potential male contraceptive target.


Subject(s)
Arrestins , Infertility, Male , Testis , Animals , Cattle , Humans , Male , Mice , Arrestins/genetics , Arrestins/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Mice, Knockout , Morphogenesis , Semen/metabolism , Spermatogenesis/genetics , Spermatozoa/metabolism , Swine , Testis/metabolism
2.
Front Vet Sci ; 9: 894075, 2022.
Article in English | MEDLINE | ID: mdl-35928111

ABSTRACT

The undifferentiated spermatogonial population in mammalian testes contains a spermatogonial stem cell (SSC) population that can regenerate continual spermatogenesis following transplantation. This capacity has the potential to be exploited as a surrogate sires breeding tool to achieve widespread dissemination of desirable genetics in livestock production. Because SSCs are relatively rare in testicular tissue, the ability to expand a population in vitro would be advantageous to provide large numbers for transplantation into surrogate recipient males. Here, we evaluated conditions that would support long-term in-vitro maintenance of undifferentiated spermatogonia from a goat breed that is endemic to Kenyan livestock production. Single-cell suspensions enriched for undifferentiated spermatogonia from pre-pubertal bucks were seeded on laminin-coated tissue culture plates and maintained in a commercial media based on serum-free composition. The serum-free media was conditioned on goat fetal fibroblasts and supplemented with a growth factor cocktail that included glial cell line-derived neurotrophic factor (GDNF), leukemia inhibitory factor (LIF), stromal cell-derived factor (SDF), and fibroblast growth factor (FGF) before use. Over 45 days, the primary cultures developed a cluster morphology indicative of in-vitro grown undifferentiated spermatogonia from other species and expressed the germ cell marker VASA, as well as the previously defined spermatogonial marker such as promyelocytic leukemia zinc finger (PLZF). Taken together, these findings provide a methodology for isolating the SSC containing undifferentiated spermatogonial population from goat testes and long-term maintenance in defined culture conditions.

3.
Biol Reprod ; 106(6): 1175-1190, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35244684

ABSTRACT

Spermatogenic regeneration is key for male fertility and relies on activities of an undifferentiated spermatogonial population. Here, a high-throughput approach with primary cultures of mouse spermatogonia was devised to rapidly predict alterations in functional capacity. Combining the platform with a large-scale RNAi screen of transcription factors, we generated a repository of new information from which pathway analysis was able to predict candidate molecular networks regulating regenerative functions. Extending from this database, the SRCAP-CREBBP/EP300 (Snf2-related CREBBP activator protein-CREB binding protein/E1A binding protein P300) complex was found to mediate differential levels of histone acetylation between stem cell and progenitor spermatogonia to influence expression of key self-renewal genes including the previously undescribed testis-specific transcription factor ZSCAN2 (zinc finger and SCAN domain containing 2). Single cell RNA sequencing analysis revealed that ZSCAN2 deficiency alters key cellular processes in undifferentiated spermatogonia such as translation, chromatin modification, and ubiquitination. In Zscan2 knockout mice, while spermatogenesis was moderately impacted during steady state, regeneration after cytotoxic insult was significantly impaired. Altogether, these findings have validated the utility of our high-throughput screening approach and have generated a transcription factor database that can be utilized for uncovering novel mechanisms governing spermatogonial functions.


Subject(s)
Spermatogenesis , Spermatogonia , Animals , Cell Differentiation , Male , Mice , Spermatogenesis/physiology , Stem Cells , Testis/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Stem Cell Reports ; 16(6): 1555-1567, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33961790

