Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Dev Cell ; 59(1): 79-90.e6, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38101411

ABSTRACT

Sperm gain fertilization competence in the female reproductive tract through a series of biochemical changes and a requisite switch from linear progressive to hyperactive motility. Despite being essential for fertilization, regulation of sperm energy transduction is poorly understood. This knowledge gap confounds interpretation of interspecies variation and limits progress in optimizing sperm selection for assisted reproduction. Here, we developed a model of mouse sperm bioenergetics using metabolic phenotyping data, quantitative microscopy, and spectral flow cytometry. The results define a mechanism of motility regulation by microenvironmental pyruvate. Rather than being consumed as a mitochondrial fuel source, pyruvate stimulates hyperactivation by repressing lactate oxidation and activating glycolysis in the flagellum through provision of nicotinamide adenine dinucleotide (NAD)+. These findings provide evidence that the transitions in motility requisite for sperm competence are governed by changes in the metabolic microenvironment, highlighting the unexplored potential of using catabolite combination to optimize sperm selection for fertilization.


Subject(s)
Pyruvic Acid , Sperm Motility , Male , Female , Animals , Mice , Pyruvic Acid/metabolism , Semen/metabolism , Energy Metabolism/physiology , Spermatozoa/metabolism , Oxidation-Reduction
2.
bioRxiv ; 2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37873304

ABSTRACT

The foundation of spermatogenesis and lifelong fertility is provided by spermatogonial stem cells (SSCs). SSCs divide asymmetrically to either replenish their numbers (self-renewal) or produce undifferentiated progenitors that proliferate before committing to differentiation. However, regulatory mechanisms governing SSC maintenance are poorly understood. Here, we show that the CCR4-NOT mRNA deadenylase complex subunit CNOT3 plays a critical role in maintaining spermatogonial populations in mice. Cnot3 is highly expressed in undifferentiated spermatogonia, and its deletion in spermatogonia resulted in germ cell loss and infertility. Single cell analyses revealed that Cnot3 deletion led to the de-repression of transcripts encoding factors involved in spermatogonial differentiation, including those in the glutathione redox pathway that are critical for SSC maintenance. Together, our study reveals that CNOT3 - likely via the CCR4-NOT complex - actively degrades transcripts encoding differentiation factors to sustain the spermatogonial pool and ensure the progression of spermatogenesis, highlighting the importance of CCR4-NOT-mediated post-transcriptional gene regulation during male germ cell development.

3.
iScience ; 26(9): 107616, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37694147

ABSTRACT

WNK1 is an important regulator in many physiological functions, yet its role in male reproduction is unexplored. In the male germline, WNK1 is upregulated in preleptotene spermatocytes indicating possible function(s) in spermatogenic meiosis. Indeed, deletion of Wnk1 in mid-pachytene spermatocytes using the Wnt7a-Cre mouse led to male sterility which resembled non-obstructive azoospermia in humans, where germ cells failed to complete spermatogenesis and produced no sperm. Mechanistically, we found elevated MTOR expression and signaling in the Wnk1-depleted spermatocytes. As MTOR is a central mediator of translation, we speculated that translation may be accelerated in these spermatocytes. Supporting this, we found the acrosome protein, ACRBP to be prematurely expressed in the spermatocytes with Wnk1 deletion. Our study uncovered an MTOR-regulating factor in the male germline with potential implications in translation, and future studies will aim to understand how WNK1 regulates MTOR activity and impact translation on a broader spectrum.

