1
|
Price KL, Tharakan DM, Salvenmoser W, Ayers K, Mah J, Dunn C, Hobmayer B, Cooley L. Examination of germline and somatic intercellular bridges in Hydra vulgaris reveals insights into the evolutionarily conserved mechanism of intercellular bridge formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639158. [PMID: 40027827 PMCID: PMC11870534 DOI: 10.1101/2025.02.19.639158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Incomplete cytokinesis results in the formation of stable intercellular bridges that have been extensively studied in bilaterians, where they play essential roles in cell-cell communication and coordination of differentiation. However, little is known about their structure and molecular composition in non-bilaterian animals. This study characterizes germline and somatic intercellular bridges in the cnidarian Hydra vulgaris, providing insights into their evolutionary origins and functional significance. We identified key conserved components, including KIF23, F-actin, and phosphotyrosine. Notably, we observed microtubule localization within Hydra ring canals, suggesting previously unrecognized functions for this cytoskeletal component in intercellular bridge formation. Bioinformatic analyses confirmed the conserved expression of Kif23 and suggested its role as a molecular marker for identifying ring canal-associated components. EdU incorporation during DNA replication demonstrated that cells connected by ring canals exhibit synchronized cell cycles, which may be critical for the coordination of division and differentiation. Our findings reveal that the molecular and structural features of intercellular bridges in Hydra are conserved across evolutionary lineages, highlighting their ancient origins and functional significance in cellular connectivity. The presence of synchronized cell cycles in ring canal-connected cells underscores their role in promoting coordinated cellular behaviors, processes fundamental to multicellular organization. This study provides new perspectives on the evolution of incomplete cytokinesis and establishes a framework for comparative investigations into the diversity and conservation of intercellular bridge mechanisms across metazoans.
Collapse
Affiliation(s)
- Kari L. Price
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- UC Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Dyuthi M. Tharakan
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Willi Salvenmoser
- Institute of Zoology and Centre for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Kathleen Ayers
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Jasmine Mah
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Casey Dunn
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
| | - Bert Hobmayer
- Institute of Zoology and Centre for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - Lynn Cooley
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT, USA
- Lead contact
| |
Collapse
|
2
|
Sorkin J, Tilton K, Lawlor MA, Sarathy SN, Liang S, Albanese A, Rabbani M, Hammoud SS, Ellison CE, Pratto F, Jain D. Intercellular bridges are essential for transposon repression and meiosis in the male germline. Nat Commun 2025; 16:1488. [PMID: 39929837 PMCID: PMC11811169 DOI: 10.1038/s41467-025-56742-9] [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: 09/04/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
Germ cell connectivity via intercellular bridges is a widely conserved feature across metazoans. However, its functional significance is poorly understood. Intercellular bridges are essential for fertility in male mice as genetic ablation of a critical bridge component, TEX14, causes spermatogenic failure, but the underlying reasons are unknown. Here we utilized a Tex14 hypomorph with reduced intercellular bridges along with Tex14-null mice that completely lack bridges to examine the roles of germ cell connectivity during spermatogenesis. We report that in males deficient for TEX14 and intercellular bridges, germ cells fail to complete meiotic DNA replication, synapsis and meiotic double-strand break repair. They also derepress retrotransposons and accumulate retrotransposon-encoded proteins during meiosis. Single-cell RNA-sequencing confirms sharing of transcripts between wild-type spermatids and demonstrates its partial attenuation in Tex14 hypomorphs, indicating that intercellular bridges enable cytoplasmic exchange between connected germ cells in testes. Our findings suggest that regulation of meiosis is non-cell-intrinsic and inform a model in which intercellular bridges influence critical meiotic events and protect germline genome integrity during spermatogenesis.
Collapse
Affiliation(s)
- Julia Sorkin
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Kevin Tilton
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Matthew A Lawlor
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Shreya N Sarathy
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Shun Liang
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Angelina Albanese
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Mashiat Rabbani
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Saher S Hammoud
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Christopher E Ellison
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | | | - Devanshi Jain
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
3
|
Levy EW, Leite I, Joyce BW, Shvartsman SY, Posfai E. A tug-of-war between germ cell motility and intercellular bridges controls germline cyst formation in mice. Curr Biol 2024; 34:5728-5738.e4. [PMID: 39566500 DOI: 10.1016/j.cub.2024.10.062] [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: 06/04/2024] [Revised: 10/08/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024]
Abstract
Gametes in many species develop in cysts-clusters of germ cells formed by incomplete cytokinesis-that remain connected through intercellular bridges (ICBs). These connections enable sharing of cytoplasmic components between germ cells and, in the female germ line, enrich select cells in the cyst to become the oocyte(s). In mice, germline cysts of variable sizes are generated during embryonic development, thought to result from cyst fractures. Studies of fixed samples failed to capture fracture events, and thus, the mechanism remained elusive. Here, we use high-resolution live imaging of germ cells within their native tissue environment to visualize germline cyst dynamics. With this novel approach, we reveal a striking motile phenotype of gonad-resident germ cells and show that this randomly oriented cell-autonomous motile behavior during cyst formation underlies fracture events. Conversely, we show that stabilized ICBs help resist excessive fracturing. Additionally, we find that motility and thus fracture rates gradually decrease during development in a sex-dependent manner, completely ceasing by the end of cyst-forming divisions. These results lead to a model where the opposing activities of developmentally regulated cell motility and stable ICBs give rise to cysts of variable sizes. We corroborate these results by developing a model that uses experimentally measured fracture rates to simulate cyst formation and fracture and show that it can reproduce experimentally measured cyst sizes in both male and female. Understanding how variable cysts form will enable further studies of mammalian oocyte selection and establishment of the ovarian reserve.
