1
|
Meng X, Yamashita YM. Intrinsically weak sex chromosome drive through sequential asymmetric meiosis. SCIENCE ADVANCES 2025; 11:eadv7089. [PMID: 40333966 PMCID: PMC12057659 DOI: 10.1126/sciadv.adv7089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/01/2025] [Indexed: 05/09/2025]
Abstract
Meiotic drivers are selfish genetic elements that bias their own transmission, violating Mendel's Law of Equal Segregation. It has long been recognized that sex chromosome-linked drivers present a paradox: Their success in transmission can severely distort populations' sex ratio and lead to extinction. This paradox is typically solved by the presence of suppressors or fitness costs associated with the driver, limiting the propagation of the driver. Here, we show that Stellate (Ste) in Drosophila melanogaster represents a novel class of X chromosome-linked driver that operates with an inherent mechanism that weakens its drive strength. Ste protein asymmetrically segregates into Y-bearing cells during meiosis I, subsequently causing their death. Unexpectedly, Ste segregates asymmetrically again during meiosis II, sparing half of the Y-bearing spermatids from Ste-induced defects, thereby weakening the drive strength. Our findings reveal a mechanism by which sex chromosome drivers avoid suicidal success.
Collapse
Affiliation(s)
- Xuefeng Meng
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Yukiko M. Yamashita
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| |
Collapse
|
2
|
Beard EK, Norris RP, Furusho M, Terasaki M, Inaba M. Soma-to-germline BMP signal is essential for Drosophila spermiogenesis. Dev Biol 2025; 517:140-147. [PMID: 39362354 PMCID: PMC12033009 DOI: 10.1016/j.ydbio.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/20/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
In the Drosophila testis, developing germ cells are encapsulated by somatic support cells throughout development. Soma-germline interactions are essential for successful spermiogenesis. However, it is still not fully understood what signaling events take place between the soma and the germline. In this study, we found that a Bone Morphogenetic Protein (BMP) ligand, Glass bottom boat (Gbb), secreted from somatic cyst cells (CCs), signals to differentiating germ cells to maintain proper spermiogenesis. Knockdown of Gbb in CCs or the type I BMP receptor Saxophone (Sax) in germ cells leads to a defect in sperm head bundling and decreased fertility. Our Transmission Electron Microscopy (TEM) analyses revealed that the mutant germ cells have aberrant morphology of mitochondria throughout the stages of spermiogenesis and exhibit a defect in nebenkern formation. Elongating spermatids show uncoupled nuclei and elongating mitochondrial derivatives, suggesting that improper mitochondrial development may cause sperm bundling defects. Taken together, we propose a new role of soma-derived BMP signaling, which is essential for spermiogenesis.
Collapse
Affiliation(s)
- Emma Kristine Beard
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06032, USA
| | - Rachael P Norris
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06032, USA
| | - Miki Furusho
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06032, USA
| | - Mark Terasaki
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06032, USA
| | - Mayu Inaba
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, 06032, USA.
| |
Collapse
|
3
|
Meng FW, Murphy PJ. Canonical and Variant Nucleosome Reprogramming from Sperm to Blastula. Methods Mol Biol 2025; 2923:17-32. [PMID: 40418441 DOI: 10.1007/978-1-0716-4522-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Epigenetic reprogramming of chromatin, mediated through modifications to canonical histones and the incorporation of histone variants, plays a central role in the establishment of cell identity and regulation of gene expression programs. Such epigenetic programming becomes particularly important during the development of highly specialized cell types such as germ cells-sperm and oocytes, and stem cells within the blastula of early embryos. Determining patterns of epigenetic modifications within these cell types can be particularly challenging due to technical limitations associated with limited starting materials. Here we present a brief overview of studies focused on epigenetic reprogramming during the transition from sperm to blastula stage embryos and compare between several diverse model systems including Drosophila, zebrafish and mammals. Furthermore, we discuss and compare prevailing genomic profiling approaches, such as ChIP-Seq, CUT&Tag, and CUT&RUN, for performing genome-wide epigenomic analysis.
Collapse
Affiliation(s)
- Fanju W Meng
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Patrick J Murphy
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
4
|
Kitaoka M, Yamashita YM. Running the gauntlet: challenges to genome integrity in spermiogenesis. Nucleus 2024; 15:2339220. [PMID: 38594652 PMCID: PMC11005813 DOI: 10.1080/19491034.2024.2339220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Species' continuity depends on gametogenesis to produce the only cell types that can transmit genetic information across generations. Spermiogenesis, which encompasses post-meiotic, haploid stages of male gametogenesis, is a process that leads to the formation of sperm cells well-known for their motility. Spermiogenesis faces three major challenges. First, after two rounds of meiotic divisions, the genome lacks repair templates (no sister chromatids, no homologous chromosomes), making it incredibly vulnerable to any genomic insults over an extended time (typically days-weeks). Second, the sperm genome becomes transcriptionally silent, making it difficult to respond to new perturbations as spermiogenesis progresses. Third, the histone-to-protamine transition, which is essential to package the sperm genome, counterintuitively involves DNA break formation. How spermiogenesis handles these challenges remains poorly understood. In this review, we discuss each challenge and their intersection with the biology of protamines. Finally, we discuss the implication of protamines in the process of evolution.
Collapse
Affiliation(s)
- Maiko Kitaoka
- Whitehead Institute for Biomedical Research and Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Yukiko M. Yamashita
- Whitehead Institute for Biomedical Research and Howard Hughes Medical Institute, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
5
|
McSwiggin H, Magalhães R, Nilsson EE, Yan W, Skinner MK. Epigenetic transgenerational inheritance of toxicant exposure-specific non-coding RNA in sperm. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae014. [PMID: 39494159 PMCID: PMC11529619 DOI: 10.1093/eep/dvae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 08/23/2024] [Accepted: 09/03/2024] [Indexed: 11/05/2024]
Abstract
Environmentally induced epigenetic transgenerational inheritance of phenotypic variation and disease susceptibility requires the germ cell (sperm or egg) transmission of integrated epigenetic mechanisms involving DNA methylation, histone modifications, and non-coding RNA (ncRNA) actions. Previous studies have demonstrated that transgenerational exposure and disease-specific differential DNA methylation regions (DMRs) in sperm are observed and that ncRNA-mediated DNA methylation occurs. The current study was designed to determine if transgenerational exposure-specific ncRNAs exist in sperm. Specifically, toxicants with distinct mechanisms of action including the fungicide vinclozolin (anti-androgenic), pesticide dichlorodiphenyltrichloroethane (estrogenic), herbicide atrazine (endocrine disruptor at cyclic adenosine monophosphate level), and hydrocarbon mixture jet fuel (JP8) (aryl hydrocarbon receptor disruptor) were used to promote transgenerational disease phenotypes in F3 generation outbred rats. New aliquots of sperm, previously collected and used for DNA methylation analyses, were used in the current study for ncRNA sequencing analyses of nuclear RNA. Significant changes in transgenerational sperm ncRNA were observed for each transgenerational exposure lineage. The majority of ncRNA was small noncoding RNAs including piwi-interacting RNA, tRNA-derived small RNAs, microRNAs, rRNA-derived small RNA, as well as long ncRNAs. Although there was some overlap among the different classes of ncRNA across the different exposures, the majority of differentially expressed ncRNAs were exposure-specific with no overlapping ncRNA between the four different exposure lineages in the transgenerational F3 generation sperm nuclear ncRNAs. The ncRNA chromosomal locations and gene associations were identified for a small number of differential expressed ncRNA. Interestingly, an overlap analysis between the transgenerational sperm DMRs and ncRNA chromosomal locations demonstrated small populations of overlapping ncRNA, but a large population of non-overlapping ncRNAs. Observations suggest that transgenerational sperm ncRNAs have both exposure-specific populations within the different classes of ncRNA, as well as some common populations of ncRNAs among the different exposures. The lack of co-localization of many of the ncRNAs with previously identified transgenerational DMRs suggests a distal integration of the different epigenetic mechanisms. The potential use of ncRNA analyses for transgenerational toxicant exposure assessment appears feasible.
Collapse
Affiliation(s)
- Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, United States
| | - Rubens Magalhães
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, United States
| | - Eric E Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, United States
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, United States
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, United States
| |
Collapse
|
6
|
Khan S, Mishra RK. Multigenerational Effect of Heat Stress on the Drosophila melanogaster Sperm Proteome. J Proteome Res 2024; 23:2265-2278. [PMID: 38743012 DOI: 10.1021/acs.jproteome.4c00205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The effect of the parental environment on offspring through non-DNA sequence-based mechanisms, such as DNA methylation, chromatin modifications, noncoding RNAs, and proteins, could only be established after the conception of "epigenetics". These effects are now broadly referred to as multigenerational epigenetic effects. Despite accumulating evidence of male gamete-mediated multigenerational epigenetic inheritance, little is known about the factors that underlie heat stress-induced multigenerational epigenetic inheritance via the male germline in Drosophila. In this study, we address this gap by utilizing an established heat stress paradigm in Drosophila and investigating its multigenerational effect on the sperm proteome. Our findings indicate that multigenerational heat stress during the early embryonic stage significantly influences proteins in the sperm associated with translation, chromatin organization, microtubule-based processes, and the generation of metabolites and energy. Assessment of life-history traits revealed that reproductive fitness and stress tolerance remained unaffected by multigenerational heat stress. Our study offers initial insights into the chromatin-based epigenetic mechanisms as a plausible means of transmitting heat stress memory through the male germline in Drosophila. Furthermore, it sheds light on the repercussions of early embryonic heat stress on male reproductive potential. The data sets from this study are available at the ProteomeXchange Consortium under the identifier PXD037488.
Collapse
Affiliation(s)
- Shagufta Khan
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad-500 007, Telangana, India
| | - Rakesh K Mishra
- CSIR - Centre for Cellular and Molecular Biology, Hyderabad-500 007, Telangana, India
- Tata Institute for Genetics and Society, Bengaluru-560 065, Karnataka, India
| |
Collapse
|
7
|
Ridges JT, Bladen J, King TD, Brown NC, Large CRL, Cooper JC, Jones AJ, Loppin B, Dubruille R, Phadnis N. Overdrive is essential for targeted sperm elimination by Segregation Distorter. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597441. [PMID: 38895353 PMCID: PMC11185633 DOI: 10.1101/2024.06.04.597441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Intra-genomic conflict driven by selfish chromosomes is a powerful force that shapes the evolution of genomes and species. In the male germline, many selfish chromosomes bias transmission in their own favor by eliminating spermatids bearing the competing homologous chromosomes. However, the mechanisms of targeted gamete elimination remain mysterious. Here, we show that Overdrive (Ovd), a gene required for both segregation distortion and male sterility in Drosophila pseudoobscura hybrids, is broadly conserved in Dipteran insects but dispensable for viability and fertility. In D. melanogaster, Ovd is required for targeted Responder spermatid elimination after the histone-to-protamine transition in the classical Segregation Distorter system. We propose that Ovd functions as a general spermatid quality checkpoint that is hijacked by independent selfish chromosomes to eliminate competing gametes.