4.
Development ; 150(14)2023 07 15.
Article in English | MEDLINE | ID: mdl-37350382

ABSTRACT

Retinoic acid (RA) is the proposed mammalian 'meiosis inducing substance'. However, evidence for this role comes from studies in the fetal ovary, where germ cell differentiation and meiotic initiation are temporally inseparable. In the postnatal testis, these events are separated by more than 1 week. Exploiting this difference, we discovered that, although RA is required for spermatogonial differentiation, it is dispensable for the subsequent initiation, progression and completion of meiosis. Indeed, in the absence of RA, the meiotic transcriptome program in both differentiating spermatogonia and spermatocytes entering meiosis was largely unaffected. Instead, transcripts encoding factors required during spermiogenesis were aberrant during preleptonema, and the subsequent spermatid morphogenesis program was disrupted such that no sperm were produced. Taken together, these data reveal a RA-independent model for male meiotic initiation.


Subject(s)
Testis , Tretinoin , Animals , Female , Male , Tretinoin/pharmacology , Spermatogenesis/genetics , Spermatogonia , Spermatozoa , Meiosis/genetics , Mammals
5.
Methods Mol Biol ; 2656: 227-237, 2023.
Article in English | MEDLINE | ID: mdl-37249875

ABSTRACT

The delivery, to newborn and juvenile mice, of drugs and other compounds that manipulate the physiology or cellular/molecular state -e.g., by activating or inhibiting signaling pathways) is a powerful, yet underutilized approach to studying spermatogenesis. Here, we provide detailed protocols we have optimized in our laboratory for safely and effectively feeding and injecting mice and discuss troubleshooting approaches.


Subject(s)
Spermatogenesis , Testis , Male , Animals , Mice , Testis/metabolism
6.
Biol Reprod ; 108(3): 447-464, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36617158

ABSTRACT

The formation of fertilisation-competent sperm requires spermatid morphogenesis (spermiogenesis), a poorly understood program that involves complex coordinated restructuring and specialised cytoskeletal structures. A major class of cytoskeletal regulators are the actin-related proteins (ARPs), which include conventional actin variants, and related proteins that play essential roles in complexes regulating actin dynamics, intracellular transport, and chromatin remodeling. Multiple testis-specific ARPs are well conserved among mammals, but their functional roles are unknown. One of these is actin-like 7b (Actl7b) that encodes an orphan ARP highly similar to the ubiquitously expressed beta actin (ACTB). Here we report ACTL7B is expressed in human and mouse spermatids through the elongation phase of spermatid development. In mice, ACTL7B specifically localises to the developing acrosome, within the nucleus of early spermatids, and to the flagellum connecting region. Based on this localisation pattern and high level of sequence conservation in mice, humans, and other mammals, we examined the requirement for ACTL7B in spermiogenesis by generating and characterising the reproductive phenotype of male Actl7b KO mice. KO mice were infertile, with severe and variable oligoteratozoospermia (OAT) and multiple morphological abnormalities of the flagellum (MMAF) and sperm head. These defects phenocopy human OAT and MMAF, which are leading causes of idiopathic male infertility. In conclusion, this work identifies ACTL7B as a key regulator of spermiogenesis that is required for male fertility.


Subject(s)
Actins , Infertility, Male , Male , Mice , Humans , Animals , Actins/genetics , Actins/metabolism , Avena , Seeds/metabolism , Spermatids/metabolism , Spermatogenesis/genetics , Flagella/metabolism , Infertility, Male/genetics , Infertility, Male/metabolism , Mammals
7.
Biol Reprod ; 108(5): 822-836, 2023 05 10.
Article in English | MEDLINE | ID: mdl-36708226