ABSTRACT

Maintenance and self-renewal of the spermatogonial stem cell (SSC) population is the cornerstone of male fertility. Here, we have identified a key role for the nucleosome remodeling protein CHD4 in regulating SSC function. Gene expression analyses revealed that CHD4 expression is highly enriched in the SSC population in the mouse testis. Using spermatogonial transplantation techniques it was established that loss of Chd4 expression significantly impairs SSC regenerative capacity, causing a ∼50% reduction in colonization of recipient testes. An scRNA-seq comparison revealed reduced expression of "self-renewal" genes following Chd4 knockdown, along with increased expression of signature progenitor genes. Co-immunoprecipitation analyses demonstrated that CHD4 regulates gene expression in spermatogonia not only through its traditional association with the remodeling complex NuRD, but also via interaction with the GDNF-responsive transcription factor SALL4. Cumulatively, the results of this study depict a previously unappreciated role for CHD4 in controlling fate decisions in the spermatogonial pool.


Subject(s)
Adult Germline Stem Cells/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Stem Cells/metabolism , Testis/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Self Renewal , DNA Helicases/genetics , Gene Expression Regulation , Gene Knockdown Techniques/methods , Male , Mice , Mice, Inbred Strains , Transcriptome
5.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33929507

ABSTRACT

The stem cell-containing undifferentiated spermatogonial population in mammals, which ensures continual sperm production, arises during development from prospermatogonial precursors. Although a period of quiescence is known to occur in prospermatogonia prior to postnatal spermatogonial transition, the importance of this has not been defined. Here, using mouse models with conditional knockout of the master cell cycle regulator Rb1 to disrupt normal timing of the quiescence period, we found that failure to initiate mitotic arrest during fetal development leads to prospermatogonial apoptosis and germline ablation. Outcomes of single-cell RNA-sequencing analysis indicate that oxidative phosphorylation activity and inhibition of meiotic initiation are disrupted in prospermatogonia that fail to enter quiescence on a normal timeline. Taken together, these findings suggest that key layers of programming are laid down during the quiescent period in prospermatogonia to ensure proper fate specification and fitness in postnatal life.


Subject(s)
Cell Division/physiology , Spermatogonia/cytology , Spermatogonia/growth & development , Stem Cells/cytology , Animals , Apoptosis , Cell Proliferation , Gene Expression Regulation, Developmental , Male , Mice , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/genetics , Retinoblastoma Binding Proteins/genetics , Sequence Analysis, RNA , Spermatogenesis/physiology , Spermatogonia/metabolism , Spermatozoa , Transcriptome
6.
Proc Natl Acad Sci U S A ; 117(39): 24195-24204, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929012

ABSTRACT

Spermatogonial stem cell transplantation (SSCT) is an experimental technique for transfer of germline between donor and recipient males that could be used as a tool for biomedical research, preservation of endangered species, and dissemination of desirable genetics in food animal populations. To fully realize these potentials, recipient males must be devoid of endogenous germline but possess normal testicular architecture and somatic cell function capable of supporting allogeneic donor stem cell engraftment and regeneration of spermatogenesis. Here we show that male mice, pigs, goats, and cattle harboring knockout alleles of the NANOS2 gene generated by CRISPR-Cas9 editing have testes that are germline ablated but otherwise structurally normal. In adult pigs and goats, SSCT with allogeneic donor stem cells led to sustained donor-derived spermatogenesis. With prepubertal mice, allogeneic SSCT resulted in attainment of natural fertility. Collectively, these advancements represent a major step toward realizing the enormous potential of surrogate sires as a tool for dissemination and regeneration of germplasm in all mammalian species.


Subject(s)
Adult Germline Stem Cells/transplantation , RNA-Binding Proteins/physiology , Spermatogenesis , Animals , Cattle , Female , Goats , Male , Mice , Mice, Knockout , Swine , Testis/anatomy & histology , Testis/physiology , Transplantation, Homologous
7.
Mol Cell ; 79(4): 645-659.e9, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32692974