Collapse
Affiliation(s)
- Jackson T. Ridges
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Jackson Bladen
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Thomas D. King
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Nora C. Brown
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | | | - Jacob C. Cooper
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Amanda J. Jones
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Benjamin Loppin
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Raphaëlle Dubruille
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Nitin Phadnis
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
8
|
Kaur R, McGarry A, Shropshire JD, Leigh BA, Bordenstein SR. Prophage proteins alter long noncoding RNA and DNA of developing sperm to induce a paternal-effect lethality. Science 2024; 383:1111-1117. [PMID: 38452081 PMCID: PMC11187695 DOI: 10.1126/science.adk9469] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024]
Abstract
The extent to which prophage proteins interact with eukaryotic macromolecules is largely unknown. In this work, we show that cytoplasmic incompatibility factor A (CifA) and B (CifB) proteins, encoded by prophage WO of the endosymbiont Wolbachia, alter long noncoding RNA (lncRNA) and DNA during Drosophila sperm development to establish a paternal-effect embryonic lethality known as cytoplasmic incompatibility (CI). CifA is a ribonuclease (RNase) that depletes a spermatocyte lncRNA important for the histone-to-protamine transition of spermiogenesis. Both CifA and CifB are deoxyribonucleases (DNases) that elevate DNA damage in late spermiogenesis. lncRNA knockdown enhances CI, and mutagenesis links lncRNA depletion and subsequent sperm chromatin integrity changes to embryonic DNA damage and CI. Hence, prophage proteins interact with eukaryotic macromolecules during gametogenesis to create a symbiosis that is fundamental to insect evolution and vector control.
Collapse
Affiliation(s)
- Rupinder Kaur
- Pennsylvania State University, Departments of Biology and Entomology, University Park, PA 16802, USA
- One Health Microbiome Center, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
- Vanderbilt University, Department of Biological Sciences, Nashville, TN 37235, USA
| | - Angelina McGarry
- Pennsylvania State University, Departments of Biology and Entomology, University Park, PA 16802, USA
- One Health Microbiome Center, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - J. Dylan Shropshire
- Vanderbilt University, Department of Biological Sciences, Nashville, TN 37235, USA
- Department of Biological Sciences, Lehigh University, Bethlehem, PA 18015, USA
| | - Brittany A. Leigh
- Vanderbilt University, Department of Biological Sciences, Nashville, TN 37235, USA
| | - Seth R. Bordenstein
- Pennsylvania State University, Departments of Biology and Entomology, University Park, PA 16802, USA
- One Health Microbiome Center, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
- Vanderbilt University, Department of Biological Sciences, Nashville, TN 37235, USA
| |
Collapse
|
9
|
Kaur R, Meier CJ, McGraw EA, Hillyer JF, Bordenstein SR. The mechanism of cytoplasmic incompatibility is conserved in Wolbachia-infected Aedes aegypti mosquitoes deployed for arbovirus control. PLoS Biol 2024; 22:e3002573. [PMID: 38547237 PMCID: PMC11014437 DOI: 10.1371/journal.pbio.3002573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 04/12/2024] [Accepted: 03/01/2024] [Indexed: 04/13/2024] Open
Abstract
The rising interest and success in deploying inherited microorganisms and cytoplasmic incompatibility (CI) for vector control strategies necessitate an explanation of the CI mechanism. Wolbachia-induced CI manifests in the form of embryonic lethality when sperm from Wolbachia-bearing testes fertilize eggs from uninfected females. Embryos from infected females however survive to sustain the maternally inherited symbiont. Previously in Drosophila melanogaster flies, we demonstrated that CI modifies chromatin integrity in developing sperm to bestow the embryonic lethality. Here, we validate these findings using wMel-transinfected Aedes aegypti mosquitoes released to control vector-borne diseases. Once again, the prophage WO CI proteins, CifA and CifB, target male gametic nuclei to modify chromatin integrity via an aberrant histone-to-protamine transition. Cifs are not detected in the embryo, and thus elicit CI via the nucleoprotein modifications established pre-fertilization. The rescue protein CifA in oogenesis localizes to stem cell, nurse cell, and oocyte nuclei, as well as embryonic DNA during embryogenesis. Discovery of the nuclear targeting Cifs and altered histone-to-protamine transition in both Aedes aegypti mosquitoes and D. melanogaster flies affirm the Host Modification Model of CI is conserved across these host species. The study also newly uncovers the cell biology of Cif proteins in the ovaries, CifA localization in the embryos, and an impaired histone-to-protamine transition during spermiogenesis of any mosquito species. Overall, these sperm modification findings may enable future optimization of CI efficacy in vectors or pests that are refractory to Wolbachia transinfections.
Collapse
Affiliation(s)
- Rupinder Kaur
- Pennsylvania State University, Departments of Biology and Entomology, University Park, Pennsylvania, United States of America
- Pennsylvania State University, One Health Microbiome Center, Huck Institutes of the Life Sciences, University Park, Pennsylvania, United States of America
- Vanderbilt University, Department of Biological Sciences, Nashville, Tennessee, United States of America
| | - Cole J. Meier
- Vanderbilt University, Department of Biological Sciences, Nashville, Tennessee, United States of America
| | - Elizabeth A. McGraw
- Pennsylvania State University, Departments of Biology and Entomology, University Park, Pennsylvania, United States of America
- Pennsylvania State University, One Health Microbiome Center, Huck Institutes of the Life Sciences, University Park, Pennsylvania, United States of America
- Pennsylvania State University, Center for Infectious Disease Dynamics, Huck Institutes of the Life Sciences, University Park, Pennsylvania, United States of America
| | - Julian F. Hillyer
- Vanderbilt University, Department of Biological Sciences, Nashville, Tennessee, United States of America
| | - Seth R. Bordenstein
- Pennsylvania State University, Departments of Biology and Entomology, University Park, Pennsylvania, United States of America
- Pennsylvania State University, One Health Microbiome Center, Huck Institutes of the Life Sciences, University Park, Pennsylvania, United States of America
- Vanderbilt University, Department of Biological Sciences, Nashville, Tennessee, United States of America
| |
Collapse
|
10
|
Hu Q, Xiao Y, Wei R, Tang T, Wen L, Lu Y, Yu XQ. Identification and functional analysis of CG3526 in spermatogenesis of Drosophila melanogaster. INSECT SCIENCE 2024; 31:79-90. [PMID: 37465843 DOI: 10.1111/1744-7917.13243] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 07/20/2023]
Abstract
Spermatogenesis is a critical part of reproduction in insects; however, its molecular mechanism is still largely unknown. In this study, we identified a testis-specific gene CG3526 in Drosophila melanogaster. Bioinformatics analysis showed that CG3526 contains a zinc binding domain and 2 C2 H2 type zinc fingers, and it is clustered to the vertebrate really interesting new gene (RING) family E3 ubiquitin-protein ligases. When CG3526 was knocked down by RNA interference (RNAi), the testis became much smaller in size, and the apical tip exhibited a sharp and thin end instead of the blunt and round shape in the control testis. More importantly, compared to the control flies, only a few mature sperm were present in the seminal vesicle of C587-Gal4 > CG3526 RNAi flies. Immunofluorescence staining of the testis from CG3526 RNAi flies showed that the homeostasis of testis stem cell niche was disrupted, cell distribution in the apical tip was scattered, and the process of spermatogenesis was not completed. Furthermore, we found that the phenotype of CG3526 RNAi flies' testis was similar to that of testis of Stat92E RNAi flies, the expression level of CG3526 was significantly downregulated in the Stat92EF06346 mutant flies, and the promoter activity of CG3526 was upregulated by STAT92E. Taken together, our results indicated that CG3526 is a downstream effector gene in the JAK-STAT signaling pathway that plays a key role in the spermatogenesis of Drosophila.
Collapse
Affiliation(s)
- Qihao Hu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yanhong Xiao
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Runnan Wei
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Ting Tang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Liang Wen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yuzhen Lu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| |
Collapse
|
11
|
Zhang S, Wang R, Zhu X, Zhang L, Liu X, Sun L. Characteristics and expression of lncRNA and transposable elements in Drosophila aneuploidy. iScience 2023; 26:108494. [PMID: 38125016 PMCID: PMC10730892 DOI: 10.1016/j.isci.2023.108494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/28/2023] [Accepted: 11/16/2023] [Indexed: 12/23/2023] Open
Abstract
Aneuploidy can globally affect the expression of the whole genome, which is detrimental to organisms. Dosage-sensitive regulators usually have multiple intermolecular interactions, and changes in their stoichiometry are responsible for the dysregulation of the regulatory network. Currently, studies on noncoding genes in aneuploidy are relatively rare. We studied the characteristics and expression profiles of long noncoding RNAs (lncRNAs) and transposable elements (TEs) in aneuploid Drosophila. It is found that lncRNAs and TEs are affected by genomic imbalance and appear to be more sensitive to an inverse dosage effect than mRNAs. Several dosage-sensitive lncRNAs and TEs were detected for their expression patterns during embryogenesis, and their biological functions in the ovary and testes were investigated using tissue-specific RNAi. This study advances our understanding of the noncoding sequences in imbalanced genomes and provides a novel perspective for the study of aneuploidy-related human diseases such as cancer.
Collapse
Affiliation(s)
- Shuai Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Ruixue Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Xilin Zhu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Ludan Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Xinyu Liu
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Lin Sun
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing 100875, China
- Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Science, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
12
|
Anderson JT, Henikoff S, Ahmad K. Chromosome-specific maturation of the epigenome in the Drosophila male germline. eLife 2023; 12:RP89373. [PMID: 38032818 PMCID: PMC10688970 DOI: 10.7554/elife.89373] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
Spermatogenesis in the Drosophila male germline proceeds through a unique transcriptional program controlled both by germline-specific transcription factors and by testis-specific versions of core transcriptional machinery. This program includes the activation of genes on the heterochromatic Y chromosome, and reduced transcription from the X chromosome, but how expression from these sex chromosomes is regulated has not been defined. To resolve this, we profiled active chromatin features in the testes from wildtype and meiotic arrest mutants and integrate this with single-cell gene expression data from the Fly Cell Atlas. These data assign the timing of promoter activation for genes with germline-enriched expression throughout spermatogenesis, and general alterations of promoter regulation in germline cells. By profiling both active RNA polymerase II and histone modifications in isolated spermatocytes, we detail widespread patterns associated with regulation of the sex chromosomes. Our results demonstrate that the X chromosome is not enriched for silencing histone modifications, implying that sex chromosome inactivation does not occur in the Drosophila male germline. Instead, a lack of dosage compensation in spermatocytes accounts for the reduced expression from this chromosome. Finally, profiling uncovers dramatic ubiquitinylation of histone H2A and lysine-16 acetylation of histone H4 across the Y chromosome in spermatocytes that may contribute to the activation of this heterochromatic chromosome.
Collapse
Affiliation(s)
- James T Anderson
- Basic Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Kami Ahmad
- Basic Sciences Division, Fred Hutchinson Cancer CenterSeattleUnited States
| |
Collapse
|
13
|
Dubruille R, Herbette M, Revel M, Horard B, Chang CH, Loppin B. Histone removal in sperm protects paternal chromosomes from premature division at fertilization. Science 2023; 382:725-731. [PMID: 37943933 PMCID: PMC11180706 DOI: 10.1126/science.adh0037] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/22/2023] [Indexed: 11/12/2023]
Abstract
The global replacement of histones with protamines in sperm chromatin is widespread in animals, including insects, but its actual function remains enigmatic. We show that in the Drosophila paternal effect mutant paternal loss (pal), sperm chromatin retains germline histones H3 and H4 genome wide without impairing sperm viability. However, after fertilization, pal sperm chromosomes are targeted by the egg chromosomal passenger complex and engage into a catastrophic premature division in synchrony with female meiosis II. We show that pal encodes a rapidly evolving transition protein specifically required for the eviction of (H3-H4)2 tetramers from spermatid DNA after the removal of H2A-H2B dimers. Our study thus reveals an unsuspected role of histone eviction from insect sperm chromatin: safeguarding the integrity of the male pronucleus during female meiosis.