ABSTRACT

The foundation of mammalian spermatogenesis is provided by undifferentiated spermatogonia, which comprise of spermatogonial stem cells (SSCs) and transit-amplifying progenitors that differentiate in response to retinoic acid (RA) and are committed to enter meiosis. Our laboratory recently reported that the foundational populations of SSCs, undifferentiated progenitors, and differentiating spermatogonia are formed in the neonatal testis in part based on their differential responsiveness to RA. Here, we expand on those findings to define the extent to which RA responsiveness during steady-state spermatogenesis in the adult testis regulates the spermatogonial fate. Our results reveal that both progenitor and differentiating spermatogonia throughout the testis are capable of responding to exogenous RA, but their resulting fates were quite distinct-undifferentiated progenitors precociously differentiated and proceeded into meiosis on a normal timeline, while differentiating spermatogonia were unable to hasten their entry into meiosis. This reveals that the spermatogonia responding to RA must still complete the 8.6 day differentiation program prior to their entry into meiosis. Addition of exogenous RA enriched testes with preleptotene and pachytene spermatocytes one and two seminiferous cycles later, respectively, supporting recent clinical studies reporting increased sperm production and enhanced fertility in subfertile men on long-term RA analog treatment. Collectively, our results reveal that a well-buffered system exists within mammalian testes to regulate spermatogonial RA exposure, that exposed undifferentiated progenitors can precociously differentiate, but must complete a normal-length differentiation program prior to entering meiosis, and that daily RA treatments increased the numbers of advanced germ cells by directing undifferentiated progenitors to continuously differentiate.


Subject(s)
Spermatogonia , Tretinoin , Animals , Infant, Newborn , Male , Humans , Tretinoin/pharmacology , Semen , Spermatogenesis/physiology , Testis , Cell Differentiation/physiology , Mammals
8.
Development ; 149(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36250451

ABSTRACT

In mammalian testes, premeiotic spermatogonia respond to retinoic acid by completing an essential lengthy differentiation program before initiating meiosis. The molecular and cellular changes directing these developmental processes remain largely undefined. This wide gap in knowledge is due to two unresolved technical challenges: (1) lack of robust and reliable in vitro models to study differentiation and meiotic initiation; and (2) lack of methods to isolate large and pure populations of male germ cells at each stage of differentiation and at meiotic initiation. Here, we report a facile in vitro differentiation and meiotic initiation system that can be readily manipulated, including the use of chemical agents that cannot be safely administered to live animals. In addition, we present a transgenic mouse model enabling fluorescence-activated cell sorting-based isolation of millions of spermatogonia at specific developmental stages as well as meiotic spermatocytes.


Subject(s)
Spermatogenesis , Spermatogonia , Male , Mice , Animals , Spermatocytes , Testis , Meiosis , Cell Differentiation , Mice, Transgenic , Mammals
9.
PLoS Genet ; 18(9): e1010416, 2022 09.
Article in English | MEDLINE | ID: mdl-36129965

ABSTRACT

Control over gene expression is exerted, in multiple stages of spermatogenesis, at the post-transcriptional level by RNA binding proteins (RBPs). We identify here an essential role in mammalian spermatogenesis and male fertility for 'RNA binding protein 46' (RBM46). A highly evolutionarily conserved gene, Rbm46 is also essential for fertility in both flies and fish. We found Rbm46 expression was restricted to the mouse germline, detectable in males in the cytoplasm of premeiotic spermatogonia and meiotic spermatocytes. To define its requirement for spermatogenesis, we generated Rbm46 knockout (KO, Rbm46-/-) mice; although male Rbm46-/- mice were viable and appeared grossly normal, they were infertile. Testes from adult Rbm46-/- mice were small, with seminiferous tubules containing only Sertoli cells and few undifferentiated spermatogonia. Using genome-wide unbiased high throughput assays RNA-seq and 'enhanced crosslinking immunoprecipitation' coupled with RNA-seq (eCLIP-seq), we discovered RBM46 could bind, via a U-rich conserved consensus sequence, to a cohort of mRNAs encoding proteins required for completion of differentiation and subsequent meiotic initiation. In summary, our studies support an essential role for RBM46 in regulating target mRNAs during spermatogonia differentiation prior to the commitment to meiosis in mice.


