Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 760
Filter
1.
Nature ; 626(8001): 1116-1124, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355802

ABSTRACT

Transposable elements (TEs) are a major constituent of human genes, occupying approximately half of the intronic space. During pre-messenger RNA synthesis, intronic TEs are transcribed along with their host genes but rarely contribute to the final mRNA product because they are spliced out together with the intron and rapidly degraded. Paradoxically, TEs are an abundant source of RNA-processing signals through which they can create new introns1, and also functional2 or non-functional chimeric transcripts3. The rarity of these events implies the existence of a resilient splicing code that is able to suppress TE exonization without compromising host pre-mRNA processing. Here we show that SAFB proteins protect genome integrity by preventing retrotransposition of L1 elements while maintaining splicing integrity, via prevention of the exonization of previously integrated TEs. This unique dual role is possible because of L1's conserved adenosine-rich coding sequences that are bound by SAFB proteins. The suppressive activity of SAFB extends to tissue-specific, giant protein-coding cassette exons, nested genes and Tigger DNA transposons. Moreover, SAFB also suppresses LTR/ERV elements in species in which they are still active, such as mice and flies. A significant subset of splicing events suppressed by SAFB in somatic cells are activated in the testis, coinciding with low SAFB expression in postmeiotic spermatids. Reminiscent of the division of labour between innate and adaptive immune systems that fight external pathogens, our results uncover SAFB proteins as an RNA-based, pattern-guided, non-adaptive defence system against TEs in the soma, complementing the RNA-based, adaptive Piwi-interacting RNA pathway of the germline.


Subject(s)
DNA Transposable Elements , Introns , RNA Precursors , RNA Splicing , RNA, Messenger , Animals , Humans , Male , Mice , DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Exons/genetics , Genome/genetics , Introns/genetics , Organ Specificity/genetics , Piwi-Interacting RNA/genetics , Piwi-Interacting RNA/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatids/cytology , Spermatids/metabolism , RNA Splicing/genetics , Testis , Meiosis
2.
Biochem Biophys Res Commun ; 596: 71-75, 2022 03 12.
Article in English | MEDLINE | ID: mdl-35121371

ABSTRACT

The mouse WD repeat and FYVE domain containing 1 (Wdfy1) gene is located in chromosome 1qC4 and spans over 73.7 kilobases. It encodes a protein of 410-amino acid protein that shares 97.8% amino acid sequence identity with the human WDFY1 protein. However, the expression pattern of WDFY1 in reproductive organs and its function in male fertility remain unknown. In this study, we generated transgenic mice expressing FLAG-Wdfy1-mCherry cDNA driven by the Wdfy1 promoter to clarify the expression of WDFY1. The results showed that WDFY1 is highly expressed in mouse testes and located in the cytoplasm of late pachytene spermatocytes to elongated spermatids. Interestingly, the global Wdfy1 knockout (KO) male mice displayed normal growth, development, and fertility. Further histological analysis of Wdfy1 knockout mouse testes revealed that all spermatogenic cells are present in Wdfy1 KO seminiferous tubules. Together, our data demonstrate that WDFY1 is dispensable for mouse spermatogenesis and male fertility.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Fertility/genetics , Gene Expression Regulation , Spermatogenesis/genetics , Testis/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Female , Gene Expression Profiling/methods , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Spermatids/cytology , Spermatids/metabolism , Testis/cytology , WD40 Repeats/genetics
3.
Int J Mol Sci ; 22(23)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34884637

ABSTRACT

The adverse effects of radiation are proportional to the total dose and dose rate. We aimed to investigate the effects of radiation dose rate on different organs in mice. The mice were subjected to low dose rate (LDR, ~3.4 mGy/h) and high dose rate (HDR, ~51 Gy/h) radiation. LDR radiation caused severe tissue toxicity, as observed in the histological analysis of testis. It adversely influenced sperm production, including sperm count and motility, and induced greater sperm abnormalities. The expression of markers of early stage spermatogonial stem cells, such as Plzf, c-Kit, and Oct4, decreased significantly after LDR irradiation, compared to that following exposure of HDR radiation, in qPCR analysis. The compositional ratios of all stages of spermatogonia and meiotic cells, except round spermatid, were considerably reduced by LDR in FACS analysis. Therefore, LDR radiation caused more adverse testicular damage than that by HDR radiation, contrary to the response observed in other organs. Therefore, the dose rate of radiation may have differential effects, depending on the organ; it is necessary to evaluate the effect of radiation in terms of radiation dose, dose rate, organ type, and other conditions.