Collapse
Affiliation(s)
- Raphaälle Dubruille
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Marion Herbette
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Maxime Revel
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Béatrice Horard
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Ching-Ho Chang
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Benjamin Loppin
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
14
|
Anderson J, Henikoff S, Ahmad K. Chromosome-specific maturation of the epigenome in the Drosophila male germline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529909. [PMID: 37873332 PMCID: PMC10592605 DOI: 10.1101/2023.02.24.529909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Spermatogenesis in the Drosophila male germline proceeds through a unique transcriptional program controlled both by germline-specific transcription factors and by testis-specific versions of core transcriptional machinery. This program includes the activation of genes on the heterochromatic Y chromosome, and reduced transcription from the X chromosome, but how expression from these sex chromosomes is regulated has not been defined. To resolve this, we profiled active chromatin features in the testes from wildtype and meiotic arrest mutants and integrate this with single-cell gene expression data from the Fly Cell Atlas. These data assign the timing of promoter activation for genes with germline-enriched expression throughout spermatogenesis, and general alterations of promoter regulation in germline cells. By profiling both active RNA polymerase II and histone modifications in isolated spermatocytes, we detail widespread patterns associated with regulation of the sex chromosomes. Our results demonstrate that the X chromosome is not enriched for silencing histone modifications, implying that sex chromosome inactivation does not occur in the Drosophila male germline. Instead, a lack of dosage compensation in spermatocytes accounts for the reduced expression from this chromosome. Finally, profiling uncovers dramatic ubiquitinylation of histone H2A and lysine-16 acetylation of histone H4 across the Y chromosome in spermatocytes that may contribute to the activation of this heterochromatic chromosome.
Collapse
Affiliation(s)
- James Anderson
- Basic Sciences Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, USA
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, USA
- Howard Hughes Medical Institute; Chevy Chase, MD, USA
| | - Kami Ahmad
- Basic Sciences Division, Fred Hutchinson Cancer Center; Seattle, WA, 98109, USA
| |
Collapse
|
15
|
Emelyanov AV, Barcenilla-Merino D, Loppin B, Fyodorov DV. APOLLO, a testis-specific Drosophila ortholog of importin-4, mediates the loading of protamine-like protein Mst77F into sperm chromatin. J Biol Chem 2023; 299:105212. [PMID: 37660905 PMCID: PMC10520872 DOI: 10.1016/j.jbc.2023.105212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
DNA in sperm is packed with small, charged proteins termed SNBPs (sperm nuclear basic proteins), including mammalian and Drosophila protamines. During spermiogenesis, somatic-type chromatin is taken apart and replaced with sperm chromatin in a multistep process leading to an extraordinary condensation of the genome. During fertilization, the ova face a similarly challenging task of SNBP eviction and reassembly of nucleosome-based chromatin. Despite its importance for the animal life cycle, sperm chromatin metabolism, including the biochemical machinery mediating the mutual replacement of histones and SNBPs, remains poorly studied. In Drosophila, Mst77F is one of the first SNBPs loaded into the spermatid nuclei. It persists in mature spermatozoa and is essential for sperm compaction and male fertility. Here, by using in vitro biochemical assays, we identify chaperones that can mediate the eviction and loading of Mst77F on DNA, thus facilitating the interconversions of chromatin forms in the male gamete. Unlike NAP1 and TAP/p32 chaperones that disassemble Mst77F-DNA complexes, ARTEMIS and APOLLO, orthologs of mammalian importin-4 (IPO4), mediate the deposition of Mst77F on DNA or oligonucleosome templates, accompanied by the dissociation of histone-DNA complexes. In vivo, a mutation of testis-specific Apollo brings about a defect of Mst77F loading, abnormal sperm morphology, and male infertility. We identify IPO4 ortholog APOLLO as a critical component of sperm chromatin assembly apparatus in Drosophila. We discover that in addition to recognized roles in protein traffic, a nuclear transport receptor (IPO4) can function directly in chromatin remodeling as a dual, histone- and SNBP-specific, chaperone.
Collapse
Affiliation(s)
- Alexander V Emelyanov
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Daniel Barcenilla-Merino
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France
| | - Benjamin Loppin
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Université Claude Bernard Lyon 1, Lyon, France.
| | - Dmitry V Fyodorov
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA.
| |
Collapse
|
16
|
Hirschhäuser A, Molitor D, Salinas G, Großhans J, Rust K, Bogdan S. Single-cell transcriptomics identifies new blood cell populations in Drosophila released at the onset of metamorphosis. Development 2023; 150:dev201767. [PMID: 37681301 PMCID: PMC10560556 DOI: 10.1242/dev.201767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
Drosophila blood cells called hemocytes form an efficient barrier against infections and tissue damage. During metamorphosis, hemocytes undergo tremendous changes in their shape and behavior, preparing them for tissue clearance. Yet, the diversity and functional plasticity of pupal blood cells have not been explored. Here, we combine single-cell transcriptomics and high-resolution microscopy to dissect the heterogeneity and plasticity of pupal hemocytes. We identified undifferentiated and specified hemocytes with different molecular signatures associated with distinct functions such as antimicrobial, antifungal immune defense, cell adhesion or secretion. Strikingly, we identified a highly migratory and immune-responsive pupal cell population expressing typical markers of the posterior signaling center (PSC), which is known to be an important niche in the larval lymph gland. PSC-like cells become restricted to the abdominal segments and are morphologically very distinct from typical Hemolectin (Hml)-positive plasmatocytes. G-TRACE lineage experiments further suggest that PSC-like cells can transdifferentiate to lamellocytes triggered by parasitoid wasp infestation. In summary, we present the first molecular description of pupal Drosophila blood cells, providing insights into blood cell functional diversification and plasticity during pupal metamorphosis.
Collapse
Affiliation(s)
- Alexander Hirschhäuser
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Darius Molitor
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit, Department of Human Genetics, University Medical Center Göttingen, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Jörg Großhans
- Department of Biology, Philipps University Marburg, Karl-von-Frisch-Strasse 8, 35043 Marburg, Germany
| | - Katja Rust
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| | - Sven Bogdan
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps University Marburg, Emil-Mannkopff-Strasse 2, 35037 Marburg, Germany
| |
Collapse
|
17
|
Gershoni M, Braun T, Hauser R, Barda S, Lehavi O, Malcov M, Frumkin T, Kalma Y, Pietrokovski S, Arama E, Kleiman SE. A pathogenic variant in the uncharacterized RNF212B gene results in severe aneuploidy male infertility and repeated IVF failure. HGG ADVANCES 2023; 4:100189. [PMID: 37124137 PMCID: PMC10133878 DOI: 10.1016/j.xhgg.2023.100189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Quantitative and qualitative spermatogenic impairments are major causes of men's infertility. Although in vitro fertilization (IVF) is effective, some couples persistently fail to conceive. To identify causal variants in patients with severe male infertility factor and repeated IVF failures, we sequenced the exome of two consanguineous family members who underwent several failed IVF cycles and were diagnosed with low sperm count and motility. We identified a rare homozygous nonsense mutation in a previously uncharacterized gene, RNF212B, as the causative variant. Recurrence was identified in another unrelated, infertile patient who also faced repeated failed IVF treatments. scRNA-seq demonstrated meiosis-specific expression of RNF212B. Sequence analysis located a protein domain known to be associated with aneuploidy, which can explain multiple IVF failures. Accordingly, FISH analysis revealed a high aneuploidy rate in the patients' sperm cells and their IVF embryos. Finally, inactivation of the Drosophila orthologs significantly reduced male fertility. Given that members of the evolutionary conserved RNF212 gene family are involved in meiotic recombination and crossover maturation, our findings indicate a critical role of RNF212B in meiosis, genome stability, and in human fertility. Since recombination is completely absent in Drosophila males, our findings may indicate an additional unrelated role for the RNF212-like paralogs in spermatogenesis.
Collapse
Affiliation(s)
- Moran Gershoni
- ARO-The Volcani Center Institute of Animal Science, Bet Dagan, Israel
- Corresponding author
| | - Tslil Braun
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Hauser
- Racine IVF Unit and Male Fertility Clinic and Sperm Bank, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shimi Barda
- Racine IVF Unit and Male Fertility Clinic and Sperm Bank, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Lehavi
- Racine IVF Unit and Male Fertility Clinic and Sperm Bank, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mira Malcov
- Racine IVF Unit and Male Fertility Clinic and Sperm Bank, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tsvia Frumkin
- Racine IVF Unit and Male Fertility Clinic and Sperm Bank, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Kalma
- Racine IVF Unit and Male Fertility Clinic and Sperm Bank, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shmuel Pietrokovski
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Corresponding author
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Corresponding author
| | - Sandra E. Kleiman
- Racine IVF Unit and Male Fertility Clinic and Sperm Bank, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Corresponding author
| |
Collapse
|
18
|
Orsi GA, Tortora MMC, Horard B, Baas D, Kleman JP, Bucevičius J, Lukinavičius G, Jost D, Loppin B. Biophysical ordering transitions underlie genome 3D re-organization during cricket spermiogenesis. Nat Commun 2023; 14:4187. [PMID: 37443316 PMCID: PMC10345107 DOI: 10.1038/s41467-023-39908-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Spermiogenesis is a radical process of differentiation whereby sperm cells acquire a compact and specialized morphology to cope with the constraints of sexual reproduction while preserving their main cargo, an intact copy of the paternal genome. In animals, this often involves the replacement of most histones by sperm-specific nuclear basic proteins (SNBPs). Yet, how the SNBP-structured genome achieves compaction and accommodates shaping remain largely unknown. Here, we exploit confocal, electron and super-resolution microscopy, coupled with polymer modeling to identify the higher-order architecture of sperm chromatin in the needle-shaped nucleus of the emerging model cricket Gryllus bimaculatus. Accompanying spermatid differentiation, the SNBP-based genome is strikingly reorganized as ~25nm-thick fibers orderly coiled along the elongated nucleus axis. This chromatin spool is further found to achieve large-scale helical twisting in the final stages of spermiogenesis, favoring its ultracompaction. We reveal that these dramatic transitions may be recapitulated by a surprisingly simple biophysical principle based on a nucleated rigidification of chromatin linked to the histone-to-SNBP transition within a confined nuclear space. Our work highlights a unique, liquid crystal-like mode of higher-order genome organization in ultracompact cricket sperm, and establishes a multidisciplinary methodological framework to explore the diversity of non-canonical modes of DNA organization.