Collapse
Affiliation(s)
- Ezra W Levy
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Isabella Leite
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA; Quantitative and Computational Biology Program, Lewis-Sigler Institute for Integrative Genomics, Washington Road, Princeton, NJ 08544, USA
| | - Bradley W Joyce
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Stanislav Y Shvartsman
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA; Quantitative and Computational Biology Program, Lewis-Sigler Institute for Integrative Genomics, Washington Road, Princeton, NJ 08544, USA; Developmental Dynamics Group, Center for Computational Biology, Flatiron Institute, 5th Avenue, New York, NY 10010, USA
| | - Eszter Posfai
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA.
| |
Collapse
|
4
|
Elkouby YM. Germ cell development: Anchoring and pulling forces shape germline cyst construction. Curr Biol 2024; 34:R1228-R1230. [PMID: 39689689 DOI: 10.1016/j.cub.2024.10.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
A new study reports a 'tug-of-war' mechanism in mouse germline cyst formation, where cell motility and intercellular bridges balance fragmentation and stabilization of the cyst. These dynamic and opposing forces that anchor and pull cells apart shape cyst construction, advancing our understanding of mammalian oogenesis and reproduction.
Collapse
Affiliation(s)
- Yaniv M Elkouby
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem, Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel; Institute for Medical Research - Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel.
| |
Collapse
|
5
|
Wong HN, Chen T, Wang PJ, Holzman LB. ARF6, a component of intercellular bridges, is essential for spermatogenesis in mice. Dev Biol 2024; 508:46-63. [PMID: 38242343 PMCID: PMC10979378 DOI: 10.1016/j.ydbio.2024.01.007] [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: 07/21/2023] [Revised: 01/05/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
Male germ cells are connected by intercellular bridges (ICBs) in a syncytium due to incomplete cytokinesis. Syncytium is thought to be important for synchronized germ cell development by interchange of cytoplasmic factors via ICBs. Mammalian ADP-ribosylation factor 6 (ARF6) is a small GTPase that is involved in many cellular mechanisms including but not limited to regulating cellular structure, motility, vesicle trafficking and cytokinesis. ARF6 localizes to ICBs in spermatogonia and spermatocytes in mice. Here we report that mice with global depletion of ARF6 in adulthood using Ubc-CreERT2 display no observable phenotypes but are male sterile. ARF6-deficient males display a progressive loss of germ cells, including LIN28A-expressing spermatogonia, and ultimately develop Sertoli-cell-only syndrome. Specifically, intercellular bridges are lost in ARF6-deficient testis. Furthermore, germ cell-specific inactivation using the Ddx4-CreERT2 results in the same testicular morphological phenotype, showing the germ cell-intrinsic requirement of ARF6. Therefore, ARF6 is essential for spermatogenesis in mice and this function is conserved from Drosophila to mammals.
Collapse
Affiliation(s)
- Hetty N Wong
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Tingfang Chen
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - P Jeremy Wang
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, 19104, USA
| | - Lawrence B Holzman
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Shi H, Li QY, Li H, Wang HY, Fan CX, Dong QY, Pan BC, Ji ZL, Li JY. ROS-induced oxidative stress is a major contributor to sperm cryoinjury. Hum Reprod 2024; 39:310-325. [PMID: 38011909 DOI: 10.1093/humrep/dead250] [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: 03/29/2023] [Revised: 11/13/2023] [Indexed: 11/29/2023] Open
Abstract
STUDY QUESTION What is the mechanism behind cryoinjury in human sperm, particularly concerning the interplay between reactive oxygen species (ROS) and autophagy, and how does it subsequently affect sperm fate? SUMMARY ANSWER The freeze-thaw operation induces oxidative stress by generating abundant ROS, which impairs sperm motility and activates autophagy, ultimately guiding the sperm toward programmed cell death such as apoptosis and necrosis, as well as triggering premature capacitation. WHAT IS KNOWN ALREADY Both ROS-induced oxidative stress and autophagy are thought to exert an influence on the quality of frozen-thawed sperm. STUDY DESIGN, SIZE, DURATION Overall, 84 semen specimens were collected from young healthy fertile males, with careful quality evaluation. The specimens were split into three groups to investigate the ROS-induced cryoinjury: normal control without any treatment, sperm treated with 0.5 mM hydrogen peroxide (H2O2) for 1 h, and sperm thawed following cryopreservation. Samples from 48 individuals underwent computer-assisted human sperm analysis (CASA) to evaluate sperm quality in response to the treatments. Semen samples from three donors were analyzed for changes in the sperm proteome after H2O2 treatment, and another set of samples from three donors were analyzed for changes following the freeze-thaw process. The other 30 samples were used for fluorescence-staining and western blotting. PARTICIPANTS/MATERIALS, SETTING, METHODS Sperm motility parameters, including progressive motility (PR %) and total motility (PR + NP %), were evaluated using the CASA system on a minimum of 200 spermatozoa. The proteomic profiles were determined with label-free mass spectrometry (MS/MS) and protein identification was performed via ion search against the NCBI human database. Subsequently, comprehensive bioinformatics was applied to detect significant proteomic changes and functional enrichment. Fluorescence-staining and western blot analyses were also conducted to confirm the proteomic changes on selected key proteins. The ROS level was measured using 