Subject(s)
Spermatogenesis/radiation effects , Testis/radiation effects , Animals , Dose-Response Relationship, Radiation , Gamma Rays , Male , Mice , Models, Animal , Radiation Dosage , Spermatids/cytology , Spermatids/radiation effects , Spermatogonia/cytology , Spermatogonia/radiation effects , Spermatozoa/cytology , Spermatozoa/radiation effects , Testis/cytology
4.
Stem Cell Reports ; 16(11): 2798-2812, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34653405

ABSTRACT

We propose a new concept that human somatic cells can be converted to become male germline stem cells by the defined factors. Here, we demonstrated that the overexpression of DAZL, DAZ2, and BOULE could directly reprogram human Sertoli cells into cells with the characteristics of human spermatogonial stem cells (SSCs), as shown by their similar transcriptomes and proteomics with human SSCs. Significantly, human SSCs derived from human Sertoli cells colonized and proliferated in vivo, and they could differentiate into spermatocytes and haploid spermatids in vitro. Human Sertoli cell-derived SSCs excluded Y chromosome microdeletions and assumed normal chromosomes. Collectively, human somatic cells could be converted directly to human SSCs with the self-renewal and differentiation potentials and high safety. This study is of unusual significance, because it provides an effective approach for reprogramming human somatic cells into male germ cells and offers invaluable male gametes for treating male infertility.


Subject(s)
Cell Differentiation/genetics , Cell Self Renewal/genetics , Cellular Reprogramming/genetics , RNA-Binding Proteins/genetics , Sertoli Cells/metabolism , Spermatogonia/metabolism , Animals , Cells, Cultured , Gene Expression Profiling/methods , Haploidy , Humans , Male , Mice, Nude , Proteomics/methods , RNA-Binding Proteins/metabolism , Sertoli Cells/cytology , Spermatids/cytology , Spermatids/metabolism , Spermatogonia/cytology , Stem Cell Transplantation/methods , Transplantation, Heterologous
5.
Development ; 148(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34473243

ABSTRACT

CPEB proteins are conserved translation regulators involved in multiple biological processes. One of these proteins in Drosophila, Orb2, is a principal player in spermatogenesis. It is required for meiosis and spermatid differentiation. During the later process, orb2 mRNA and protein are localized within the developing spermatid. To evaluate the role of the orb2 mRNA 3'UTR in spermatogenesis, we used the CRISPR/Cas9 system to generate a deletion of the orb2 3'UTR, orb2R. This deletion disrupts the process of spermatid differentiation but has no apparent effect on meiosis. Differentiation abnormalities include defects in the initial polarization of the 64-cell spermatid cysts, mislocalization of mRNAs and proteins in the elongating spermatid tails, altered morphology of the elongating spermatid tails, and defects in the assembly of the individualization complex. These disruptions in differentiation appear to arise because orb2 mRNA and protein are not properly localized within the 64-cell spermatid cyst.


Subject(s)
3' Untranslated Regions , Drosophila Proteins/genetics , Spermatogenesis , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Animals , Cell Differentiation , Cell Polarity , Drosophila , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Spermatids/cytology , Spermatids/metabolism , Testis/metabolism
6.
PLoS Genet ; 17(6): e1009655, 2021 06.
Article in English | MEDLINE | ID: mdl-34181646