ABSTRACT

Stress granules (SGs) are membrane-less ribonucleoprotein condensates that form in response to various stress stimuli via phase separation. SGs act as a protective mechanism to cope with acute stress, but persistent SGs have cytotoxic effects that are associated with several age-related diseases. Here, we demonstrate that the testis-specific protein, MAGE-B2, increases cellular stress tolerance by suppressing SG formation through translational inhibition of the key SG nucleator G3BP. MAGE-B2 reduces G3BP protein levels below the critical concentration for phase separation and suppresses SG initiation. Knockout of the MAGE-B2 mouse ortholog or overexpression of G3BP1 confers hypersensitivity of the male germline to heat stress in vivo. Thus, MAGE-B2 provides cytoprotection to maintain mammalian spermatogenesis, a highly thermosensitive process that must be preserved throughout reproductive life. These results demonstrate a mechanism that allows for tissue-specific resistance against stress and could aid in the development of male fertility therapies.


Subject(s)
Cytoplasmic Granules/genetics , DNA Helicases/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Protein Biosynthesis , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , Stress, Physiological/genetics , 5' Untranslated Regions , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/pathology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA Helicases/metabolism , Female , HCT116 Cells , HeLa Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Spermatogonia/cytology , Spermatogonia/pathology , Testis/cytology , Testis/metabolism
8.
Nat Commun ; 10(1): 2787, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31243281

ABSTRACT

Continuity, robustness, and regeneration of cell lineages relies on stem cell pools that are established during development. For the mammalian spermatogenic lineage, a foundational spermatogonial stem cell (SSC) pool arises from prospermatogonial precursors during neonatal life via mechanisms that remain undefined. Here, we mapped the kinetics of this process in vivo using a multi-transgenic reporter mouse model, in silico with single-cell RNA sequencing, and functionally with transplantation analyses to define the SSC trajectory from prospermatogonia. Outcomes revealed that a heterogeneous prospermatogonial population undergoes dynamic changes during late fetal and neonatal development. Differential transcriptome profiles predicted divergent developmental trajectories from fetal prospermatogonia to descendant postnatal spermatogonia. Furthermore, transplantation analyses demonstrated that a defined subset of fetal prospermatogonia is fated to function as SSCs. Collectively, these findings suggest that SSC fate is preprogrammed within a subset of fetal prospermatogonia prior to building of the foundational pool during early neonatal development.


Subject(s)
Adult Germline Stem Cells/physiology , Cell Lineage , Testis/embryology , Animals , Cell Differentiation , Embryonic Development , Gene Expression Regulation, Developmental/physiology , Genes, Reporter , Male , Mice , Mice, Transgenic , RNA/genetics , Spermatogenesis/physiology , Spermatogonia/physiology
9.
Sci Adv ; 5(5): eaav4832, 2019 05.
Article in English | MEDLINE | ID: mdl-31149633

ABSTRACT

Ensuring robust gamete production even in the face of environmental stress is of utmost importance for species survival, especially in mammals that have low reproductive rates. Here, we describe a family of genes called melanoma antigens (MAGEs) that evolved in eutherian mammals and are normally restricted to expression in the testis (http://MAGE.stjude.org) but are often aberrantly activated in cancer. Depletion of Mage-a genes disrupts spermatogonial stem cell maintenance and impairs repopulation efficiency in vivo. Exposure of Mage-a knockout mice to genotoxic stress or long-term starvation that mimics famine in nature causes defects in spermatogenesis, decreased testis weights, diminished sperm production, and reduced fertility. Last, human MAGE-As are activated in many cancers where they promote fuel switching and growth of cells. These results suggest that mammalian-specific MAGE genes have evolved to protect the male germline against environmental stress, ensure reproductive success under non-optimal conditions, and are hijacked by cancer cells.