Subject(s)
RNA-Binding Proteins/metabolism , Spermatogenesis , Spermatogonia , Animals , Cell Differentiation/genetics , Male , Mammals/genetics , Meiosis/genetics , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Spermatocytes/metabolism , Spermatogenesis/genetics , Spermatogonia/metabolism , Testis
10.
Nat Commun ; 12(1): 6581, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34772938

ABSTRACT

The mammalian SWI/SNF nucleosome remodeler is essential for spermatogenesis. Here, we identify a role for ARID2, a PBAF (Polybromo - Brg1 Associated Factor)-specific subunit, in meiotic division. Arid2cKO spermatocytes arrest at metaphase-I and are deficient in spindle assembly, kinetochore-associated Polo-like kinase1 (PLK1), and centromeric targeting of Histone H3 threonine3 phosphorylation (H3T3P) and Histone H2A threonine120 phosphorylation (H2AT120P). By determining ARID2 and BRG1 genomic associations, we show that PBAF localizes to centromeres and promoters of genes known to govern spindle assembly and nuclear division in spermatocytes. Consistent with gene ontology of target genes, we also identify a role for ARID2 in centrosome stability. Additionally, misexpression of genes such as Aurkc and Ppp1cc (Pp1γ), known to govern chromosome segregation, potentially compromises the function of the chromosome passenger complex (CPC) and deposition of H3T3P, respectively. Our data support a model where-in PBAF activates genes essential for meiotic cell division.


Subject(s)
Chromatin , Mammals/genetics , Meiosis , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/metabolism , Centromere/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , DNA Helicases/metabolism , Gene Silencing , Histones/metabolism , Male , Metaphase , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Transcription Factors/genetics , Transcriptome , Polo-Like Kinase 1
11.
Biol Reprod ; 105(6): 1603-1616, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34518881

ABSTRACT

Aurora A kinase (AURKA) is an important regulator of cell division and is required for assembly of the mitotic spindle. We recently reported the unusual finding that this mitotic kinase is also found on the sperm flagellum. To determine its requirement in spermatogenesis, we generated conditional knockout animals with deletion of the Aurka gene in either spermatogonia or spermatocytes to assess its role in mitotic and postmitotic cells, respectively. Deletion of Aurka in spermatogonia resulted in disappearance of all developing germ cells in the testis, as expected, given its vital role in mitotic cell division. Deletion of Aurka in spermatocytes reduced testis size, sperm count, and fertility, indicating disruption of meiosis or an effect on spermiogenesis in developing mice. Interestingly, deletion of Aurka in spermatocytes increased apoptosis in spermatocytes along with an increase in the percentage of sperm with abnormal morphology. Despite the increase in abnormal sperm, sperm from spermatocyte Aurka knockout mice displayed increased progressive motility. In addition, sperm lysate prepared from Aurka knockout animals had decreased protein phosphatase 1 (PP1) activity. Together, our results show that AURKA plays multiple roles in spermatogenesis, from mitotic divisions of spermatogonia to sperm morphology and motility.


Subject(s)
Aurora Kinase A/genetics , Mice/physiology , Sperm Motility/genetics , Spermatozoa/enzymology , Testis/growth & development , Animals , Aurora Kinase A/deficiency , Aurora Kinase A/metabolism , Male , Mice/genetics , Mice, Knockout , Spermatogenesis/genetics
13.
iScience ; 23(10): 101596, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33083754

ABSTRACT

Spermatogonial stem cells (SSCs) both self-renew and give rise to progenitors that initiate spermatogenic differentiation in the mammalian testis. Questions remain regarding the extent to which the SSC and progenitor states are functionally distinct. Here we provide the first multiparametric integrative analysis of mammalian germ cell epigenomes comparable with that done for >100 somatic cell types by the ENCODE Project. Differentially expressed genes distinguishing SSC- and progenitor-enriched spermatogonia showed distinct histone modification patterns, particularly for H3K27ac and H3K27me3. Motif analysis predicted transcription factors that may regulate spermatogonial subtype-specific fate, and immunohistochemistry and gene-specific chromatin immunoprecipitation analyses confirmed subtype-specific differences in target gene binding of a subset of these factors. Taken together, these results show that SSCs and progenitors display distinct epigenetic profiling consistent with these spermatogonial subtypes being differentially programmed to either self-renew and maintain regenerative capacity as SSCs or lose regenerative capacity and initiate lineage commitment as progenitors.