2',7'-dichlorodihydrofluorescein diacetate labeling and the abundance of bioactive mitochondria was determined by evaluating the inner mitochondrial membrane potential (MMP) level. Molecular behaviors of sequestosome-1 (p62 or SQSTM1) and microtubule-associated proteins 1A/1B light chain 3 (LC3) were monitored to evaluate the state of apoptosis in human sperm. Fluorescent probes oxazole yellow (YO-PRO-1) and propidium iodide (PI) were utilized to monitor programmed cell death, namely apoptosis and necrosis. Additionally, gradient concentrations of antioxidant coenzyme Q10 (CoQ10) were introduced to suppress ROS impacts on sperm. MAIN RESULTS AND THE ROLE OF CHANCE The CASA analysis revealed a significant decrease in sperm motility for both the H2O2-treatment and freeze-thaw groups. Fluorescence staining showed that high ROS levels were produced in the treated sperm and the MMPs were largely reduced. The introduction of CoQ10 at concentrations of 20 and 30 μM resulted in a significant rescue of progressive motility (P < 0.05). The result suggested that excessive ROS could be the major cause of sperm motility impairment, likely by damaging mitochondrial energy generation. Autophagy was significantly activated in sperm when they were under oxidative stress, as evidenced by the upregulation of p62 and the increased conversion of LC3 as well as the upregulation of several autophagy-related proteins, such as charged multivesicular body protein 2a, mitochondrial import receptor subunit TOM22 homolog, and WD repeat domain phosphoinositide-interacting protein 2. Additionally, fluorescent staining indicated the occurrence of apoptosis and necrosis in both H2O2-treated sperm and post-thaw sperm. The cell death process can be suppressed when CoQ10 is introduced, which consolidates the view that ROS could be the major contributor to sperm cryoinjury. The freeze-thaw process could also initiate sperm premature capacitation, demonstrated by the prominent increase in tyrosine phosphorylated proteins, verified with anti-phosphotyrosine antibody and immunofluorescence assays. The upregulation of capacitation-related proteins, such as hyaluronidase 3 and Folate receptor alpha, supported this finding. LARGE SCALE DATA The data underlying this article are available in the article and its online supplementary material. LIMITATIONS, REASONS FOR CAUTION The semen samples were obtained exclusively from young, healthy, and fertile males with progressive motility exceeding 60%, which might overemphasize the positive effects while possibly neglecting the negative impacts of cryoinjury. Additionally, the H2O2 treatment conditions in this study may not precisely mimic the oxidative stress experienced by sperm after thawing from cryopreservation, potentially resulting in the omission of certain molecular alterations. WIDER IMPLICATIONS OF THE FINDINGS This study provides substantial proteomic data for a comprehensive and deeper understanding of the impact of cryopreservation on sperm quality. It will facilitate the design of optimal protocols for utilizing cryopreserved sperm to improve applications, such as ART, and help resolve various adverse situations caused by chemotherapy, radiotherapy, and surgery. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from the Major Innovation Project of Research Institute of National Health Commission (#2022GJZD01-3) and the National Key R&D Program of China (#2018YFC1003600). All authors declare no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Hui Shi
- College of Life Science, Yantai University, Yantai, Shandong, China
| | - Qian-Ying Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Hui Li
- College of Life Science, Yantai University, Yantai, Shandong, China
| | - Hai-Yan Wang
- College of Life Science, Yantai University, Yantai, Shandong, China
| | - Chuan-Xi Fan
- College of Life Science, Yantai University, Yantai, Shandong, China
| | - Qiao-Yan Dong
- College of Life Science, Yantai University, Yantai, Shandong, China
| | - Bo-Chen Pan
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhi-Liang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jian-Yuan Li
- Institute of Science and Technology, National Health Commission, Beijing, China
| |
Collapse
|
7
|
Czukiewska SM, Fan X, Mulder AA, Van Der Helm T, Hillenius S, Van Der Meeren L, Matorras R, Eguizabal C, Lei L, Koning RI, Chuva De Sousa Lopes SM. Cell-cell interactions during the formation of primordial follicles in humans. Life Sci Alliance 2023; 6:e202301926. [PMID: 37643865 PMCID: PMC10465921 DOI: 10.26508/lsa.202301926] [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: 01/14/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Gametogenesis is a complex and sex-specific multistep process during which the gonadal somatic niche plays an essential regulatory role. One of the most crucial steps during human female gametogenesis is the formation of primordial follicles, the functional unit of the ovary that constitutes the pool of follicles available at birth during the entire reproductive life. However, the relation between human fetal germ cells (hFGCs) and gonadal somatic cells during the formation of the primordial follicles remains largely unexplored. We have discovered that hFGCs can form multinucleated syncytia, some connected via interconnecting intercellular bridges, and that not all nuclei in hFGC-syncytia were synchronous regarding meiotic stage. As hFGCs progressed in development, pre-granulosa cells formed protrusions that seemed to progressively constrict individual hFGCs, perhaps contributing to separate them from the multinucleated syncytia. Our findings highlighted the cell-cell interaction and molecular dynamics between hFGCs and (pre)granulosa cells during the formation of primordial follicles in humans. Knowledge on how the pool of primordial follicle is formed is important to understand human infertility.