Subject(s)
Melanoma-Specific Antigens/genetics , Neoplasms/genetics , Spermatogenesis/genetics , Stress, Physiological/genetics , Testis/physiology , Animals , DNA Damage , Deoxyglucose/pharmacology , Evolution, Molecular , Female , Gene Expression Regulation, Neoplastic , Germ Cells , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Spermatogonia/drug effects , Starvation
10.
J Anim Sci ; 97(4): 1468-1477, 2019 Apr 03.
Article in English | MEDLINE | ID: mdl-30576512

ABSTRACT

Male reproductive capacity is a critical component of cattle production and the majority of genetic gain is made via selective utilization of gametes from desirable sires. Thus, strategies that enhance sperm production increase the availability of elite genetics for use in improving production characteristics of populations on a worldwide scale. In all mammals, the amount of sperm produced is strongly correlated to the number of Sertoli cells in testes. Studies with rodents showed that the size of the Sertoli cell population is set during prepubertal development via signaling from thyroid hormones. Here, we devised a strategy to increase Sertoli cell number in bulls via induction of a transient hypothyroidic state just prior to and extending beyond the period of Sertoli cell proliferation that we found to normally cease between 4.5 and 5 mo of age. In adulthood, these bulls produced a significantly greater number of sperm compared to age-matched controls and their testes contained nearly 2 times more Sertoli cells. Importantly, sperm motility, morphology, fertilizing ability, and viability after cryopreservation were found to be no different for treated bulls compared to untreated control bulls. This strategy of transient induction of hypothyroidism during a defined period of prepubertal development in bulls could prove to be an efficacious approach for enhancing daily sperm production in genetically desirable sires that will, in turn, provide an avenue for improving the efficiency of commercial cattle production.

11.
Cell Rep ; 25(6): 1650-1667.e8, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30404016

ABSTRACT

Spermatogenesis is a complex and dynamic cellular differentiation process critical to male reproduction and sustained by spermatogonial stem cells (SSCs). Although patterns of gene expression have been described for aggregates of certain spermatogenic cell types, the full continuum of gene expression patterns underlying ongoing spermatogenesis in steady state was previously unclear. Here, we catalog single-cell transcriptomes for >62,000 individual spermatogenic cells from immature (postnatal day 6) and adult male mice and adult men. This allowed us to resolve SSC and progenitor spermatogonia, elucidate the full range of gene expression changes during male meiosis and spermiogenesis, and derive unique gene expression signatures for multiple mouse and human spermatogenic cell types and/or subtypes. These transcriptome datasets provide an information-rich resource for studies of SSCs, male meiosis, testicular cancer, male infertility, or contraceptive development, as well as a gene expression roadmap to be emulated in efforts to achieve spermatogenesis in vitro.


Subject(s)
Mammals/genetics , Single-Cell Analysis , Spermatids/cytology , Spermatogenesis/genetics , Spermatogonia/cytology , Transcriptome/genetics , Adult , Aging/genetics , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Haploidy , Humans , Male , Meiosis , Mice, Inbred C57BL , Signal Transduction , Spermatids/metabolism , Spermatogonia/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Testis/cytology
12.
Stem Cell Reports ; 10(2): 538-552, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29398482

ABSTRACT

Spermatogenesis requires retinoic acid (RA) induction of the undifferentiated to differentiating transition in transit amplifying (TA) progenitor spermatogonia, whereas continuity of the spermatogenic lineage relies on the RA response being suppressed in spermatogonial stem cells (SSCs). Here, we discovered that, in mouse testes, both spermatogonial populations possess intrinsic RA-response machinery and exhibit hallmarks of the differentiating transition following direct exposure to RA, including loss of SSC regenerative capacity. We determined that SSCs are only resistant to RA-driven differentiation when situated in the normal topological organization of the testis. Furthermore, we show that the soma is instrumental in "priming" TA progenitors for RA-induced differentiation through elevated RA receptor expression. Collectively, these findings indicate that SSCs and TA progenitor spermatogonia inhabit disparate niche microenvironments within seminiferous tubules that are critical for mediating extrinsic cues that drive fate decisions.