Collapse
Affiliation(s)
- Guillermo A Orsi
- Institute for Advanced Biosciences, University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, 38000, Grenoble, France.
| | - Maxime M C Tortora
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Inserm U1293, Université Claude Bernard Lyon 1, Lyon, France
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Béatrice Horard
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Inserm U1293, Université Claude Bernard Lyon 1, Lyon, France
| | - Dominique Baas
- Laboratoire MeLiS, CNRS UMR 52684, Inserm U 1314, Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Lyon, France
| | - Jean-Philippe Kleman
- Institut de Biologie Structurale, UMR5075, University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Jonas Bucevičius
- Chromatin Labeling and Imaging Group, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gražvydas Lukinavičius
- Chromatin Labeling and Imaging Group, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Daniel Jost
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Inserm U1293, Université Claude Bernard Lyon 1, Lyon, France.
| | - Benjamin Loppin
- Laboratoire de Biologie et Modélisation de la Cellule, École Normale Supérieure de Lyon, CNRS UMR5239, Inserm U1293, Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
19
|
Li P, Messina G, Lehner CF. Nuclear elongation during spermiogenesis depends on physical linkage of nuclear pore complexes to bundled microtubules by Drosophila Mst27D. PLoS Genet 2023; 19:e1010837. [PMID: 37428798 PMCID: PMC10359004 DOI: 10.1371/journal.pgen.1010837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023] Open
Abstract
Spermatozoa in animal species are usually highly elongated cells with a long motile tail attached to a head that contains the haploid genome in a compact and often elongated nucleus. In Drosophila melanogaster, the nucleus is compacted two hundred-fold in volume during spermiogenesis and re-modeled into a needle that is thirty-fold longer than its diameter. Nuclear elongation is preceded by a striking relocalization of nuclear pore complexes (NPCs). While NPCs are initially located throughout the nuclear envelope (NE) around the spherical nucleus of early round spermatids, they are later confined to one hemisphere. In the cytoplasm adjacent to this NPC-containing NE, the so-called dense complex with a strong bundle of microtubules is assembled. While this conspicuous proximity argued for functional significance of NPC-NE and microtubule bundle, experimental confirmation of their contributions to nuclear elongation has not yet been reported. Our functional characterization of the spermatid specific Mst27D protein now resolves this deficit. We demonstrate that Mst27D establishes physical linkage between NPC-NE and dense complex. The C-terminal region of Mst27D binds to the nuclear pore protein Nup358. The N-terminal CH domain of Mst27D, which is similar to that of EB1 family proteins, binds to microtubules. At high expression levels, Mst27D promotes bundling of microtubules in cultured cells. Microscopic analyses indicated co-localization of Mst27D with Nup358 and with the microtubule bundles of the dense complex. Time-lapse imaging revealed that nuclear elongation is accompanied by a progressive bundling of microtubules into a single elongated bundle. In Mst27D null mutants, this bundling process does not occur and nuclear elongation is abnormal. Thus, we propose that Mst27D permits normal nuclear elongation by promoting the attachment of the NPC-NE to the microtubules of the dense complex, as well as the progressive bundling of these microtubules.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Molecular Life Science (DMLS), University of Zurich, Zurich, Switzerland
| | - Giovanni Messina
- Department of Molecular Life Science (DMLS), University of Zurich, Zurich, Switzerland
| | - Christian F Lehner
- Department of Molecular Life Science (DMLS), University of Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Park JI, Bell GW, Yamashita YM. Derepression of Y-linked multicopy protamine-like genes interferes with sperm nuclear compaction in D. melanogaster. Proc Natl Acad Sci U S A 2023; 120:e2220576120. [PMID: 37036962 PMCID: PMC10120018 DOI: 10.1073/pnas.2220576120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/17/2023] [Indexed: 04/12/2023] Open
Abstract
Across species, sperm maturation involves the dramatic reconfiguration of chromatin into highly compact nuclei that enhance hydrodynamic ability and ensure paternal genomic integrity. This process is mediated by the replacement of histones by sperm nuclear basic proteins, also referred to as protamines. In humans, a carefully balanced dosage between two known protamine genes is required for optimal fertility. However, it remains unknown how their proper balance is regulated and how defects in balance may lead to compromised fertility. Here, we show that a nucleolar protein, modulo, a homolog of nucleolin, mediates the histone-to-protamine transition during Drosophila spermatogenesis. We find that modulo mutants display nuclear compaction defects during late spermatogenesis due to decreased expression of autosomal protamine genes (including Mst77F) and derepression of Y-linked multicopy Mst77F homologs (Mst77Y), leading to the mutant's known sterility. Overexpression of Mst77Y in a wild-type background is sufficient to cause nuclear compaction defects, similar to modulo mutant, indicating that Mst77Y is a dominant-negative variant interfering with the process of histone-to-protamine transition. Interestingly, ectopic overexpression of Mst77Y caused decompaction of X-bearing spermatids nuclei more frequently than Y-bearing spermatid nuclei, although this did not greatly affect the sex ratio of offspring. We further show that modulo regulates these protamine genes at the step of transcript polyadenylation. We conclude that the regulation of protamines mediated by modulo, ensuring the expression of functional ones while repressing dominant-negative ones, is critical for male fertility.
Collapse
Affiliation(s)
- Jun I. Park
- Life Sciences Institute, University of Michigan, Ann Arbor, MI48109
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI48109
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI48109
| | - George W. Bell
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
| | - Yukiko M. Yamashita
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, School of Science, Massachusetts Institute of Technology, Cambridge, MA02142
- HHMI, Cambridge, MA02142
| |
Collapse
|
21
|
Shao L, Fingerhut JM, Falk BL, Han H, Maldonado G, Qiao Y, Lee V, Hall E, Chen L, Polevoy G, Hernández G, Lasko P, Brill JA. Eukaryotic translation initiation factor eIF4E-5 is required for spermiogenesis in Drosophila melanogaster. Development 2023; 150:286752. [PMID: 36695474 DOI: 10.1242/dev.200477] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Drosophila sperm development is characterized by extensive post-transcriptional regulation whereby thousands of transcripts are preserved for translation during later stages. A key step in translation initiation is the binding of eukaryotic initiation factor 4E (eIF4E) to the 5' mRNA cap. In addition to canonical eIF4E-1, Drosophila has multiple eIF4E paralogs, including four (eIF4E-3, -4, -5, and -7) that are highly expressed in the testis. Among these, only eIF4E-3 has been characterized genetically. Here, using CRISPR/Cas9 mutagenesis, we determined that eIF4E-5 is essential for male fertility. eIF4E-5 protein localizes to the distal ends of elongated spermatid cysts, and eIF4E-5 mutants exhibit defects during post-meiotic stages, including a mild defect in spermatid cyst polarization. eIF4E-5 mutants also have a fully penetrant defect in individualization, resulting in failure to produce mature sperm. Indeed, our data indicate that eIF4E-5 regulates non-apoptotic caspase activity during individualization by promoting local accumulation of the E3 ubiquitin ligase inhibitor Soti. Our results further extend the diversity of non-canonical eIF4Es that carry out distinct spatiotemporal roles during spermatogenesis.
Collapse
Affiliation(s)
- Lisa Shao
- Cell Biology Program, The Hospital for Sick Children, PGCRL Building, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Jaclyn M Fingerhut
- Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, 455 Main Street, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, 455 Main Street, Cambridge, MA 02142, USA
| | - Brook L Falk
- Cell Biology Program, The Hospital for Sick Children, PGCRL Building, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Hong Han
- Department of Biology, McGill University, 3649 Promenade Sir William Osler, Montréal, Quebec, H3G 0B1, Canada
| | - Giovanna Maldonado
- Laboratory of Translation and Cancer, Unit of Biomedical Research on Cancer, Instituto Nacional de Cancerología (INCan), Av San Fernando 22, Mexico City 14080, Mexico
| | - Yuemeng Qiao
- Cell Biology Program, The Hospital for Sick Children, PGCRL Building, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- Human Biology Program, University of Toronto, 300 Huron Street, Toronto, Ontario, M5S 3J6, Canada
| | - Vincent Lee
- Cell Biology Program, The Hospital for Sick Children, PGCRL Building, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Elizabeth Hall
- Cell Biology Program, The Hospital for Sick Children, PGCRL Building, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Liang Chen
- Department of Biology, McGill University, 3649 Promenade Sir William Osler, Montréal, Quebec, H3G 0B1, Canada
| | - Gordon Polevoy
- Cell Biology Program, The Hospital for Sick Children, PGCRL Building, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
| | - Greco Hernández
- Laboratory of Translation and Cancer, Unit of Biomedical Research on Cancer, Instituto Nacional de Cancerología (INCan), Av San Fernando 22, Mexico City 14080, Mexico
| | - Paul Lasko
- Department of Biology, McGill University, 3649 Promenade Sir William Osler, Montréal, Quebec, H3G 0B1, Canada
| | - Julie A Brill
- Cell Biology Program, The Hospital for Sick Children, PGCRL Building, 686 Bay Street, Toronto, Ontario, M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
22
|
Chang CH, Mejia Natividad I, Malik HS. Expansion and loss of sperm nuclear basic protein genes in Drosophila correspond with genetic conflicts between sex chromosomes. eLife 2023; 12:85249. [PMID: 36763410 PMCID: PMC9917458 DOI: 10.7554/elife.85249] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/04/2023] [Indexed: 02/11/2023] Open
Abstract
Many animal species employ sperm nuclear basic proteins (SNBPs) or protamines to package sperm genomes tightly. SNBPs vary across animal lineages and evolve rapidly in mammals. We used a phylogenomic approach to investigate SNBP diversification in Drosophila species. We found that most SNBP genes in Drosophila melanogaster evolve under positive selection except for genes essential for male fertility. Unexpectedly, evolutionarily young SNBP genes are more likely to be critical for fertility than ancient, conserved SNBP genes. For example, CG30056 is dispensable for male fertility despite being one of three SNBP genes universally retained in Drosophila species. We found 19 independent SNBP gene amplification events that occurred preferentially on sex chromosomes. Conversely, the montium group of Drosophila species lost otherwise-conserved SNBP genes, coincident with an X-Y chromosomal fusion. Furthermore, SNBP genes that became linked to sex chromosomes via chromosomal fusions were more likely to degenerate or relocate back to autosomes. We hypothesize that autosomal SNBP genes suppress meiotic drive, whereas sex-chromosomal SNBP expansions lead to meiotic drive. X-Y fusions in the montium group render autosomal SNBPs dispensable by making X-versus-Y meiotic drive obsolete or costly. Thus, genetic conflicts between sex chromosomes may drive SNBP rapid evolution during spermatogenesis in Drosophila species.
Collapse
Affiliation(s)
- Ching-Ho Chang
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, United States
| | - Isabel Mejia Natividad
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, United States.,Howard Hughes Medical Institute, Fred Hutchinson Cancer Center, Seattle, United States
| | - Harmit S Malik
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, United States.,Howard Hughes Medical Institute, Fred Hutchinson Cancer Center, Seattle, United States
| |
Collapse
|
23
|
Scheuren M, Möhner J, Zischler H. R-loop landscape in mature human sperm: Regulatory and evolutionary implications. Front Genet 2023; 14:1069871. [PMID: 37139234 PMCID: PMC10149866 DOI: 10.3389/fgene.2023.1069871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 04/03/2023] [Indexed: 05/05/2023] Open
Abstract
R-loops are three-stranded nucleic acid structures consisting of an RNA:DNA hybrid and a displaced DNA strand. While R-loops pose a potential threat to genome integrity, they constitute 5% of the human genome. The role of R-loops in transcriptional regulation, DNA replication, and chromatin signature is becoming increasingly clear. R-loops are associated with various histone modifications, suggesting that they may modulate chromatin accessibility. To potentially harness transcription-coupled repair mechanisms in the germline, nearly the entire genome is expressed during the early stages of male gametogenesis in mammals, providing ample opportunity for the formation of a transcriptome-dependent R-loop landscape in male germ cells. In this study, our data demonstrated the presence of R-loops in fully mature human and bonobo sperm heads and their partial correspondence to transcribed regions and chromatin structure, which is massively reorganized from mainly histone to mainly protamine-packed chromatin in mature sperm. The sperm R-loop landscape resembles characteristic patterns of somatic cells. Surprisingly, we detected R-loops in both residual histone and protamine-packed chromatin and localize them to still-active retroposons, ALUs and SINE-VNTR-ALUs (SVAs), the latter has recently arisen in hominoid primates. We detected both evolutionarily conserved and species-specific localizations. Comparing our DNA-RNA immunoprecipitation (DRIP) data with published DNA methylation and histone chromatin immunoprecipitation (ChIP) data, we hypothesize that R-loops epigenetically reduce methylation of SVAs. Strikingly, we observe a strong influence of R-loops on the transcriptomes of zygotes from early developmental stages before zygotic genome activation. Overall, these findings suggest that chromatin accessibility influenced by R-loops may represent a system of inherited gene regulation.