Collapse
Affiliation(s)
- Sylwia M Czukiewska
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Xueying Fan
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Adriaan A Mulder
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Talia Van Der Helm
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Sanne Hillenius
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Lotte Van Der Meeren
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Roberto Matorras
- IVIRMA, IVI Bilbao, Bilbao, Spain
- Human Reproduction Unit, Cruces University Hospital, Bilbao, Spain
- Department of Obstetrics and Gynecology, Basque Country University, Bilbao, Spain
- Biocruces Bizkaia Health Research Institute, Bilbao, Spain
| | - Cristina Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Lei Lei
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA
| | - Roman I Koning
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Susana M Chuva De Sousa Lopes
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department for Reproductive Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
8
|
Ikami K, Shoffner-Beck S, Tyczynska Weh M, Schnell S, Yoshida S, Diaz Miranda EA, Ko S, Lei L. Branched germline cysts and female-specific cyst fragmentation facilitate oocyte determination in mice. Proc Natl Acad Sci U S A 2023; 120:e2219683120. [PMID: 37155904 PMCID: PMC10194012 DOI: 10.1073/pnas.2219683120] [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: 11/17/2022] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
During mouse gametogenesis, germ cells derived from the same progenitor are connected via intercellular bridges forming germline cysts, within which asymmetrical or symmetrical cell fate occurs in female and male germ cells, respectively. Here, we have identified branched cyst structures in mice, and investigated their formation and function in oocyte determination. In fetal female cysts, 16.8% of the germ cells are connected by three or four bridges, namely branching germ cells. These germ cells are preferentially protected from cell death and cyst fragmentation and accumulate cytoplasm and organelles from sister germ cells to become primary oocytes. Changes in cyst structure and differential cell volumes among cyst germ cells suggest that cytoplasmic transport in germline cysts is conducted in a directional manner, in which cellular content is first transported locally between peripheral germ cells and further enriched in branching germ cells, a process causing selective germ cell loss in cysts. Cyst fragmentation occurs extensively in female cysts, but not in male cysts. Male cysts in fetal and adult testes have branched cyst structures, without differential cell fates between germ cells. During fetal cyst formation, E-cadherin (E-cad) junctions between germ cells position intercellular bridges to form branched cysts. Disrupted junction formation in E-cad-depleted cysts led to an altered ratio in branched cysts. Germ cell-specific E-cad knockout resulted in reductions in primary oocyte number and oocyte size. These findings shed light on how oocyte fate is determined within mouse germline cysts.
Collapse
Affiliation(s)
- Kanako Ikami
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI48109
- Buck Institute for Research on Aging, Novato, CA94945
| | - Suzanne Shoffner-Beck
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Malgorzata Tyczynska Weh
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Santiago Schnell
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI48109
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, Okazaki, Aichi444-8585, Japan
- Graduate Institute for Advanced Studies, Sokendai, Okazaki, Aichi444-8585, Japan
| | - Edgar Andres Diaz Miranda
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO65211
| | - Sooah Ko
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO65211
| | - Lei Lei
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO65211
- Division of Biological Sciences, College of Arts and Sciences, University of Missouri, Columbia, MO65211
| |
Collapse
|
9
|
Pan B, Yuan S, Mayernik L, Yap YT, Moin K, Chung CS, Maddipati K, Krawetz SA, Zhang Z, Hess RA, Chen X. Disrupted intercellular bridges and spermatogenesis in fatty acyl-CoA reductase 1 knockout mice: A new model of ether lipid deficiency. FASEB J 2023; 37:e22908. [PMID: 37039784 PMCID: PMC10150578 DOI: 10.1096/fj.202201848r] [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: 11/09/2022] [Revised: 03/10/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023]
Abstract
Peroxisomal fatty acyl-CoA reductase 1 (FAR1) is a rate-limiting enzyme for ether lipid (EL) synthesis. Gene mutations in FAR1 cause a rare human disease. Furthermore, altered EL homeostasis has also been associated with various prevalent human diseases. Despite their importance in human health, the exact cellular functions of FAR1 and EL are not well-understood. Here, we report the generation and initial characterization of the first Far1 knockout (KO) mouse model. Far1 KO mice were subviable and displayed growth retardation. The adult KO male mice had smaller testes and were infertile. H&E and immunofluorescent staining showed fewer germ cells in seminiferous tubules. Round spermatids were present but no elongated spermatids or spermatozoa were observed, suggesting a spermatogenesis arrest at this stage. Large multi-nucleated giant cells (MGC) were found lining the lumen of seminiferous tubules with many of them undergoing apoptosis. The immunofluorescent signal of TEX14, an essential component of intercellular bridges (ICB) between developing germ cells, was greatly reduced and mislocalized in KO testis, suggesting the disrupted ICBs as an underlying cause of MGC formation. Integrative analysis of our total testis RNA-sequencing results and published single-cell RNA-sequencing data unveiled cell type-specific molecular alterations underlying the spermatogenesis arrest. Many genes essential for late germ cell development showed dramatic downregulation, whereas genes essential for extracellular matrix dynamics and cell-cell interactions were among the most upregulated genes. Together, this work identified the cell type-specific requirement of ELs in spermatogenesis and suggested a critical role of Far1/ELs in the formation/maintenance of ICB during meiosis.