Subject(s)
Adult Germline Stem Cells/drug effects , Spermatogenesis/genetics , Spermatogonia/cytology , Testis/cytology , Adult Germline Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Male , Mice , Seminiferous Tubules/cytology , Seminiferous Tubules/growth & development , Spermatogenesis/drug effects , Spermatogonia/growth & development , Testis/growth & development , Tretinoin/administration & dosage , Tretinoin/metabolism
13.
Dev Biol ; 432(2): 229-236, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29037932

ABSTRACT

The onset of spermatogenesis occurs in response to retinoic acid (RA), the active metabolite of vitamin A. However, whether RA plays any role during establishment of the spermatogonial stem cell (SSC) pool is unknown. Because designation of the SSC population and the onset of RA signaling in the testis that induces differentiation have similar timing, this study asked whether RA influenced SSC establishment. Whole mount immunofluorescence and flow cytometric analysis using the Id4-eGfp transgenic reporter mouse line revealed an enrichment for ID4-EGFP+ cells within the testis following inhibition of RA synthesis by WIN 18,446 treatment. Transplantation analyses confirmed a significant increase in the number of SSCs in testes from RA-deficient animals. Conversely, no difference in the ID4-EGFP+ population or change in SSC number were detected following exposure to an excess of RA. Collectively, reduced RA altered the number of SSCs present in the neonatal testis but precocious RA exposure in the neonatal testis did not, suggesting that RA deficiency causes a greater proportion of progenitor undifferentiated spermatogonia to retain their SSC state past the age when the pool is thought to be determined.


Subject(s)
Spermatogenesis/physiology , Tretinoin/metabolism , Adult Germline Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Male , Mice , Mice, Transgenic , Signal Transduction/drug effects , Spermatogenesis/genetics , Spermatogonia/cytology , Testis/metabolism
14.
Stem Cell Reports ; 8(5): 1430-1441, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28392219

ABSTRACT

The application of spermatogonial stem cell (SSC) transplantation for regenerating male fertility requires amplification of SSC number in vitro during which the integrity to re-establish spermatogenesis must be preserved. Conventional conditions supporting proliferation of SSCs from mouse pups have been the basis for developing methodology with adult human cells but are unrefined. We found that the integrity to regenerate spermatogenesis after transplantation declines with advancing time in primary cultures of pup SSCs and that the efficacy of deriving cultures from adult SSCs is limited with conventional conditions. To address these deficiencies, we optimized the culture environment to favor glycolysis as the primary bioenergetics process. In these conditions, regenerative integrity of pup and adult SSCs was significantly improved and the efficiency of establishing primary cultures was 100%. Collectively, these findings suggest that SSCs are primed for conditions favoring glycolytic activity, and matching culture environments to their bioenergetics is critical for maintaining functional integrity.


Subject(s)
Glycolysis , Spermatogenesis , Spermatogonia/transplantation , Animals , Cells, Cultured , Male , Mice , Primary Cell Culture/methods , Regeneration , Spermatogonia/cytology , Spermatogonia/metabolism , Spermatogonia/physiology , Stem Cell Transplantation/methods
15.
Development ; 144(4): 624-634, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087628

ABSTRACT

Spermatogenesis is a classic model of cycling cell lineages that depend on a balance between stem cell self-renewal for continuity and the formation of progenitors as the initial step in the production of differentiated cells. The mechanisms that guide the continuum of spermatogonial stem cell (SSC) to progenitor spermatogonial transition and precise identifiers of subtypes in the process are undefined. Here we used an Id4-eGfp reporter mouse to discover that EGFP intensity is predictive of the subsets, with the ID4-EGFPBright population being mostly, if not purely, SSCs, whereas the ID4-EGFPDim population is in transition to the progenitor state. These subsets are also distinguishable by transcriptome signatures. Moreover, using a conditional overexpression mouse model, we found that transition from the stem cell to the immediate progenitor state requires downregulation of Id4 coincident with a major change in the transcriptome. Collectively, our results demonstrate that the level of ID4 is predictive of stem cell or progenitor capacity in spermatogonia and dictates the interface of transition between the different functional states.