ABSTRACT

During spermatogenesis, the process in which sperm for fertilization are produced from germline cells, gene expression is spatiotemporally highly regulated. In Drosophila, successful expression of extremely large male fertility factor genes on Y-chromosome spanning some megabases due to their gigantic intron sizes is crucial for spermatogenesis. Expression of such extremely large genes must be challenging, but the molecular mechanism that allows it remains unknown. Here we report that a novel RNA-binding protein Maca, which contains two RNA-recognition motifs, is crucial for this process. maca null mutant male flies exhibited a failure in the spermatid individualization process during spermatogenesis, lacked mature sperm, and were completely sterile, while maca mutant female flies were fully fertile. Proteomics and transcriptome analyses revealed that both protein and mRNA abundance of the gigantic male fertility factor genes kl-2, kl-3, and kl-5 (kl genes) are significantly decreased, where the decreases of kl-2 are particularly dramatic, in maca mutant testes. Splicing of the kl-3 transcripts was also dysregulated in maca mutant testes. All these physiological and molecular phenotypes were rescued by a maca transgene in the maca mutant background. Furthermore, we found that in the control genetic background, Maca is exclusively expressed in spermatocytes in testes and enriched at Y-loop A/C in the nucleus, where the kl-5 primary transcripts are localized. Our data suggest that Maca increases transcription processivity, promotes successful splicing of gigantic introns, and/or protects transcripts from premature degradation, of the kl genes. Our study identified a novel RNA-binding protein Maca that is crucial for successful expression of the gigantic male fertility factor genes, spermatogenesis, and male fertility.


Subject(s)
Drosophila melanogaster/genetics , RNA-Binding Proteins/genetics , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Transcriptome , Animals , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Female , Fertility/genetics , Gene Expression Regulation , Gene Ontology , Genes, Reporter , Genetic Complementation Test , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Molecular Sequence Annotation , Mutation , RNA-Binding Proteins/metabolism , Spermatids/cytology , Spermatids/growth & development , Spermatocytes/cytology , Spermatocytes/growth & development , Testis/cytology , Testis/metabolism , Y Chromosome/chemistry
7.
Int J Mol Sci ; 22(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34072207

ABSTRACT

During spermatogenesis, the Golgi apparatus serves important roles including the formation of the acrosome, which is a sperm-specific organelle essential for fertilization. We have previously demonstrated that D. melanogaster ATP-dependent Citrate Lyase (ATPCL) is required for spindle organization, cytokinesis, and fusome assembly during male meiosis, mainly due to is activity on fatty acid biosynthesis. Here, we show that depletion of DmATPCL also affects the organization of acrosome and suggest a role for this enzyme in the assembly of Golgi-derived structures during Drosophila spermatogenesis.


Subject(s)
ATP Citrate (pro-S)-Lyase/genetics , ATP Citrate (pro-S)-Lyase/metabolism , Drosophila/physiology , Golgi Apparatus/metabolism , Meiosis , Animals , Gene Expression , Male , Spermatids/cytology , Spermatids/metabolism , Spermatogenesis/genetics
8.
Nat Commun ; 12(1): 3184, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075040

ABSTRACT

During spermatogenesis, meiosis is accompanied by a robust alteration in gene expression and chromatin status. However, it remains elusive how the meiotic transcriptional program is established to ensure completion of meiotic prophase. Here, we identify a protein complex that consists of germ-cell-specific zinc-finger protein ZFP541 and its interactor KCTD19 as the key transcriptional regulators in mouse meiotic prophase progression. Our genetic study shows that ZFP541 and KCTD19 are co-expressed from pachytene onward and play an essential role in the completion of the meiotic prophase program in the testis. Furthermore, our ChIP-seq and transcriptome analyses identify that ZFP541 binds to and suppresses a broad range of genes whose function is associated with biological processes of transcriptional regulation and covalent chromatin modification. The present study demonstrates that a germ-cell specific complex that contains ZFP541 and KCTD19 promotes the progression of meiotic prophase towards completion in male mice, and triggers the reconstruction of the transcriptional network and chromatin organization leading to post-meiotic development.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Pachytene Stage/genetics , Potassium Channels, Voltage-Gated/metabolism , Spermatids/cytology , Spermatogenesis/genetics , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Chromatin Immunoprecipitation Sequencing , Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Female , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Oocytes/cytology , Oocytes/metabolism , Potassium Channels, Voltage-Gated/genetics , RNA-Seq , Spermatids/metabolism , Transcription Factors/genetics , Transcription, Genetic
9.
Commun Biol ; 4(1): 691, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099857

ABSTRACT

Chromatin of male and female gametes undergoes a number of reprogramming events during the transition from germ cell to embryonic developmental programs. Although the rearrangement of DNA methylation patterns occurring in the zygote has been extensively characterized, little is known about the dynamics of DNA modifications during spermatid maturation. Here, we demonstrate that the dynamics of 5-carboxylcytosine (5caC) correlate with active transcription of LINE-1 retroelements during murine spermiogenesis. We show that the open reading frames of active and evolutionary young LINE-1s are 5caC-enriched in round spermatids and 5caC is eliminated from LINE-1s and spermiogenesis-specific genes during spermatid maturation, being simultaneously retained at promoters and introns of developmental genes. Our results reveal an association of 5caC with activity of LINE-1 retrotransposons suggesting a potential direct role for this DNA modification in fine regulation of their transcription.