Collapse
|
24
|
Mao B, Zhang W, Zheng Y, Li D, Chen MY, Wang YF. Comparative phosphoproteomics reveal new candidates in the regulation of spermatogenesis of Drosophila melanogaster. INSECT SCIENCE 2022; 29:1703-1720. [PMID: 35271765 DOI: 10.1111/1744-7917.13031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 06/14/2023]
Abstract
The most common phenotype induced by the endosymbiont Wolbachia in insects is cytoplasmic incompatibility, where none or fewer progenies can be produced when Wolbachia-infected males mate with uninfected females. This suggests that some modifications are induced in host sperms during spermatogenesis by Wolbachia. To identify the proteins whose phosphorylation states play essential roles in male reproduction in Drosophila melanogaster, we applied isobaric tags for relative and absolute quantitation (iTRAQ)-based proteomic strategy combined with titanium dioxide (TiO2 ) enrichment to compare the phosphoproteome of Wolbachia-infected with that of uninfected male reproductive systems in D. melanogaster. We identified 182 phosphopeptides, defining 140 phosphoproteins, that have at least a 1.2 fold change in abundance with a P-value of <0.05. Most of the differentially abundant phosphoproteins (DAPPs) were associated with microtubule cytoskeleton organization and spermatid differentiation. The DAPPs included proteins already known to be associated with spermatogenesis, as well as many not previously studied during this process. Six genes coding for DAPPs were knocked down, respectively, in Wolbachia-free fly testes. Among them, Slmap knockdown caused the most severe damage in spermatogenesis, with no mature sperm observed in seminal vesicles. Immunofluorescence staining showed that the formation of individualization complex composed of actin cones was completely disrupted. These results suggest that Wolbachia may induce wide changes in the abundance of phosphorylated proteins which are closely related to male reproduction. By identifying phospho-modulated proteins we also provide a significant candidate set for future studies on their roles in spermatogenesis.
Collapse
Affiliation(s)
- Bin Mao
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Wei Zhang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Ya Zheng
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Dong Li
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Meng-Yan Chen
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| | - Yu-Feng Wang
- School of Life Sciences, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, China
| |
Collapse
|
25
|
A Tremendous Reorganization Journey for the 3D Chromatin Structure from Gametes to Embryos. Genes (Basel) 2022; 13:genes13101864. [PMID: 36292750 PMCID: PMC9602195 DOI: 10.3390/genes13101864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/02/2022] [Accepted: 10/12/2022] [Indexed: 11/04/2022] Open
Abstract
The 3D chromatin structure within the nucleus is important for gene expression regulation and correct developmental programs. Recently, the rapid development of low-input chromatin conformation capture technologies has made it possible to study 3D chromatin structures in gametes, zygotes and early embryos in a variety of species, including flies, vertebrates and mammals. There are distinct 3D chromatin structures within the male and female gametes. Following the fertilization of male and female gametes, fertilized eggs undergo drastic epigenetic reprogramming at multi levels, including the 3D chromatin structure, to convert the terminally differentiated gamete state into the totipotent state, which can give rise to an individual. However, to what extent the 3D chromatin structure reorganization is evolutionarily conserved and what the underlying mechanisms are for the tremendous reorganization in early embryos remain elusive. Here, we review the latest findings on the 3D chromatin structure reorganization during embryogenesis, and discuss the convergent and divergent reprogramming patterns and key molecular mechanisms for the 3D chromatin structure reorganization from gametes to embryos in different species. These findings shed light on how the 3D chromatin structure reorganization contribute to embryo development in different species. The findings also indicate the role of the 3D chromatin structure on the acquisition of totipotent developmental potential.
Collapse
|
26
|
Kaur R, Leigh BA, Ritchie IT, Bordenstein SR. The Cif proteins from Wolbachia prophage WO modify sperm genome integrity to establish cytoplasmic incompatibility. PLoS Biol 2022; 20:e3001584. [PMID: 35609042 PMCID: PMC9128985 DOI: 10.1371/journal.pbio.3001584] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/25/2022] [Indexed: 01/27/2023] Open
Abstract
Inherited microorganisms can selfishly manipulate host reproduction to drive through populations. In Drosophila melanogaster, germline expression of the native Wolbachia prophage WO proteins CifA and CifB cause cytoplasmic incompatibility (CI) in which embryos from infected males and uninfected females suffer catastrophic mitotic defects and lethality; however, in infected females, CifA expression rescues the embryonic lethality and thus imparts a fitness advantage to the maternally transmitted Wolbachia. Despite widespread relevance to sex determination, evolution, and vector control, the mechanisms underlying when and how CI impairs male reproduction remain unknown and a topic of debate. Here, we use cytochemical, microscopic, and transgenic assays in D. melanogaster to demonstrate that CifA and CifB proteins of wMel localize to nuclear DNA throughout the process of spermatogenesis. Cif proteins cause abnormal histone retention in elongating spermatids and protamine deficiency in mature sperms that travel to the female reproductive tract with Cif proteins. Notably, protamine gene knockouts enhance wild-type CI. In ovaries, CifA localizes to germ cell nuclei and cytoplasm of early-stage egg chambers; however, Cifs are absent in late-stage oocytes and subsequently in fertilized embryos. Finally, CI and rescue are contingent upon a newly annotated CifA bipartite nuclear localization sequence. Together, our results strongly support the Host modification model of CI in which Cifs initially modify the paternal and maternal gametes to bestow CI-defining embryonic lethality and rescue.
Collapse
Affiliation(s)
- Rupinder Kaur
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Brittany A. Leigh
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Isabella T. Ritchie
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Seth R. Bordenstein
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, United States of America
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
27
|
Vedanayagam J, Lin CJ, Lai EC. Rapid evolutionary dynamics of an expanding family of meiotic drive factors and their hpRNA suppressors. Nat Ecol Evol 2021; 5:1613-1623. [PMID: 34862477 PMCID: PMC8665063 DOI: 10.1038/s41559-021-01592-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/19/2021] [Indexed: 11/25/2022]
Abstract
Meiotic drivers are a class of selfish genetic elements whose existence is frequently hidden due to concomitant suppressor systems. Accordingly, we know little of their evolutionary breadth and molecular mechanisms. Here, we trace the evolution of the Dox meiotic drive system in Drosophila simulans, which affects male-female balance (sex ratio). Dox emerged via stepwise mobilization and acquisition of multiple D. melanogaster gene segments including from protamine, which mediates compaction of sperm chromatin. Moreover, we reveal novel Dox homologs and massive amplification of Dox superfamily genes on X chromosomes of its closest sisters D. mauritiana and D. sechellia. Emergence of Dox loci is tightly associated with 359-class satellite repeats that flank de novo genomic copies. In concert, we find coordinated diversification of autosomal hairpin RNA-class siRNA loci that target subsets of Dox superfamily genes. Overall, we reveal fierce genetic arms races between meiotic drive factors and siRNA suppressors associated with recent speciation.
Collapse
Affiliation(s)
- Jeffrey Vedanayagam
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.
| | - Ching-Jung Lin
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
- Weill Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Eric C Lai
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.
- Weill Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
28
|
Rivard EL, Ludwig AG, Patel PH, Grandchamp A, Arnold SE, Berger A, Scott EM, Kelly BJ, Mascha GC, Bornberg-Bauer E, Findlay GD. A putative de novo evolved gene required for spermatid chromatin condensation in Drosophila melanogaster. PLoS Genet 2021; 17:e1009787. [PMID: 34478447 PMCID: PMC8445463 DOI: 10.1371/journal.pgen.1009787] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/16/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023] Open
Abstract
Comparative genomics has enabled the identification of genes that potentially evolved de novo from non-coding sequences. Many such genes are expressed in male reproductive tissues, but their functions remain poorly understood. To address this, we conducted a functional genetic screen of over 40 putative de novo genes with testis-enriched expression in Drosophila melanogaster and identified one gene, atlas, required for male fertility. Detailed genetic and cytological analyses showed that atlas is required for proper chromatin condensation during the final stages of spermatogenesis. Atlas protein is expressed in spermatid nuclei and facilitates the transition from histone- to protamine-based chromatin packaging. Complementary evolutionary analyses revealed the complex evolutionary history of atlas. The protein-coding portion of the gene likely arose at the base of the Drosophila genus on the X chromosome but was unlikely to be essential, as it was then lost in several independent lineages. Within the last ~15 million years, however, the gene moved to an autosome, where it fused with a conserved non-coding RNA and evolved a non-redundant role in male fertility. Altogether, this study provides insight into the integration of novel genes into biological processes, the links between genomic innovation and functional evolution, and the genetic control of a fundamental developmental process, gametogenesis.
Collapse
Affiliation(s)
- Emily L. Rivard
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| | - Andrew G. Ludwig
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| | - Prajal H. Patel
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| | | | - Sarah E. Arnold
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| | | | - Emilie M. Scott
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| | - Brendan J. Kelly
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| | - Grace C. Mascha
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| | - Erich Bornberg-Bauer
- University of Münster, Münster, Germany
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Geoffrey D. Findlay
- College of the Holy Cross, Worcester, Massachusetts, United States of America
| |
Collapse
|
29
|
Gilmutdinov R, Kozlov EN, Yakovlev KV, Olenina LV, Kotov AA, Barr J, Zhukova M, Schedl P, Shidlovskii YV. The 3'UTR of the Drosophila CPEB translation factor gene orb2 plays a crucial role in spermatogenesis. Development 2021; 148:272122. [PMID: 34473243 DOI: 10.1242/dev.198788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/22/2021] [Indexed: 01/27/2023]
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.
Collapse
Affiliation(s)
- Rudolf Gilmutdinov
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Eugene N Kozlov
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Konstantin V Yakovlev
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia.,Laboratory of Cytotechnology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Ludmila V Olenina
- Department of Molecular Genetics of Cell, Institute of Molecular Genetics, National Research Centre Kurchatov Institute, Moscow 123182, Russia
| | - Alexei A Kotov
- Department of Molecular Genetics of Cell, Institute of Molecular Genetics, National Research Centre Kurchatov Institute, Moscow 123182, Russia
| | - Justinn Barr
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA
| | - Mariya Zhukova
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544-1014, USA
| | - Yulii V Shidlovskii
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia.,Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University, Moscow 119048, Russia
| |
Collapse
|
30
|
Herbette M, Wei X, Chang CH, Larracuente AM, Loppin B, Dubruille R. Distinct spermiogenic phenotypes underlie sperm elimination in the Segregation Distorter meiotic drive system. PLoS Genet 2021; 17:e1009662. [PMID: 34228705 PMCID: PMC8284685 DOI: 10.1371/journal.pgen.1009662] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/16/2021] [Accepted: 06/10/2021] [Indexed: 12/28/2022] Open
Abstract
Segregation Distorter (SD) is a male meiotic drive system in Drosophila melanogaster. Males heterozygous for a selfish SD chromosome rarely transmit the homologous SD+ chromosome. It is well established that distortion results from an interaction between Sd, the primary distorting locus on the SD chromosome and its target, a satellite DNA called Rsp, on the SD+ chromosome. However, the molecular and cellular mechanisms leading to post-meiotic SD+ sperm elimination remain unclear. Here we show that SD/SD+ males of different genotypes but with similarly strong degrees of distortion have distinct spermiogenic phenotypes. In some genotypes, SD+ spermatids fail to fully incorporate protamines after the removal of histones, and degenerate during the individualization stage of spermiogenesis. In contrast, in other SD/SD+ genotypes, protamine incorporation appears less disturbed, yet spermatid nuclei are abnormally compacted, and mature sperm nuclei are eventually released in the seminal vesicle. Our analyses of different SD+ chromosomes suggest that the severity of the spermiogenic defects associates with the copy number of the Rsp satellite. We propose that when Rsp copy number is very high (> 2000), spermatid nuclear compaction defects reach a threshold that triggers a checkpoint controlling sperm chromatin quality to eliminate abnormal spermatids during individualization.