Collapse
Affiliation(s)
- Bo Pan
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Shuo Yuan
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
- Department of Occupational and Environmental Medicine, School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Linda Mayernik
- Department of Pharmacology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Yi Tian Yap
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Kamiar Moin
- Department of Pharmacology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Charles S. Chung
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Krishnarao Maddipati
- Department of Pathology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| | - Stephen A. Krawetz
- Department of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics & Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Rex A. Hess
- Comparative Biosciences, College of Veterinary Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Xuequn Chen
- Department of Physiology, Wayne State University, School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
10
|
Mytlis A, Levy K, Elkouby YM. The many faces of the bouquet centrosome MTOC in meiosis and germ cell development. Curr Opin Cell Biol 2023; 81:102158. [PMID: 36913831 DOI: 10.1016/j.ceb.2023.102158] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/28/2022] [Accepted: 02/12/2023] [Indexed: 03/13/2023]
Abstract
Meiotic chromosomal pairing is facilitated by a conserved cytoskeletal organization. Telomeres associate with perinuclear microtubules via Sun/KASH complexes on the nuclear envelope (NE) and dynein. Telomere sliding on perinuclear microtubules contributes to chromosome homology searches and is essential for meiosis. Telomeres ultimately cluster on the NE, facing the centrosome, in a configuration called the chromosomal bouquet. Here, we discuss novel components and functions of the bouquet microtubule organizing center (MTOC) in meiosis, but also broadly in gamete development. The cellular mechanics of chromosome movements and the bouquet MTOC dynamics are striking. The newly identified zygotene cilium mechanically anchors the bouquet centrosome and completes the bouquet MTOC machinery in zebrafish and mice. We hypothesize that various centrosome anchoring strategies evolved in different species. Evidence suggests that the bouquet MTOC machinery is a cellular organizer, linking meiotic mechanisms with gamete development and morphogenesis. We highlight this cytoskeletal organization as a new platform for creating a holistic understanding of early gametogenesis, with direct implications to fertility and reproduction.
Collapse
Affiliation(s)
- Avishag Mytlis
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem, 9112102, Israel; Institute for Medical Research - Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Karine Levy
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem, 9112102, Israel; Institute for Medical Research - Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Yaniv M Elkouby
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem, 9112102, Israel; Institute for Medical Research - Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel.
| |
Collapse
|
11
|
Polyakova N, Kalashnikova M, Belyavsky A. Non-Classical Intercellular Communications: Basic Mechanisms and Roles in Biology and Medicine. Int J Mol Sci 2023; 24:ijms24076455. [PMID: 37047428 PMCID: PMC10095225 DOI: 10.3390/ijms24076455] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
In multicellular organisms, interactions between cells and intercellular communications form the very basis of the organism’s survival, the functioning of its systems, the maintenance of homeostasis and adequate response to the environment. The accumulated experimental data point to the particular importance of intercellular communications in determining the fate of cells, as well as their differentiation and plasticity. For a long time, it was believed that the properties and behavior of cells were primarily governed by the interactions of secreted or membrane-bound ligands with corresponding receptors, as well as direct intercellular adhesion contacts. In this review, we describe various types of other, non-classical intercellular interactions and communications that have recently come into the limelight—in particular, the broad repertoire of extracellular vesicles and membrane protrusions. These communications are mediated by large macromolecular structural and functional ensembles, and we explore here the mechanisms underlying their formation and present current data that reveal their roles in multiple biological processes. The effects mediated by these new types of intercellular communications in normal and pathological states, as well as therapeutic applications, are also discussed. The in-depth study of novel intercellular interaction mechanisms is required for the establishment of effective approaches for the control and modification of cell properties both for basic research and the development of radically new therapeutic strategies.