Subject(s)
Gene Expression Regulation , Inhibitor of Differentiation Proteins/physiology , Spermatogenesis , Spermatogonia/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Self Renewal , Gene Expression Regulation, Developmental , Genes, Reporter , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Testis/metabolism , Transcriptome , Transgenes
16.
Sci Rep ; 7: 40176, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28071690

ABSTRACT

Genome editing tools have revolutionized the generation of genetically modified animals including livestock. In particular, the domestic pig is a proven model of human physiology and an agriculturally important species. In this study, we utilized the CRISPR/Cas9 system to edit the NANOS2 gene in pig embryos to generate offspring with mono-allelic and bi-allelic mutations. We found that NANOS2 knockout pigs phenocopy knockout mice with male specific germline ablation but other aspects of testicular development are normal. Moreover, male pigs with one intact NANOS2 allele and female knockout pigs are fertile. From an agriculture perspective, NANOS2 knockout male pigs are expected to serve as an ideal surrogate for transplantation of donor spermatogonial stem cells to expand the availability of gametes from genetically desirable sires.


Subject(s)
Animals, Genetically Modified , Gene Knockout Techniques , RNA-Binding Proteins/genetics , Sus scrofa/genetics , Animals , CRISPR-Cas Systems , Fertility , Infertility, Male , Male
17.
Biol Reprod ; 95(6): 117, 2016 12.
Article in English | MEDLINE | ID: mdl-27733379

ABSTRACT

Precise separation of spermatogonial stem cells (SSCs) from progenitor spermatogonia that lack stem cell activity and are committed to differentiation remains a challenge. To distinguish between these spermatogonial subtypes, we identified genes that exhibited bimodal mRNA levels at the single-cell level among undifferentiated spermatogonia from Postnatal Day 6 mouse testes, including Tspan8, Epha2, and Pvr, each of which encode cell surface proteins useful for cell selection. Transplantation studies provided definitive evidence that a TSPAN8-high subpopulation is enriched for SSCs. RNA-seq analyses identified genes differentially expressed between TSPAN8-high and -low subpopulations that clustered into multiple biological pathways potentially involved in SSC renewal or differentiation, respectively. Methyl-seq analysis identified hypomethylated domains in the promoters of these genes in both subpopulations that colocalized with peaks of histone modifications defined by ChIP-seq analysis. Taken together, these results demonstrate functional heterogeneity among mouse undifferentiated spermatogonia and point to key biological characteristics that distinguish SSCs from progenitor spermatogonia.


Subject(s)
Adult Germline Stem Cells/cytology , Testis/cytology , Tetraspanins/metabolism , Adult Germline Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Cycle/physiology , Gene Expression Profiling , Male , Mice , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Spermatogenesis , Testis/metabolism , Tetraspanins/genetics
18.
Biol Reprod ; 95(1): 14, 2016 07.
Article in English | MEDLINE | ID: mdl-27251094

ABSTRACT

Continual and robust spermatogenesis relies on the actions of an undifferentiated spermatogonial population that contains stem cells. A remarkable feature of spermatogonial stem cells (SSCs) is the capacity to regenerate spermatogenesis following isolation from a donor testis and transplantation into a permissive recipient testis. This capacity has enormous potential as a tool for enhancing the reproductive capacity of livestock, which can improve production efficiency. Because SSCs are a rare subset of the undifferentiated spermatogonial population, a period of in vitro amplification in number following isolation from donor testicular tissue is essential. Here, we describe methodology for isolation of a cell fraction from prepubertal bull testes that is enriched for undifferentiated spermatogonia and long-term maintenance of the cells in both the feeder cell coculture and the feeder-free format. To achieve this method, we derived bovine fetal fibroblasts (BFF) to serve as feeders for optimizing medium conditions that promote maintenance of bovine undifferentiated spermatogonia for at least 2 mo. In addition, we devised a feeder-free system with BFF-conditioned medium that sustained bovine undifferentiated spermatogonia for at least 1 mo in vitro. The methodologies described could be optimized to provide platforms for exponential expansion of bovine SSCs that will provide the numbers needed for transplantation into recipient testes.