14.
Biol Reprod ; 103(5): 1132-1143, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32716476

ABSTRACT

Sirolimus, also known as rapamycin, and its closely related rapamycin analog (rapalog) Everolimus inhibit "mammalian target of rapamycin complex 1" (mTORC1), whose activity is required for spermatogenesis. Everolimus is Food and Drug Administration approved for treating human patients to slow growth of aggressive cancers and preventing organ transplant rejection. Here, we test the hypothesis that rapalog inhibition of mTORC1 activity has a negative, but reversible, impact upon spermatogenesis. Juvenile (P20) or adult (P>60) mice received daily injections of sirolimus or Everolimus for 30 days, and tissues were examined at completion of treatment or following a recovery period. Rapalog treatments reduced body and testis weights, testis weight/body weight ratios, cauda epididymal sperm counts, and seminal vesicle weights in animals of both ages. Following rapalog treatment, numbers of differentiating spermatogonia were reduced, with concomitant increases in the ratio of undifferentiated spermatogonia to total number of remaining germ cells. To determine if even low doses of Everolimus can inhibit spermatogenesis, an additional group of adult mice received a dose of Everolimus ∼6-fold lower than a human clinical dose used to treat cancer. In these animals, only testis weights, testis weight/body weight ratios, and tubule diameters were reduced. Return to control values following a recovery period was variable for each of the measured parameters and was duration and dose dependent. Together, these data indicate rapalogs exerted a dose-dependent restriction on overall growth of juvenile and adult mice and negative impact upon spermatogenesis that were largely reversed; following treatment cessation, males from all treatment groups were able to sire offspring.


Subject(s)
Cell Differentiation/drug effects , Everolimus/pharmacology , Fertility/drug effects , Spermatogenesis/drug effects , Spermatogonia/drug effects , Animals , Male , Mice
15.
Mol Reprod Dev ; 86(11): 1462-1484, 2019 11.
Article in English | MEDLINE | ID: mdl-31642147

ABSTRACT

This is a translation and modern interpretation of one of the most important manuscripts on spermatogenesis, published by Victor von Ebner, in 1871. It was originally written in Italian, and to the best of my knowledge has never been translated to English or examined by modern scientists.


Subject(s)
Spermatogenesis , Translating , Animals , Humans
16.
J Biol Chem ; 294(39): 14394-14405, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31399511

ABSTRACT

Docosahexaenoic acid (DHA) is an ω-3 dietary-derived polyunsaturated fatty acid of marine origin enriched in testes and necessary for normal fertility, yet the mechanisms regulating the enrichment of DHA in the testes remain unclear. Long-chain ACSL6 (acyl-CoA synthetase isoform 6) activates fatty acids for cellular anabolic and catabolic metabolism by ligating a CoA to a fatty acid, is highly expressed in testes, and has high preference for DHA. Here, we investigated the role of ACSL6 for DHA enrichment in the testes and its requirement for male fertility. Acsl6-/- males were severely subfertile with smaller testes, reduced cauda epididymal sperm counts, germ cell loss, and disorganization of the seminiferous epithelium. Total fatty acid profiling of Acsl6-/- testes revealed reduced DHA and increased ω-6 arachidonic acid, a fatty acid profile also reflected in phospholipid composition. Strikingly, lipid imaging demonstrated spatial redistribution of phospholipids in Acsl6-/- testes. Arachidonic acid-containing phospholipids were predominantly interstitial in control testes but diffusely localized across Acsl6-/- testes. In control testes, DHA-containing phospholipids were predominantly within seminiferous tubules, which contain Sertoli cells and spermatogenic cells but relocalized to the interstitium in Acsl6-/- testes. Taken together, these data demonstrate that ACSL6 is an initial driving force for germ cell DHA enrichment and is required for normal spermatogenesis and male fertility.