Collapse
Affiliation(s)
- Natalia Polyakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| | - Maria Kalashnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova 5A, 117485 Moscow, Russia
| | - Alexander Belyavsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerova 5A, 117485 Moscow, Russia
- Correspondence:
| |
Collapse
|
12
|
Price KL, Tharakan DM, Cooley L. Evolutionarily conserved midbody remodeling precedes ring canal formation during gametogenesis. Dev Cell 2023; 58:474-488.e5. [PMID: 36898376 PMCID: PMC10059090 DOI: 10.1016/j.devcel.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/18/2022] [Accepted: 02/10/2023] [Indexed: 03/12/2023]
Abstract
How canonical cytokinesis is altered during germ cell division to produce stable intercellular bridges, called "ring canals," is poorly understood. Here, using time-lapse imaging in Drosophila, we observe that ring canal formation occurs through extensive remodeling of the germ cell midbody, a structure classically associated with its function in recruiting abscission-regulating proteins in complete cytokinesis. Germ cell midbody cores reorganize and join the midbody ring rather than being discarded, and this transition is accompanied by changes in centralspindlin dynamics. The midbody-to-ring canal transformation is conserved in the Drosophila male and female germlines and during mouse and Hydra spermatogenesis. In Drosophila, ring canal formation depends on Citron kinase function to stabilize the midbody, similar to its role during somatic cell cytokinesis. Our results provide important insights into the broader functions of incomplete cytokinesis events across biological systems, such as those observed during development and disease states.
Collapse
Affiliation(s)
- Kari L Price
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Dyuthi M Tharakan
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Lynn Cooley
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA; Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT, USA.
| |
Collapse
|
13
|
Liu M, Liu S, Song C, Zhu H, Wu B, Zhang A, Zhao H, Wen Z, Gao J. Pre-meiotic deletion of PEX5 causes spermatogenesis failure and infertility in mice. Cell Prolif 2023; 56:e13365. [PMID: 36433756 PMCID: PMC9977671 DOI: 10.1111/cpr.13365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/27/2022] Open
Abstract
Peroxisomes are involved in the regulation of various pathological processes. Peroxisomal biogenesis factor 5 (PEX5), which plays an essential role in peroxisomal biogenesis, is critical for reactive oxygen species (ROS) accumulation. However, its underlying functions in spermatogenesis have not yet been identified. Pex5 was deleted by crossing Stra8-Cre mice with Pex5flox/flox mice before the onset of meiosis. The morphology of testes and epididymides, spermatogenesis function, and fertility in both wild type (WT) and Pex5-/- mice were analysed by haematoxylin and eosin (HE) and immunofluorescent staining. Mechanism of PEX5 affecting peroxisomes and spermatogenesis were validated by Western blot and transmission electron microscopy (TEM). Transcriptome RNA sequencing (RNA-seq) was used to profile the dysregulated genes in testes from WT and Pex5-/- mice on postnatal day (P) 35. The adult Pex5 knockout male mice were completely sterile with no mature sperm production. Loss of Pex5 in spermatocytes resulted in multinucleated giant cell formation, meiotic arrest, abnormal tubulin expression, and deformed acrosome formation. Furthermore, Pex5 deletion led to delayed DNA double-strand break repair and improper crossover at the pachytene stage. Impaired peroxisome function in Pex5 knockout mice induced ROS redundancy, which in turn led to an increase in germ cell apoptosis and a decline in autophagy. Pex5 regulates ROS during meiosis and is essential for spermatogenesis and male fertility in mice.
Collapse
Affiliation(s)
- Min Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Shuangyuan Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Chenyang Song
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Haixia Zhu
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Bin Wu
- Department of Reproductive Medicine, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Aizhen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Hui Zhao
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Zongzhuang Wen
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Jiangang Gao
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China.,School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| |
Collapse
|
14
|
Overland MR, Li Y, Derpinghaus A, Aksel S, Cao M, Ladwig N, Cunha GR, Himelreich-Perić M, Baskin LS. Development of the human ovary: Fetal through pubertal ovarian morphology, folliculogenesis and expression of cellular differentiation markers. Differentiation 2023; 129:37-59. [PMID: 36347737 DOI: 10.1016/j.diff.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 01/28/2023]
Abstract
A definition of normal human fetal and early postnatal ovarian development is critical to the ability to accurately diagnose the presence or absence of functional ovarian tissue in clinical specimens. Through assembling an extensive histologic and immunohistochemical developmental ontogeny of human ovarian specimens from 8 weeks of gestation through 16 years of postnatal, we present a comprehensive immunohistochemical mapping of normal protein expression patterns in the early fetal through post-pubertal human ovary and detail a specific expression-based definition of the early stages of follicular development. Normal fetal and postnatal ovarian tissue is defined by the presence of follicular structures and characteristic immunohistochemical staining patterns, including granulosa cells expressing Forkhead Box Protein L2 (FOXL2). However, the current standard array of immunohistochemical markers poorly defines ovarian stromal tissue, and additional work is needed to identify new markers to advance our ability to accurately identify ovarian stromal components in gonadal specimens from patients with disorders of sexual differentiation.
Collapse
Affiliation(s)
- Maya R Overland
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Yi Li
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Amber Derpinghaus
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Sena Aksel
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Mei Cao
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Nicholas Ladwig
- Department of Pathology, University of California, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Gerald R Cunha
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | - Marta Himelreich-Perić
- Scientific Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Laurence S Baskin
- Department of Urology, University of California, 400 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
15
|
Gerhold AR, Labbé JC, Singh R. Uncoupling cell division and cytokinesis during germline development in metazoans. Front Cell Dev Biol 2022; 10:1001689. [PMID: 36407108 PMCID: PMC9669650 DOI: 10.3389/fcell.2022.1001689] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
The canonical eukaryotic cell cycle ends with cytokinesis, which physically divides the mother cell in two and allows the cycle to resume in the newly individualized daughter cells. However, during germline development in nearly all metazoans, dividing germ cells undergo incomplete cytokinesis and germ cells stay connected by intercellular bridges which allow the exchange of cytoplasm and organelles between cells. The near ubiquity of incomplete cytokinesis in animal germ lines suggests that this is an ancient feature that is fundamental for the development and function of this tissue. While cytokinesis has been studied for several decades, the mechanisms that enable regulated incomplete cytokinesis in germ cells are only beginning to emerge. Here we review the current knowledge on the regulation of germ cell intercellular bridge formation, focusing on findings made using mouse, Drosophila melanogaster and Caenorhabditis elegans as experimental systems.