Collapse
Affiliation(s)
- Marion Herbette
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS UMR 5239, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Xiaolu Wei
- University of Rochester Medical Center, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Ching-Ho Chang
- University of Rochester Department of Biology, Rochester, New York, United States of America
| | - Amanda M. Larracuente
- University of Rochester Department of Biology, Rochester, New York, United States of America
| | - Benjamin Loppin
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS UMR 5239, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| | - Raphaëlle Dubruille
- Laboratoire de Biologie et Modélisation de la Cellule, CNRS UMR 5239, École Normale Supérieure de Lyon, University of Lyon, Lyon, France
| |
Collapse
|
31
|
Presgraves DC, Meiklejohn CD. Hybrid Sterility, Genetic Conflict and Complex Speciation: Lessons From the Drosophila simulans Clade Species. Front Genet 2021; 12:669045. [PMID: 34249091 PMCID: PMC8261240 DOI: 10.3389/fgene.2021.669045] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/20/2021] [Indexed: 12/31/2022] Open
Abstract
The three fruitfly species of the Drosophila simulans clade- D. simulans, D. mauritiana, and D. sechellia- have served as important models in speciation genetics for over 40 years. These species are reproductively isolated by geography, ecology, sexual signals, postmating-prezygotic interactions, and postzygotic genetic incompatibilities. All pairwise crosses between these species conform to Haldane's rule, producing fertile F1 hybrid females and sterile F1 hybrid males. The close phylogenetic proximity of the D. simulans clade species to the model organism, D. melanogaster, has empowered genetic analyses of their species differences, including reproductive incompatibilities. But perhaps no phenotype has been subject to more continuous and intensive genetic scrutiny than hybrid male sterility. Here we review the history, progress, and current state of our understanding of hybrid male sterility among the D. simulans clade species. Our aim is to integrate the available information from experimental and population genetics analyses bearing on the causes and consequences of hybrid male sterility. We highlight numerous conclusions that have emerged as well as issues that remain unresolved. We focus on the special role of sex chromosomes, the fine-scale genetic architecture of hybrid male sterility, and the history of gene flow between species. The biggest surprises to emerge from this work are that (i) genetic conflicts may be an important general force in the evolution of hybrid incompatibility, (ii) hybrid male sterility is polygenic with contributions of complex epistasis, and (iii) speciation, even among these geographically allopatric taxa, has involved the interplay of gene flow, negative selection, and positive selection. These three conclusions are marked departures from the classical views of speciation that emerged from the modern evolutionary synthesis.
Collapse
Affiliation(s)
- Daven C. Presgraves
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Colin D. Meiklejohn
- School of Biological Sciences, University of Nebraska, Lincoln, NE, United States
| |
Collapse
|
32
|
Reduce, Retain, Recycle: Mechanisms for Promoting Histone Protein Degradation versus Stability and Retention. Mol Cell Biol 2021; 41:e0000721. [PMID: 33753462 DOI: 10.1128/mcb.00007-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The eukaryotic genome is packaged into chromatin. The nucleosome, the basic unit of chromatin, is composed of DNA coiled around a histone octamer. Histones are among the longest-lived protein species in mammalian cells due to their thermodynamic stability and their associations with DNA and histone chaperones. Histone metabolism plays an integral role in homeostasis. While histones are largely stable, the degradation of histone proteins is necessary under specific conditions. Here, we review the physiological and cellular contexts that promote histone degradation. We describe specific known mechanisms that drive histone proteolysis. Finally, we discuss the importance of histone degradation and regulation of histone supply for organismal and cellular fitness.
Collapse
|
33
|
Palacios V, Kimble GC, Tootle TL, Buszczak M. Importin-9 regulates chromosome segregation and packaging in Drosophila germ cells. J Cell Sci 2021; 134:237786. [PMID: 33632744 DOI: 10.1242/jcs.258391] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/10/2021] [Indexed: 12/29/2022] Open
Abstract
Germ cells undergo distinct nuclear processes as they differentiate into gametes. Although these events must be coordinated to ensure proper maturation, the stage-specific transport of proteins in and out of germ cell nuclei remains incompletely understood. Our efforts to genetically characterize Drosophila genes that exhibit enriched expression in germ cells led to the finding that loss of the highly conserved Importin β/karyopherin family member Importin-9 (Ipo9, herein referring to Ranbp9) results in female and male sterility. Immunofluorescence and fluorescent in situ hybridization revealed that Ipo9KO mutants display chromosome condensation and segregation defects during meiosis. In addition, Ipo9KO mutant males form abnormally structured sperm and fail to properly exchange histones for protamines. Ipo9 physically interacts with proteasome proteins, and Ipo9 mutant males exhibit disruption of the nuclear localization of several proteasome components. Thus, Ipo9 coordinates the nuclear import of functionally related factors necessary for the completion of gametogenesis. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Victor Palacios
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Garrett C Kimble
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Tina L Tootle
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Martinet B, Zambra E, Przybyla K, Lecocq T, Anselmo A, Nonclercq D, Rasmont P, Michez D, Hennebert E. Mating under climate change: Impact of simulated heatwaves on the reproduction of model pollinators. Funct Ecol 2020. [DOI: 10.1111/1365-2435.13738] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Baptiste Martinet
- Laboratory of Zoology Research Institute of Biosciences University of Mons Mons Belgium
- Evolutionary Biology & Ecology Université Libre de Bruxelles Bruxelles Belgium
| | - Ella Zambra
- Laboratory of Zoology Research Institute of Biosciences University of Mons Mons Belgium
| | - Kimberly Przybyla
- Laboratory of Zoology Research Institute of Biosciences University of Mons Mons Belgium
| | - Thomas Lecocq
- Laboratory of Zoology Research Institute of Biosciences University of Mons Mons Belgium
- INRAEURAFPAUniversity of Lorraine Nancy France
| | - Abigaël Anselmo
- Laboratory of Zoology Research Institute of Biosciences University of Mons Mons Belgium
| | - Denis Nonclercq
- Laboratory of Histology Research Institute of BiosciencesUniversity of Mons Mons Belgium
| | - Pierre Rasmont
- Laboratory of Zoology Research Institute of Biosciences University of Mons Mons Belgium
| | - Denis Michez
- Laboratory of Zoology Research Institute of Biosciences University of Mons Mons Belgium
| | - Elise Hennebert
- Laboratory of Cell Biology Research Institute of BiosciencesUniversity of Mons Mons Belgium
| |
Collapse
|
35
|
Dong C, Nakagawa R, Oyama K, Yamamoto Y, Zhang W, Dong A, Li Y, Yoshimura Y, Kamiya H, Nakayama JI, Ueda J, Min J. Structural basis for histone variant H3tK27me3 recognition by PHF1 and PHF19. eLife 2020; 9:58675. [PMID: 32869745 PMCID: PMC7492083 DOI: 10.7554/elife.58675] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/29/2020] [Indexed: 12/30/2022] Open
Abstract
The Polycomb repressive complex 2 (PRC2) is a multicomponent histone H3K27 methyltransferase complex, best known for silencing the Hox genes during embryonic development. The Polycomb-like proteins PHF1, MTF2, and PHF19 are critical components of PRC2 by stimulating its catalytic activity in embryonic stem cells. The Tudor domains of PHF1/19 have been previously shown to be readers of H3K36me3 in vitro. However, some other studies suggest that PHF1 and PHF19 co-localize with the H3K27me3 mark but not H3K36me3 in cells. Here, we provide further evidence that PHF1 co-localizes with H3t in testis and its Tudor domain preferentially binds to H3tK27me3 over canonical H3K27me3 in vitro. Our complex structures of the Tudor domains of PHF1 and PHF19 with H3tK27me3 shed light on the molecular basis for preferential recognition of H3tK27me3 by PHF1 and PHF19 over canonical H3K27me3, implicating that H3tK27me3 might be a physiological ligand of PHF1/19.
Collapse
Affiliation(s)
- Cheng Dong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Reiko Nakagawa
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Kyohei Oyama
- Department of Cardiac Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Yusuke Yamamoto
- Department of Cardiac Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Weilian Zhang
- Structural Genomics Consortium, University of Toronto, Toronto, Canada
| | - Aiping Dong
- Structural Genomics Consortium, University of Toronto, Toronto, Canada
| | - Yanjun Li
- Structural Genomics Consortium, University of Toronto, Toronto, Canada
| | - Yuriko Yoshimura
- Division of Chromatin Regulation, National Institute for Basic Biology, Okazaki, Japan
| | - Hiroyuki Kamiya
- Department of Cardiac Surgery, Asahikawa Medical University, Asahikawa, Japan
| | - Jun-Ichi Nakayama
- Division of Chromatin Regulation, National Institute for Basic Biology, Okazaki, Japan.,Department of Basic Biology, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Jun Ueda
- Centre for Advanced Research and Education, Asahikawa Medical University, Asahikawa, Japan
| | - Jinrong Min
- Structural Genomics Consortium, University of Toronto, Toronto, Canada.,Department of Physiology, University of Toronto, Toronto, Canada
| |
Collapse
|
36
|
Torres-Flores U, Hernández-Hernández A. The Interplay Between Replacement and Retention of Histones in the Sperm Genome. Front Genet 2020; 11:780. [PMID: 32765595 PMCID: PMC7378789 DOI: 10.3389/fgene.2020.00780] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/30/2020] [Indexed: 12/21/2022] Open
Abstract
The genome of eukaryotes is highly organized within the cell nucleus, this organization per se elicits gene regulation and favors other mechanisms like cell memory throughout histones and their post-translational modifications. In highly specialized cells, like sperm, the genome is mostly organized by protamines, yet a significant portion of it remains organized by histones. This protamine-histone-DNA organization, known as sperm epigenome, is established during spermiogenesis. Specific histones and their post-translational modifications are retained at specific genomic sites and during embryo development these sites recapitulate their histone profile that harbored in the sperm nucleus. It is known that histones are the conduit of epigenetic memory from cell to cell, hence histones in the sperm epigenome may have a role in transmitting epigenetic memory from the sperm to the embryo. However, the exact function and mechanism of histone retention remains elusive. During spermatogenesis, most of the histones that organize the genome are replaced by protamines and their retention at specific regions may be deeply intertwined with the eviction and replacement mechanism. In this review we will cover some relevant aspects of histone replacement that in turn may help us to contextualize histone retention. In the end, we focus on the architectonical protein CTCF that is, so far, the only factor that has been directly linked to the histone retention process.
Collapse
Affiliation(s)
- Ulises Torres-Flores
- Biología de Células Individuales (BIOCELIN), Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Abrahan Hernández-Hernández
- Biología de Células Individuales (BIOCELIN), Laboratorio de Investigación en Patología Experimental, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| |
Collapse
|
37
|
Dong C, Liu Y, Lyu TJ, Beldar S, Lamb KN, Tempel W, Li Y, Li Z, James LI, Qin S, Wang Y, Min J. Structural Basis for the Binding Selectivity of Human CDY Chromodomains. Cell Chem Biol 2020; 27:827-838.e7. [DOI: 10.1016/j.chembiol.2020.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/21/2020] [Accepted: 05/11/2020] [Indexed: 01/22/2023]
|
38
|
A Protamine Knockdown Mimics the Function of Sd in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2020; 10:2111-2115. [PMID: 32321837 PMCID: PMC7263674 DOI: 10.1534/g3.120.401307] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Segregation Distorter (SD) is an autosomal meiotic drive system found worldwide in natural populations of Drosophila melanogaster. This gene complex induces the preferential and nearly exclusive transmission of the SD chromosome in SD/SD+ males. This selfish propagation occurs through the interplay of the Sd locus, its enhancers and the Rsps locus during spermatid development. The key distorter locus, Sd, encodes a truncated but enzymatically active RanGAP (RanGTPase-activating protein), a key nuclear transport factor in the Ran signaling pathway. When encoded by Sd, RanGAP is mislocalized to the nucleus interior, which then traps Ran inside the nucleus and disrupts nuclear import. As a result of this aberrant nuclear transport, a process known as the histone-to-protamine transition that is required for proper spermatid condensation fails to occur in SD/SD+ males. In this process, sperm-specific protamine proteins enter the spermatid nucleus and replace the formerly chromatin-complexed histones. Previously, we have shown that mutations affecting nuclear import and export can enhance distortion in an SD background, thus verifying that a defect in nuclear transport is responsible for the unequal transmission of chromosomes. Herein, we show that specifically reducing protamines induces distortion in an SD background, verifying that protamines are transported via the RanGAP/GEF pathway and indicating that E(SD) plays a significant and unique role in the process of distortion.