Subject(s)
Cytosine/analogs & derivatives , Long Interspersed Nucleotide Elements , Open Reading Frames , Spermatids/metabolism , Animals , Cytosine/metabolism , Male , Mice , Spermatids/cytology , Spermatogenesis , Transcription, Genetic
10.
Nucleic Acids Res ; 49(9): 5106-5123, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33939832

ABSTRACT

The transition from meiotic spermatocytes to postmeiotic haploid germ cells constitutes an essential step in spermatogenesis. The epigenomic regulatory mechanisms underlying this transition remain unclear. Here, we find a prominent transcriptomic switch from the late spermatocytes to the early round spermatids during the meiotic-to-postmeiotic transition, which is associated with robust histone acetylation changes across the genome. Among histone deacetylases (HDACs) and acetyltransferases, we find that HDAC3 is selectively expressed in the late meiotic and early haploid stages. Three independent mouse lines with the testis-specific knockout of HDAC3 show infertility and defects in meiotic exit with an arrest at the late stage of meiosis or early stage of round spermatids. Stage-specific RNA-seq and histone acetylation ChIP-seq analyses reveal that HDAC3 represses meiotic/spermatogonial genes and activates postmeiotic haploid gene programs during meiotic exit, with associated histone acetylation alterations. Unexpectedly, abolishing HDAC3 catalytic activity by missense mutations in the nuclear receptor corepressor (NCOR or SMRT) does not cause infertility, despite causing histone hyperacetylation as HDAC3 knockout, demonstrating that HDAC3 enzyme activity is not required for spermatogenesis. Motif analysis of the HDAC3 cistrome in the testes identified SOX30, which has a similar spatiotemporal expression pattern as HDAC3 during spermatogenesis. Depletion of SOX30 in the testes abolishes the genomic recruitment of the HDAC3 to the binding sites. Collectively, these results establish the SOX30/HDAC3 signaling as a key regulator of the transcriptional program in a deacetylase-independent manner during the meiotic-to-postmeiotic transition in spermatogenesis.


Subject(s)
Fertility/genetics , Gene Expression Regulation , Histone Deacetylases/physiology , Meiosis/genetics , Spermatogenesis/genetics , Transcriptional Activation , Acetylation , Animals , Cellular Reprogramming/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , SOX Transcription Factors/metabolism , Spermatids/cytology , Spermatids/metabolism , Testis/metabolism
11.
PLoS Genet ; 17(5): e1009412, 2021 05.
Article in English | MEDLINE | ID: mdl-33961623

ABSTRACT

Meiosis is a cell division process with complex chromosome events where various molecules must work in tandem. To find meiosis-related genes, we screened evolutionarily conserved and reproductive tract-enriched genes using the CRISPR/Cas9 system and identified potassium channel tetramerization domain containing 19 (Kctd19) as an essential factor for meiosis. In prophase I, Kctd19 deficiency did not affect synapsis or the DNA damage response, and chiasma structures were also observed in metaphase I spermatocytes of Kctd19 KO mice. However, spermatocytes underwent apoptotic elimination during the metaphase-anaphase transition. We were able to rescue the Kctd19 KO phenotype with an epitope-tagged Kctd19 transgene. By immunoprecipitation-mass spectrometry, we confirmed the association of KCTD19 with zinc finger protein 541 (ZFP541) and histone deacetylase 1 (HDAC1). Phenotyping of Zfp541 KO spermatocytes demonstrated XY chromosome asynapsis and recurrent DNA damage in the late pachytene stage, leading to apoptosis. In summary, our study reveals that KCTD19 associates with ZFP541 and HDAC1, and that both KCTD19 and ZFP541 are essential for meiosis in male mice.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Genes, Essential , Meiosis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Anaphase , Animals , CRISPR-Cas Systems/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/deficiency , Chromosomal Proteins, Non-Histone/genetics , Chromosome Pairing , Conserved Sequence , DNA Damage , Evolution, Molecular , Fertility/genetics , Histone Deacetylase 1/metabolism , Male , Meiotic Prophase I , Metaphase , Mice , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Pachytene Stage , Phenotype , Spermatids/cytology , Spermatocytes/cytology , Spermatocytes/metabolism , Testis/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transgenes
12.
Sci Rep ; 11(1): 8422, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33875704