Collapse
Affiliation(s)
- Abigail R. Gerhold
- Department of Biology, McGill University, Montréal, QC, Canada
- *Correspondence: Abigail R. Gerhold, ; Jean-Claude Labbé,
| | - Jean-Claude Labbé
- Institute for Research in Immunology and Cancer (IRIC), Montréal, QC, Canada
- Department of Pathology and Cell Biology, Université de Montréal, Succ. Centre-ville, Montréal, QC, Canada
- *Correspondence: Abigail R. Gerhold, ; Jean-Claude Labbé,
| | - Ramya Singh
- Department of Biology, McGill University, Montréal, QC, Canada
- Institute for Research in Immunology and Cancer (IRIC), Montréal, QC, Canada
| |
Collapse
|
16
|
Bogoch Y, Jamieson-Lucy A, Vejnar CE, Levy K, Giraldez AJ, Mullins MC, Elkouby YM. Stage Specific Transcriptomic Analysis and Database for Zebrafish Oogenesis. Front Cell Dev Biol 2022; 10:826892. [PMID: 35733854 PMCID: PMC9207522 DOI: 10.3389/fcell.2022.826892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/11/2022] [Indexed: 01/21/2023] Open
Abstract
Oogenesis produces functional eggs and is essential for fertility, embryonic development, and reproduction. The zebrafish ovary is an excellent model to study oogenesis in vertebrates, and recent studies have identified multiple regulators in oocyte development through forward genetic screens, as well as reverse genetics by CRISPR mutagenesis. However, many developmental steps in oogenesis, in zebrafish and other species, remain poorly understood, and their underlying mechanisms are unknown. Here, we take a genomic approach to systematically uncover biological activities throughout oogenesis. We performed transcriptomic analysis on five stages of oogenesis, from the onset of oocyte differentiation through Stage III, which precedes oocyte maturation. These transcriptomes revealed thousands of differentially expressed genes across stages of oogenesis. We analyzed trends of gene expression dynamics along oogenesis, as well as their expression in pair-wise comparisons between stages. We determined their functionally enriched terms, identifying uniquely characteristic biological activities in each stage. These data identified two prominent developmental phases in oocyte differentiation and traced the accumulation of maternally deposited embryonic regulator transcripts in the developing oocyte. Our analysis provides the first molecular description for oogenesis in zebrafish, which we deposit online as a resource for the community. Further, the presence of multiple gene paralogs in zebrafish, and the exclusive curation by many bioinformatic tools of the single paralogs present in humans, challenge zebrafish genomic analyses. We offer an approach for converting zebrafish gene name nomenclature to the human nomenclature for supporting genomic analyses generally in zebrafish. Altogether, our work provides a valuable resource as a first step to uncover oogenesis mechanisms and candidate regulators and track accumulating transcripts of maternal regulators of embryonic development.
Collapse
Affiliation(s)
- Yoel Bogoch
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Jerusalem, Israel
- Institute for Biomedical Research, Israel-Canada, Jerusalem, Israel
| | - Allison Jamieson-Lucy
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | | | - Karine Levy
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Jerusalem, Israel
- Institute for Biomedical Research, Israel-Canada, Jerusalem, Israel
| | | | - Mary C. Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Yaniv M. Elkouby
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Jerusalem, Israel
- Institute for Biomedical Research, Israel-Canada, Jerusalem, Israel
| |
Collapse
|
17
|
Day TC, Márquez-Zacarías P, Bravo P, Pokhrel AR, MacGillivray KA, Ratcliff WC, Yunker PJ. Varied solutions to multicellularity: The biophysical and evolutionary consequences of diverse intercellular bonds. BIOPHYSICS REVIEWS 2022; 3:021305. [PMID: 35673523 PMCID: PMC9164275 DOI: 10.1063/5.0080845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 04/29/2022] [Indexed: 11/16/2022]
Abstract
The diversity of multicellular organisms is, in large part, due to the fact that multicellularity has independently evolved many times. Nonetheless, multicellular organisms all share a universal biophysical trait: cells are attached to each other. All mechanisms of cellular attachment belong to one of two broad classes; intercellular bonds are either reformable or they are not. Both classes of multicellular assembly are common in nature, having independently evolved dozens of times. In this review, we detail these varied mechanisms as they exist in multicellular organisms. We also discuss the evolutionary implications of different intercellular attachment mechanisms on nascent multicellular organisms. The type of intercellular bond present during early steps in the transition to multicellularity constrains future evolutionary and biophysical dynamics for the lineage, affecting the origin of multicellular life cycles, cell-cell communication, cellular differentiation, and multicellular morphogenesis. The types of intercellular bonds used by multicellular organisms may thus result in some of the most impactful historical constraints on the evolution of multicellularity.