Collapse
|
39
|
Taşdemir U, Yeni D, İnanç ME, Avdatek F, Çil B, Türkmen R, Güngör Ş, Tuncer PB. Red pine (Pinus brutia Ten) bark tree extract preserves sperm quality by reducing oxidative stress and preventing chromatin damage. Andrologia 2020; 52:e13603. [PMID: 32294298 DOI: 10.1111/and.13603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
This study aimed to investigate the effectiveness of using red pine bark tree extract (P; Pinus brutia Ten) as a TRIS extender in an attempt to prevent oxidative stress in bull spermatozoa after freezing. Semen specimens were obtained from Simmental bulls via an artificial vagina and pooled. They were separated into five specimens and diluted with Tris extender consisting of P (200, 100, 50 and 25 µg/ml) and P free (control; C) up to a final concentration of 16 × 106 per straw. All specimens were equilibrated for a period of 4 hr at a temperature of 4°C, following which they were filled in 0.25-ml French straws and frozen. Addition of P resulted in favourable tail length in comparison with C (p < .05). The lowest malondialdehyde levels and the highest glutathione levels were detected in all P groups (p < .05). Supplementation with P did not show advanced results in terms of total, progressive sperm motility and total abnormality in comparison with C (p > .05). In conclusion, it has been shown that although P added to a Tris extender does not have a positive effect on sperm motility, it prevents chromatin damage by reducing oxidative stress, in addition to reducing head abnormalities when used at the amount of 50 μg/ml.
Collapse
Affiliation(s)
- Umut Taşdemir
- Technical Sciences Vocational School, Aksaray University, Aksaray, Turkey
| | - Deniz Yeni
- Faculty of Veterinary Medicine, Department of Reproduction and Artificial Insemination, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Muhammed Enes İnanç
- Faculty of Veterinary Medicine, Department of Reproduction and Artificial Insemination, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Fatih Avdatek
- Faculty of Veterinary Medicine, Department of Reproduction and Artificial Insemination, Afyon Kocatepe University, Afyonkarahisar, Turkey
| | - Beste Çil
- Faculty of Veterinary Medicine, Department of Reproduction and Artificial Insemination, Ankara University, Ankara, Turkey
| | - Ruhi Türkmen
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Afyon Kocatepe University, Afyon, Turkey
| | - Şükrü Güngör
- Faculty of Veterinary Medicine, Department of Reproduction and Artificial Insemination, Mehmet Akif Ersoy University, Burdur, Turkey
| | | |
Collapse
|
40
|
Dissecting Fertility Functions of Drosophila Y Chromosome Genes with CRISPR. Genetics 2020; 214:977-990. [PMID: 32098759 DOI: 10.1534/genetics.120.302672] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/20/2020] [Indexed: 12/27/2022] Open
Abstract
Gene-poor, repeat-rich regions of the genome are poorly understood and have been understudied due to technical challenges and the misconception that they are degenerating "junk." Yet multiple lines of evidence indicate these regions may be an important source of variation that could drive adaptation and species divergence, particularly through regulation of fertility. The ∼40 Mb Y chromosome of Drosophila melanoga st er contains only 16 known protein-coding genes, and is highly repetitive and entirely heterochromatic. Most of the genes originated from duplication of autosomal genes and have reduced nonsynonymous substitution rates, suggesting functional constraint. We devised a genetic strategy for recovering and retaining stocks with sterile Y-linked mutations and combined it with CRISPR to create mutants with deletions that disrupt three Y-linked genes. Two genes, PRY and FDY, had no previously identified functions. We found that PRY mutant males are subfertile, but FDY mutant males had no detectable fertility defects. FDY, the newest known gene on the Y chromosome, may have fertility effects that are conditional or too subtle to detect. The third gene, CCY, had been predicted but never formally shown to be required for male fertility. CRISPR targeting and RNA interference of CCY caused male sterility. Surprisingly, however, our CCY mutants were sterile even in the presence of an extra wild-type Y chromosome, suggesting that perturbation of the Y chromosome can lead to dominant sterility. Our approach provides an important step toward understanding the complex functions of the Y chromosome and parsing which functions are accomplished by genes vs. repeat elements.
Collapse
|
41
|
Dokshin GA, Davis GM, Sawle AD, Eldridge MD, Nicholls PK, Gourley TE, Romer KA, Molesworth LW, Tatnell HR, Ozturk AR, de Rooij DG, Hannon GJ, Page DC, Mello CC, Carmell MA. GCNA Interacts with Spartan and Topoisomerase II to Regulate Genome Stability. Dev Cell 2020; 52:53-68.e6. [PMID: 31839538 PMCID: PMC7227305 DOI: 10.1016/j.devcel.2019.11.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 08/14/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022]
Abstract
GCNA proteins are expressed across eukarya in pluripotent cells and have conserved functions in fertility. GCNA homologs Spartan (DVC-1) and Wss1 resolve DNA-protein crosslinks (DPCs), including Topoisomerase-DNA adducts, during DNA replication. Here, we show that GCNA mutants in mouse and C. elegans display defects in genome maintenance including DNA damage, aberrant chromosome condensation, and crossover defects in mouse spermatocytes and spontaneous genomic rearrangements in C. elegans. We show that GCNA and topoisomerase II (TOP2) physically interact in both mice and worms and colocalize on condensed chromosomes during mitosis in C. elegans embryos. Moreover, C. elegans gcna-1 mutants are hypersensitive to TOP2 poison. Together, our findings support a model in which GCNA provides genome maintenance functions in the germline and may do so, in part, by promoting the resolution of TOP2 DPCs.
Collapse
Affiliation(s)
- Gregoriy A Dokshin
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Gregory M Davis
- School of Health and Life Sciences, Federation University, VIC 3841, Australia
| | - Ashley D Sawle
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Matthew D Eldridge
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | | | - Taylin E Gourley
- School of Health and Life Sciences, Federation University, VIC 3841, Australia
| | - Katherine A Romer
- Whitehead Institute, 455 Main Street, Cambridge, MA 02142, USA; Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Luke W Molesworth
- School of Health and Life Sciences, Federation University, VIC 3841, Australia
| | - Hannah R Tatnell
- School of Health and Life Sciences, Federation University, VIC 3841, Australia
| | - Ahmet R Ozturk
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Dirk G de Rooij
- Whitehead Institute, 455 Main Street, Cambridge, MA 02142, USA; Reproductive Biology Group, Division of Developmental Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584, the Netherlands; Center for Reproductive Medicine, Academic Medical Center, University of Amsterdam 1105, the Netherlands
| | - Gregory J Hannon
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - David C Page
- Whitehead Institute, 455 Main Street, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| | - Craig C Mello
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Michelle A Carmell
- RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA; Whitehead Institute, 455 Main Street, Cambridge, MA 02142, USA; Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
42
|
Srivastav SP, Rahman R, Ma Q, Pierre J, Bandyopadhyay S, Lau NC. Har-P, a short P-element variant, weaponizes P-transposase to severely impair Drosophila development. eLife 2019; 8:49948. [PMID: 31845649 PMCID: PMC6917496 DOI: 10.7554/elife.49948] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/16/2019] [Indexed: 12/20/2022] Open
Abstract
Without transposon-silencing Piwi-interacting RNAs (piRNAs), transposition causes an ovarian atrophy syndrome in Drosophila called gonadal dysgenesis (GD). Harwich (Har) strains with P-elements cause severe GD in F1 daughters when Har fathers mate with mothers lacking P-element-piRNAs (i.e. ISO1 strain). To address the mystery of why Har induces severe GD, we bred hybrid Drosophila with Har genomic fragments into the ISO1 background to create HISR-D or HISR-N lines that still cause Dysgenesis or are Non-dysgenic, respectively. In these lines, we discovered a highly truncated P-element variant we named ‘Har-P’ as the most frequent de novo insertion. Although HISR-D lines still contain full-length P-elements, HISR-N lines lost functional P-transposase but retained Har-P’s that when crossed back to P-transposase restores GD induction. Finally, we uncovered P-element-piRNA-directed repression on Har-P’s transmitted paternally to suppress somatic transposition. The Drosophila short Har-P’s and full-length P-elements relationship parallels the MITEs/DNA-transposase in plants and SINEs/LINEs in mammals. DNA provides the instructions needed for life, a role that relies on it being a very stable and organized molecule. However, some sections of DNA are able to move from one place in the genome to another. When these “mobile genetic elements” move they may disrupt other genes and cause disease. For example, a mobile section of DNA known as the P-element causes a condition called gonadal dysgenesis in female fruit flies, leading to infertility. Only certain strains of fruit flies carry P-elements and the severity of gonadal dysgenesis in their daughters varies. For example, when male fruit flies of a strain known as Harwich (or Har for short) is crossed with female fruit flies that do not contain P-elements, all of their daughters develop severe gonadal dysgenesis and are infertile. However, if the cross is done the other way around, and female Har flies mate with males that do not contain P-elements, the daughters are fertile because the Har mothers provide their daughters with protective molecules that silence the P-elements. But it was a mystery as to why the P-elements from the Har fathers always caused such severe gonadal dysgenesis in all the daughters. Here, Srivastav et al. bred fruit flies to create offspring that had different pieces of Har DNA in a genetic background that was normally free from P-elements; they then analyzed the ‘hybrid’ offspring to identify which pieces of the Har genome caused gonadal dysgenesis in the daughter flies. These experiments showed that Har flies possess a very short variant of the P-element (named “Har-P”) that is more mobile than other variants. However, the Har-P variants still depended on an enzyme known as P-transposase encoded by the full-length P-elements to move around the genome. Further experiments showed that other strains of fruit flies that cause severe gonadal dysgenesis also had very short P-element variants that were almost identical to Har-P. These findings may explain why Har and some other strains of fruit flies drive severe gonadal dysgenesis. In the future, it may be possible to transfer P-transposase and Har-P into mosquitoes, ticks and other biting insects to make them infertile and help reduce the spread of certain diseases in humans.
Collapse
Affiliation(s)
- Satyam P Srivastav
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, United States
| | - Reazur Rahman
- Department of Biology, Brandeis University, Waltham, United States
| | - Qicheng Ma
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, United States
| | - Jasmine Pierre
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, United States
| | - Saptaparni Bandyopadhyay
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, United States
| | - Nelson C Lau
- Department of Biochemistry, Boston University School of Medicine, Boston University, Boston, United States.,Department of Biology, Brandeis University, Waltham, United States.,Genome Science Institute, Boston University School of Medicine, Boston, United States
| |
Collapse
|
43
|
Mačinković I, Theofel I, Hundertmark T, Kovač K, Awe S, Lenz J, Forné I, Lamp B, Nist A, Imhof A, Stiewe T, Renkawitz-Pohl R, Rathke C, Brehm A. Distinct CoREST complexes act in a cell-type-specific manner. Nucleic Acids Res 2019; 47:11649-11666. [PMID: 31701127 PMCID: PMC7145674 DOI: 10.1093/nar/gkz1050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 01/10/2023] Open
Abstract
CoREST has been identified as a subunit of several protein complexes that generate transcriptionally repressive chromatin structures during development. However, a comprehensive analysis of the CoREST interactome has not been carried out. We use proteomic approaches to define the interactomes of two dCoREST isoforms, dCoREST-L and dCoREST-M, in Drosophila. We identify three distinct histone deacetylase complexes built around a common dCoREST/dRPD3 core: A dLSD1/dCoREST complex, the LINT complex and a dG9a/dCoREST complex. The latter two complexes can incorporate both dCoREST isoforms. By contrast, the dLSD1/dCoREST complex exclusively assembles with the dCoREST-L isoform. Genome-wide studies show that the three dCoREST complexes associate with chromatin predominantly at promoters. Transcriptome analyses in S2 cells and testes reveal that different cell lineages utilize distinct dCoREST complexes to maintain cell-type-specific gene expression programmes: In macrophage-like S2 cells, LINT represses germ line-related genes whereas other dCoREST complexes are largely dispensable. By contrast, in testes, the dLSD1/dCoREST complex prevents transcription of germ line-inappropriate genes and is essential for spermatogenesis and fertility, whereas depletion of other dCoREST complexes has no effect. Our study uncovers three distinct dCoREST complexes that function in a lineage-restricted fashion to repress specific sets of genes thereby maintaining cell-type-specific gene expression programmes.