ABSTRACT

The human orthologue of the tumor suppressor protein FBW7 is encoded by the Drosophila archipelago (ago) gene. Ago is an F-box protein that gives substrate specificity to its SCF ubiquitin ligase complex. It has a central role in multiple biological processes in a tissue-specific manner such as cell proliferation, cellular differentiation, hypoxia-induced gene expression. Here we present a previously unknown tissue-specific role of Ago in spermatid differentiation. We identified a classical mutant of ago which is semi-lethal and male-sterile. During the characterization of ago function in testis, we found that ago plays role in spermatid development, following meiosis. We confirmed spermatogenesis defects by silencing ago by RNAi in testes. The ago mutants show multiple abnormalities in elongating and elongated spermatids, including aberration of the cyst morphology, malformed mitochondrial structures, and individualization defects. Additionally, we determined the subcellular localization of Ago protein with mCherry-Ago transgene in spermatids. Our findings highlight the potential roles of Ago in different cellular processes of spermatogenesis, like spermatid individualization, and regulation of mitochondrial morphology.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , F-Box Proteins , Spermatogenesis/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , Genes, Tumor Suppressor , Infertility, Male/genetics , Male , Mitochondria , Mutation , RNA Interference , Spermatids/cytology , Testis/cytology , Testis/metabolism
13.
Sci Rep ; 11(1): 3458, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33568686

ABSTRACT

In vitro spermatogenesis (IVS) using air-liquid interphase organ culture method is possible with mouse testis tissues. The same method, however, has been hardly applicable to animals other than mice, only producing no or limited progression of spermatogenesis. In the present study, we challenged IVS of rats with modifications of culture medium, by supplementing chemical substances, including hormones, antioxidants, and lysophospholipids. In addition, reducing oxygen tension by placing tissues in an incubator of lower oxygen concentration and/or applying silicone cover ceiling on top of the tissue were effective for improving the spermatogenic efficiency. Through these modifications of the culture condition, rat spermatogenesis up to round spermatids was maintained over 70 days in the cultured tissue. Present results demonstrated a significant progress in rat IVS, revealing conditions commonly favorable for mice and rats as well as finding rat-specific optimizations. This is an important step towards successful IVS in many animal species, including humans.


Subject(s)
Organ Culture Techniques , Spermatids/growth & development , Spermatogenesis , Animals , Animals, Genetically Modified , Antioxidants , Culture Media , Hormones , Male , Meiosis , Oxygen/analysis , Rats , Spermatids/cytology , Spermatocytes/growth & development
14.
J Vis Exp ; (167)2021 01 14.
Article in English | MEDLINE | ID: mdl-33522502

ABSTRACT

Isolation of meiotic spermatocytes is essential to investigate molecular mechanisms underlying meiosis and spermatogenesis. Although there are established cell isolation protocols using Hoechst 33342 staining in combination with fluorescence-activated cell sorting, it requires cell sorters equipped with an ultraviolet laser. Here we describe a cell isolation protocol using the DyeCycle Violet (DCV) stain, a low cytotoxicity DNA binding dye structurally similar to Hoechst 33342. DCV can be excited by both ultraviolet and violet lasers, which improves the flexibility of equipment choice, including a cell sorter not equipped with an ultraviolet laser. Using this protocol, one can isolate three live-cell subpopulations in meiotic prophase I, including leptotene/zygotene, pachytene, and diplotene spermatocytes, as well as post-meiotic round spermatids. We also describe a protocol to prepare single-cell suspension from mouse testes. Overall, the procedure requires a short time to complete (4-5 hours depending on the number of needed cells), which facilitates many downstream applications.