Subject(s)
Coenzyme A Ligases/genetics , Fatty Acids, Omega-6/metabolism , Infertility, Male/genetics , Seminiferous Tubules/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Phospholipids/metabolism , Seminiferous Tubules/cytology , Spermatogenesis
17.
Development ; 146(12)2019 05 13.
Article in English | MEDLINE | ID: mdl-31023878

ABSTRACT

In the mammalian testis, sustained spermatogenesis relies on spermatogonial stem cells (SSCs); their progeny either remain as stem cells (self-renewal) or proliferate and differentiate to enter meiosis in response to retinoic acid (RA). Here, we sought to uncover elusive mechanisms regulating a key switch fundamental to spermatogonial fate: the capacity of spermatogonia to respond to RA. Using the developing mouse testis as a model, we found that spermatogonia and precursor prospermatogonia exhibit a heterogeneous capacity to respond to RA with at least two underlying causes. First, progenitor spermatogonia are prevented from responding to RA by catabolic activity of cytochrome P450 family 26 enzymes. Second, a smaller subset of undifferentiated spermatogonia enriched for SSCs exhibit catabolism-independent RA insensitivity. Moreover, for the first time, we observed that precursor prospermatogonia are heterogeneous and comprise subpopulations that exhibit the same differential RA responsiveness found in neonatal spermatogonia. We propose a novel model by which mammalian prospermatogonial and spermatogonial fates are regulated by their intrinsic capacity to respond (or not) to the differentiation signal provided by RA before, and concurrent with, the initiation of spermatogenesis.


Subject(s)
Gene Expression Regulation , Spermatogenesis , Spermatogonia/cytology , Stem Cells/cytology , Testis/growth & development , Tretinoin/metabolism , Animals , Cell Differentiation , Cell Lineage , Cytochrome P450 Family 26/metabolism , Genomics , Green Fluorescent Proteins/metabolism , Male , Meiosis , Mice , Sertoli Cells/cytology , Signal Transduction , Testis/embryology
18.
Biol Reprod ; 100(2): 429-439, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30202948

ABSTRACT

The self-renewal, proliferation, and differentiation of the spermatogonial populations must be finely coordinated in the mammalian testis, as dysregulation of these processes can lead to subfertility, infertility, or the formation of tumors. There are wide gaps in our understanding of how these spermatogonial populations are formed and maintained, and our laboratory has focused on identifying the molecular and cellular pathways that direct their development. Others and we have shown, using a combination of pharmacologic inhibitors and genetic models, that activation of mTOR complex 1 (mTORC1) is important for spermatogonial differentiation in vivo. Here, we extend those studies to directly test the germ cell-autonomous requirement for mTORC1 in spermatogonial differentiation. We created germ cell conditional knockout mice for "regulatory associated protein of MTOR, complex 1" (Rptor), which encodes an essential component of mTORC1. While germ cell KO mice were viable and healthy, they had smaller testes than littermate controls, and no sperm were present in their cauda epididymides. We found that an initial cohort of Rptor KO spermatogonia proliferated, differentiated, and entered meiosis (which they were unable to complete). However, no self-renewing spermatogonia were formed, and thus the entire germline was lost by adulthood, resulting in Sertoli cell-only testes. These results reveal the cell autonomous requirement for RPTOR in the formation or maintenance of the foundational self-renewing spermatogonial stem cell pool in the mouse testis and underscore complex roles for mTORC1 and its constituent proteins in male germ cell development.


Subject(s)
Adult Germline Stem Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Spermatozoa/physiology , Animals , Epididymis/cytology , Gene Expression Regulation/physiology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Meiosis , Mice , Mice, Knockout , Regulatory-Associated Protein of mTOR/genetics , Spermatogenesis , Testis
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...