Collapse
Affiliation(s)
- Igor Mačinković
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Ina Theofel
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Tim Hundertmark
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Kristina Kovač
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Stephan Awe
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Jonathan Lenz
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| | - Ignasi Forné
- Protein Analysis Unit, BioMedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich, Großhadernerstrasse 9, 82152 Martinsried, Germany
| | - Boris Lamp
- Genomics Core Facility, Institute of Molecular Oncology, Philipps-University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Institute of Molecular Oncology, Philipps-University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Axel Imhof
- Protein Analysis Unit, BioMedical Center, Faculty of Medicine, Ludwig-Maximilians-University Munich, Großhadernerstrasse 9, 82152 Martinsried, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Institute of Molecular Oncology, Philipps-University, Hans-Meerwein-Strasse 3, 35043 Marburg, Germany
| | - Renate Renkawitz-Pohl
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Christina Rathke
- Department of Biology, Philipps-University, Karl-von-Frisch-Strasse 8, 35043, Marburg, Germany
| | - Alexander Brehm
- Institute of Molecular Biology and Tumor Research, Biomedical Research Center, Philipps-University, Hans-Meerwein-Strasse 2, 35043, Marburg, Germany
| |
Collapse
|
44
|
Štiavnická M, García-Álvarez O, Ulčová-Gallová Z, Sutovsky P, Abril-Parreño L, Dolejšová M, Řimnáčová H, Moravec J, Hošek P, Lošan P, Gold L, Fenclová T, Králíčková M, Nevoral J. H3K4me2 accompanies chromatin immaturity in human spermatozoa: an epigenetic marker for sperm quality assessment. Syst Biol Reprod Med 2019; 66:3-11. [DOI: 10.1080/19396368.2019.1666435] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Miriama Štiavnická
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Olga García-Álvarez
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Health and Biotechnology (SaBio) GroupIREC, (CSIC-UCLM-JCCM), Albacete, Spain
| | - Zděnka Ulčová-Gallová
- Genetika Plzeň, s.r.o. (Ltd.), Pilsen – Černice, Czech Republic
- Department of Gynecology and Obstetrics, Charles University, Pilsen, Czech Republic
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, USA
| | - Laura Abril-Parreño
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Laboratory of Animal Reproduction, Department of Biological Sciences, University of Limerick, Limerick, Irelan
| | - Martina Dolejšová
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Hedvika Řimnáčová
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Jiří Moravec
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Hošek
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Lošan
- Genetika Plzeň, s.r.o. (Ltd.), Pilsen – Černice, Czech Republic
| | - Lukáš Gold
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Tereza Fenclová
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Milena Králíčková
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Plzeň, Czech Republic
| | - Jan Nevoral
- Biomedical Center in Pilsen, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Plzeň, Czech Republic
| |
Collapse
|
45
|
The dynamics and regulation of chromatin remodeling during spermiogenesis. Gene 2019; 706:201-210. [DOI: 10.1016/j.gene.2019.05.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 01/06/2023]
|
46
|
Gärtner SM, Hundertmark T, Nolte H, Theofel I, Eren-Ghiani Z, Tetzner C, Duchow TB, Rathke C, Krüger M, Renkawitz-Pohl R. Stage-specific testes proteomics of Drosophila melanogaster identifies essential proteins for male fertility. Eur J Cell Biol 2019; 98:103-115. [DOI: 10.1016/j.ejcb.2019.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 02/01/2023] Open
|
47
|
Chen T, Sun Z, Mu S, Jiang L, Li C, Li L, Guo M, Zhang Z, Kang X. Ultrastructure of spermiogenesis and the distribution of spermatozoal nuclear histones in the Japanese mantis shrimp, Oratosquilla oratoria (Crustacea: Stomatopoda). J Morphol 2019; 280:1170-1184. [PMID: 31141207 PMCID: PMC6771690 DOI: 10.1002/jmor.21008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/11/2019] [Accepted: 05/14/2019] [Indexed: 12/30/2022]
Abstract
The Japanese mantis shrimp Oratosquilla oratoria (Stomatopoda; Crustacea) is one of the most economically important aquatic species of Pacific shrimp and it is distributed from Japan to the coast of China, the Philippines, the Malay Peninsula, and the Hawaiian Islands. Early studies described certain characteristics of spermatogenesis and the sperm ultrastructure in Stomatopoda, but the composition of sperm basic nuclear proteins (SBNPs) remains completely unknown. We studied the sperm ultrastructure of O. oratoria using transmission electron microscopy and the histone composition using immunofluorescence and immunoelectron microscopy. We found that the spherical nucleus is adjacent to the electron translucent external coat, which occurs in early spermatids. The acrosomal structure begins to form at the junction of the nucleus and the external coat. At the mid-spermatid stage, part of the chromatin appears to be more electron-dense than the external coat side. The aflagellate sperm of O. oratoria, are rounded or slightly ovoid in shape and have a consistent granular nucleus, an acrosome structure of pushpin shape and a spherical vesicular body in which faintly granular material is scattered. The acrosome consists of an acrosomal vesicle, perforatorium, and subacrosomal material. The sperm contains histones H2A, H2B, H3, H4, H3.3, H2AX, and H2AZ as well as some histone modifications, that is, H3K9me3, H3K4me2, H3S10ph, H4Kac, and H2A + H4S1ph. Histones are localized not only in the nucleus of the sperm but also in other structures outside the nucleus. The results may provide new perspectives for systematic studies of crustaceans and their sperm chromatin components. These findings extend the study of the sperm structure of Stomatopoda and provide basic data to elucidate the epigenetic mechanism of fertilization.
Collapse
Affiliation(s)
- Tingrong Chen
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Zhe Sun
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Shumei Mu
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Lingling Jiang
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Chao Li
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Lu Li
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Mingshen Guo
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Zhaohui Zhang
- Department of Reproductive Medicine, Baoding No. 1 Central Hospital, Baoding, Hebei, China
| | - Xianjiang Kang
- College of Life Sciences, Hebei University, Baoding, Hebei, China
| |
Collapse
|
48
|
Drosophila Pif1A is essential for spermatogenesis and is the homolog of human CCDC157, a gene associated with idiopathic NOA. Cell Death Dis 2019; 10:125. [PMID: 30741974 PMCID: PMC6370830 DOI: 10.1038/s41419-019-1398-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 01/05/2023]
Abstract
The dynamic process of spermatogenesis shows little variation between invertebrate models such as Drosophila, and vertebrate models such as mice and rats. In each case, germ stem cells undergo mitotic division to proliferate and then continue, via meiosis, through various stages of elongation and individualization from spermatogonia to spermatid to finally to form mature sperm. Mature sperm are then stored in the seminal vesicles for fertilization. Errors in any of these stages can lead to male infertility. Here, we identify that Drosophila Pif1A acts as a key regulator for sperm individualization. Loss of Pif1A leads to male sterility associated with irregular individualization complex and empty seminal vesicles without mature sperm. Pif1A is highly expressed in the testes of mated male adult flies and the Pif1A protein is expressed at a higher level in male than in female flies. Pif1A is homologous to mammalian coiled-coil domain-containing protein 157 (CCDC157), which is also enriched in the testes of humans and mice. Human CCDC157, with unknown function, was identified to be downregulated in men with idiopathic non-obstructive azoospermia (NOA). We map the function of Drosophila Pif1A during spermatogenesis, showing that Pif1A is essential for spermatide individualization and involved in the regulation of the lipid metabolism genes. Our findings might be applicable for studying the function of CCDC157 in spermatogenesis and other aspects of human male fertility.
Collapse
|
49
|
Genetic Instability and Chromatin Remodeling in Spermatids. Genes (Basel) 2019; 10:genes10010040. [PMID: 30646585 PMCID: PMC6356297 DOI: 10.3390/genes10010040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/13/2022] Open
Abstract
The near complete replacement of somatic chromatin in spermatids is, perhaps, the most striking nuclear event known to the eukaryotic domain. The process is far from being fully understood, but research has nevertheless unraveled its complexity as an expression of histone variants and post-translational modifications that must be finely orchestrated to promote the DNA topological change and compaction provided by the deposition of protamines. That this major transition may not be genetically inert came from early observations that transient DNA strand breaks were detected in situ at chromatin remodeling steps. The potential for genetic instability was later emphasized by our demonstration that a significant number of DNA double-strand breaks (DSBs) are formed and then repaired in the haploid context of spermatids. The detection of DNA breaks by 3'OH end labeling in the whole population of spermatids suggests that a reversible enzymatic process is involved, which differs from canonical apoptosis. We have set the stage for a better characterization of the genetic impact of this transition by showing that post-meiotic DNA fragmentation is conserved from human to yeast, and by providing tools for the initial mapping of the genome-wide DSB distribution in the mouse model. Hence, the molecular mechanism of post-meiotic DSB formation and repair in spermatids may prove to be a significant component of the well-known male mutation bias. Based on our recent observations and a survey of the literature, we propose that the chromatin remodeling in spermatids offers a proper context for the induction of de novo polymorphism and structural variations that can be transmitted to the next generation.
Collapse
|
50
|
Augière C, Lapart JA, Duteyrat JL, Cortier E, Maire C, Thomas J, Durand B. salto/CG13164 is required for sperm head morphogenesis in Drosophila. Mol Biol Cell 2019; 30:636-645. [PMID: 30601696 PMCID: PMC6589691 DOI: 10.1091/mbc.e18-07-0429] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Producing mature spermatozoa is essential for sexual reproduction in metazoans. Spermiogenesis involves dramatic cell morphological changes going from sperm tail elongation and nuclear reshaping to cell membrane remodeling during sperm individualization and release. The sperm manchette plays a critical scaffolding function during nuclear remodeling by linking the nuclear lamina to the cytoskeleton. Here, we describe the role of an uncharacterized protein in Drosophila, salto/CG13164, involved in nuclear shaping and spermatid individualization. Salto has dynamic localization during spermatid differentiation, being progressively relocated from the sperm-nuclear dense body, which is equivalent to the mammalian sperm manchette, to the centriolar adjunct and acrosomal cap during spermiogenesis. salto-null male flies are sterile and exhibit complete spermatid individualization defects. salto-deficient spermatids show coiled spermatid nuclei at late maturation stages and stalled individualization complexes. Our work sheds light on a novel component involved in cytoskeleton-based cell-morphological changes during spermiogenesis.
Collapse
Affiliation(s)
- Céline Augière
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| | - Jean-André Lapart
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| | - Jean-Luc Duteyrat
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| | - Elisabeth Cortier
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| | - Charline Maire
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| | - Joëlle Thomas
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| | - Bénédicte Durand
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, Institut NeuroMyoGène, F-69008 Lyon, France
| |
Collapse
|