Subject(s)
Cell Membrane Permeability , Cell Separation/methods , DNA/metabolism , Spermatocytes/cytology , Spermatogenesis , Animals , Benzimidazoles/metabolism , Cell Survival , Dissection , Flow Cytometry , Fluorescence , Male , Meiosis , Mice , Pachytene Stage , Scattering, Radiation , Spermatids/cytology , Staining and Labeling , Testis/cytology
15.
STAR Protoc ; 2(1): 100254, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33490974

ABSTRACT

Post-meiotic spermatids become spermatozoa through developmental stages during spermiogenesis. Isolation of spermatid fractions is required to examine the change of protein expression during spermiogenesis. Here, we present a simple method to isolate spermatid fractions from mouse testes using unit gravity sedimentation in a BSA density gradient. Isolation of spermatid fractions can be used to analyze changes of transcript or protein during spermiogenesis. For complete details on the use and execution of this protocol, please refer to Kim et al. (2020).


Subject(s)
Cell Separation , Spermatids/cytology , Testis/cytology , Animals , Male , Mice , Spermatids/metabolism , Spermatogenesis , Testis/metabolism
16.
Endocrinology ; 162(4)2021 04 01.
Article in English | MEDLINE | ID: mdl-33453102

ABSTRACT

Throughout spermatogenesis, cellular cargoes including haploid spermatids are required to be transported across the seminiferous epithelium, either toward the microtubule (MT) plus (+) end near the basement membrane at stage V, or to the MT minus (-) end near the tubule lumen at stages VI to VIII of the epithelial cycle. Furthermore, preleptotene spermatocytes, differentiated from type B spermatogonia, are transported across the Sertoli cell blood-testis barrier (BTB) to enter the adluminal compartment. Few studies, however, have been conducted to explore the function of MT-dependent motor proteins to support spermatid transport during spermiogenesis. Herein, we examined the role of MT-dependent and microtubule plus (+) end-directed motor protein kinesin 15 (KIF15) in the testis. KIF15 displayed a stage-specific expression across the seminiferous epithelium, associated with MTs, and appeared as aggregates on the MT tracks that aligned perpendicular to the basement membrane and laid across the entire epithelium. KIF15 also tightly associated with apical ectoplasmic specialization, displaying strict stage-specific distribution, apparently to support spermatid transport across the epithelium. We used a loss-of-function approach by RNAi to examine the role of KIF15 in Sertoli cell epithelium in vitro to examine its role in cytoskeletal-dependent Sertoli cell function. It was noted that KIF15 knockdown by RNAi that reduced KIF15 expression by ~70% in Sertoli cells with an established functional tight junction barrier impeded the barrier function. This effect was mediated through remarkable changes in the cytoskeletal organization of MTs, but also actin-, vimentin-, and septin-based cytoskeletons, illustrating that KIF15 exerts its regulatory effects well beyond microtubules.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Kinesins/metabolism , Microtubules/metabolism , Sertoli Cells/metabolism , Spermatids/metabolism , Spermatogenesis , Vimentin/metabolism , Actins/genetics , Animals , Blood-Testis Barrier/metabolism , Kinesins/genetics , Male , Microtubules/genetics , Rats , Sertoli Cells/cytology , Spermatids/cytology , Vimentin/genetics
17.
Biochim Biophys Acta Gen Subj ; 1865(5): 129845, 2021 05.
Article in English | MEDLINE | ID: mdl-33476744

ABSTRACT

BACKGROUND: Mitochondrial fission counterbalances fusion to maintain organelle morphology, but its role during development remains poorly characterized. Mammalian spermatogenesis is a complex developmental process involving several drastic changes to mitochondrial shape and organization. Mitochondria are generally small and spherical in spermatogonia, elongate during meiosis, and fragment in haploid round spermatids. Near the end of spermatid maturation, small mitochondrial spheres line the axoneme, elongate, and tightly wrap around the midpiece to form the mitochondrial sheath, which is critical for fueling flagellar movements. It remains unclear how these changes in mitochondrial morphology are regulated and how they affect sperm development. METHODS: We used genetic ablation of Mff (mitochondrial fission factor) in mice to investigate the role of mitochondrial fission during mammalian spermatogenesis. RESULTS: Our analysis indicates that Mff is required for mitochondrial fragmentation in haploid round spermatids and for organizing mitochondria in the midpiece in elongating spermatids. In Mff mutant mice, round spermatids have aberrantly elongated mitochondria that often show central constrictions, suggestive of failed fission events. In elongating spermatids and spermatozoa, mitochondrial sheaths are disjointed, containing swollen mitochondria with large gaps between organelles. These mitochondrial abnormalities in Mff mutant sperm are associated with reduced respiratory chain Complex IV activity, aberrant sperm morphology and motility, and reduced fertility. CONCLUSIONS: Mff is required for organization of the mitochondrial sheath in mouse sperm. GENERAL SIGNIFICANCE: Mitochondrial fission plays an important role in regulating mitochondrial organization during a complex developmental process.