Subject(s)
Cell Culture Techniques/methods , Spermatogenesis/physiology , Spermatogonia/cytology , Stem Cells/cytology , Testis/cytology , Animals , Cattle , Cells, Cultured , Culture Media , Male
19.
PLoS Genet ; 11(10): e1005569, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26496357

ABSTRACT

Skeletal dysplasias are a common, genetically heterogeneous cause of short stature that can result from disruptions in many cellular processes. We report the identification of the lesion responsible for skeletal dysplasia and male infertility in the spontaneous, recessive mouse mutant chagun. We determined that Poc1a, encoding protein of the centriole 1a, is disrupted by the insertion of a processed Cenpw cDNA, which is flanked by target site duplications, suggestive of a LINE-1 retrotransposon-mediated event. Mutant fibroblasts have impaired cilia formation and multipolar spindles. Male infertility is caused by defective spermatogenesis early in meiosis and progressive germ cell loss. Spermatogonial stem cell transplantation studies revealed that Poc1a is essential for normal function of both Sertoli cells and germ cells. The proliferative zone of the growth plate is small and disorganized because chondrocytes fail to re-align after cell division and undergo increased apoptosis. Poc1a and several other genes associated with centrosome function can affect the skeleton and lead to skeletal dysplasias and primordial dwarfisms. This mouse mutant reveals how centrosome dysfunction contributes to defects in skeletal growth and male infertility.


Subject(s)
Cytoskeletal Proteins/genetics , Dwarfism/genetics , Infertility, Male/genetics , Long Interspersed Nucleotide Elements/genetics , Spermatogenesis/genetics , Animals , Cell Cycle Proteins , Centrioles/genetics , Centrosome/metabolism , Chromosomal Proteins, Non-Histone/genetics , Dwarfism/pathology , Humans , Infertility, Male/pathology , Male , Meiosis/genetics , Mice , Proteins/genetics , Proteins/metabolism , Sertoli Cells/metabolism , Spermatogonia/metabolism
20.
Genes Dev ; 28(12): 1351-62, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24939937

ABSTRACT

The maintenance of cycling cell lineages relies on undifferentiated subpopulations consisting of stem and progenitor pools. Features that delineate these cell types are undefined for many lineages, including spermatogenesis, which is supported by an undifferentiated spermatogonial population. Here, we generated a transgenic mouse line in which spermatogonial stem cells are marked by expression of an inhibitor of differentiation 4 (Id4)-green fluorescent protein (Gfp) transgene. We found that Id4-Gfp(+) cells exist primarily as a subset of the type A(single) pool, and their frequency is greatest in neonatal development and then decreases in proportion during establishment of the spermatogenic lineage, eventually comprising ∼ 2% of the undifferentiated spermatogonial population in adulthood. RNA sequencing analysis revealed that expression of 11 and 25 genes is unique for the Id4-Gfp(+)/stem cell and Id4-Gfp(-)/progenitor fractions, respectively. Collectively, these findings provide the first definitive evidence that stem cells exist as a rare subset of the A(single) pool and reveal transcriptome features distinguishing stem cell and progenitor states within the mammalian male germline.


Subject(s)
Germ Cells/cytology , Inhibitor of Differentiation Proteins/metabolism , Stem Cells/cytology , Testis/cytology , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inhibitor of Differentiation Proteins/genetics , Male , Mice , Mice, Transgenic , Spermatogenesis/genetics , Spermatogonia/metabolism , Stem Cells/metabolism , Testis/metabolism , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...