Subject(s)
Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Spermatids/metabolism , Animals , Female , Fertilization in Vitro , Male , Mice, Inbred C57BL , Mitochondrial Dynamics , Sperm Motility , Spermatids/cytology , Spermatogenesis
18.
J Morphol ; 282(4): 500-510, 2021 04.
Article in English | MEDLINE | ID: mdl-33459394

ABSTRACT

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


Subject(s)
Cytoplasm/metabolism , Octopodiformes/physiology , Spermatids/growth & development , Spermatogenesis , Animals , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Male , Microtubules/metabolism , Microtubules/ultrastructure , Octopodiformes/ultrastructure , Seminiferous Tubules/cytology , Spermatids/cytology , Spermatids/ultrastructure , Spermatozoa/cytology , Spermatozoa/ultrastructure , Testis/cytology
19.
Tissue Cell ; 68: 101457, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33212325

ABSTRACT

Avian species comprise more than half of all vertebrates yet there is a dearth of information regarding spermatid development in this class of animals. This report of spermiogenesis in the cattle egret, Bubulcus ibis, is the first in the order Pelecaniformes. Five sexually mature and reproductively active male cattle egrets were captured in the wild, humanely euthanized, the reproductive organs dissected out, and tissues from the testes routinely prepared for transmission electron microscopy. Twelve steps of spermatid development, using the step-wise system, were determined. Acrosomogenesis in the egret results in a relatively short, solid, bullet-shaped acrosome that ends bluntly anteriorly and flat posteriorly or basally. The nucleus displays remarkable morphological changes, with the anterior end of the mature spermatid becoming flat, lacking a rostrum and an endonuclear canal. A perforatorium does not develop. It is noteworthy that a longitudinal, but not a circular, manchette develops during spermiogenesis in this bird. The proximal centriole is attached to the nucleus, at the implantation fossa, by means of well-formed, electron dense struts of material. An amorphous fibrous sheath develops in the principal piece. The interesting development and peculiar features of the acrosome and nucleus, as well as the absence of the perforatorium and circular manchette in the spermatozoon of the cattle egret, may be of phylogenetic significance.


Subject(s)
Birds/physiology , Cattle/parasitology , Spermatogenesis , Animals , Male , Spermatids/cytology , Spermatids/ultrastructure
20.
Cell Tissue Res ; 383(3): 1167-1182, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33237480

ABSTRACT

The acrosome is a special organelle that develops from the Golgi apparatus and the endolysosomal compartment in the spermatids. Centromere protein E (CENP-E) is an essential kinesin motor in chromosome congression and alignment. This study is aimed at investigating the roles and mechanisms of kinesin-7 CENP-E in the formation of the acrosome during spermatogenesis. Male ICR mice are injected with GSK923295 for long-term inhibition of CENP-E. Chemical inhibition and siRNA-mediated knockdown of CENP-E are carried out in the GC-2 spd cells. The morphology of the acrosomes is determined by the HE staining, immunofluorescence, and transmission electron microscopy. We have identified CENP-E is a key factor in the formation and structural maintenance of the acrosome during acrosome biogenesis. Long-term inhibition of CENP-E by GSK923295 results in the asymmetric acrosome and the dispersed acrosome. CENP-E depletion leads to the malformation of the Golgi complex and abnormal targeting of the PICK1- and PIST-positive Golgi-associated vesicles. Our findings uncover an essential role of CENP-E in membrane trafficking and structural organization of the acrosome in the spermatids during spermatogenesis. Our results shed light on the molecular mechanisms involved in vesicle trafficking and architecture maintenance of the acrosome.


Subject(s)
Acrosome/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Golgi Apparatus/metabolism , Kinesins/metabolism , Spermatids , Spermatogenesis , Animals , Cell Line , Male , Mice , Mice, Inbred ICR , Protein Transport , Spermatids/cytology , Spermatids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...