Collapse
Affiliation(s)
- Thomas C. Day
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | | | - Aawaz R. Pokhrel
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | - William C. Ratcliff
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Peter J. Yunker
- School of Physics, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
18
|
Yao M, Qu H, Han Y, Cheng CY, Xiao X. Kinesins in Mammalian Spermatogenesis and Germ Cell Transport. Front Cell Dev Biol 2022; 10:837542. [PMID: 35547823 PMCID: PMC9083010 DOI: 10.3389/fcell.2022.837542] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
In mammalian testes, the apical cytoplasm of each Sertoli cell holds up to several dozens of germ cells, especially spermatids that are transported up and down the seminiferous epithelium. The blood-testis barrier (BTB) established by neighboring Sertoli cells in the basal compartment restructures on a regular basis to allow preleptotene/leptotene spermatocytes to pass through. The timely transfer of germ cells and other cellular organelles such as residual bodies, phagosomes, and lysosomes across the epithelium to facilitate spermatogenesis is important and requires the microtubule-based cytoskeleton in Sertoli cells. Kinesins, a superfamily of the microtubule-dependent motor proteins, are abundantly and preferentially expressed in the testis, but their functions are poorly understood. This review summarizes recent findings on kinesins in mammalian spermatogenesis, highlighting their potential role in germ cell traversing through the BTB and the remodeling of Sertoli cell-spermatid junctions to advance spermatid transport. The possibility of kinesins acting as a mediator and/or synchronizer for cell cycle progression, germ cell transit, and junctional rearrangement and turnover is also discussed. We mostly cover findings in rodents, but we also make special remarks regarding humans. We anticipate that this information will provide a framework for future research in the field.
Collapse
Affiliation(s)
- Mingxia Yao
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Haoyang Qu
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - Yating Han
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - C Yan Cheng
- Department of Urology and Andrology, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang Xiao
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China.,Zhejiang Provincial Laboratory of Experimental Animal's & Nonclinical Laboratory Studies, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
19
|
Mytlis A, Kumar V, Qiu T, Deis R, Hart N, Levy K, Masek M, Shawahny A, Ahmad A, Eitan H, Nather F, Adar-Levor S, Birnbaum RY, Elia N, Bachmann-Gagescu R, Roy S, Elkouby YM. Control of meiotic chromosomal bouquet and germ cell morphogenesis by the zygotene cilium. Science 2022; 376:eabh3104. [PMID: 35549308 DOI: 10.1126/science.abh3104] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A hallmark of meiosis is chromosomal pairing, which requires telomere tethering and rotation on the nuclear envelope via microtubules, driving chromosome homology searches. Telomere pulling toward the centrosome forms the "zygotene chromosomal bouquet". Here, we identified the "zygotene cilium" in oocytes. This cilium provides a cable system for the bouquet machinery, extending throughout the germline cyst. Using zebrafish mutants and live manipulations, we demonstrate that the cilium anchors the centrosome to counterbalance telomere pulling. The cilium is essential for bouquet and synaptonemal complex formation, oogenesis, ovarian development, and fertility. Thus, a cilium represents a conserved player in zebrafish and mouse meiosis, which sheds light on reproductive aspects in ciliopathies, and suggests that cilia can control chromosomal dynamics.
Collapse
Affiliation(s)
- Avishag Mytlis
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Vineet Kumar
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Tao Qiu
- Institute of Molecular and Cell Biology, Proteos, 138673 Singapore
| | - Rachael Deis
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Neta Hart
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Karine Levy
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Markus Masek
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland.,Institute of Medical Genetics, University of Zurich, 8952 Schlieren, Switzerland
| | - Amal Shawahny
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Adam Ahmad
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Hagai Eitan
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Farouq Nather
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| | - Shai Adar-Levor
- Departments of Life Sciences, Ben-Gurion University of the Negev, Beer Shave 84105, Israel
| | - Ramon Y Birnbaum
- Departments of Life Sciences, Ben-Gurion University of the Negev, Beer Shave 84105, Israel
| | - Natalie Elia
- Departments of Life Sciences, Ben-Gurion University of the Negev, Beer Shave 84105, Israel
| | - Ruxandra Bachmann-Gagescu
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland.,Institute of Medical Genetics, University of Zurich, 8952 Schlieren, Switzerland
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Proteos, 138673 Singapore.,Department of Biological Sciences, National University of Singapore, 117543 Singapore.,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 119288 Singapore
| | - Yaniv M Elkouby
- Department of Developmental Biology and Cancer Research, Hebrew University of Jerusalem Faculty of Medicine, Ein-Kerem Campus, Jerusalem 9112102, Israel.,Institute for Medical Research-Israel-Canada (IMRIC), Ein-Kerem Campus, Jerusalem 9112102, Israel
| |
Collapse
|
20
|
Incomplete abscission and cytoplasmic bridges in the evolution of eukaryotic multicellularity. Curr Biol 2022; 32:R385-R397. [DOI: 10.1016/j.cub.2022.03.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
21
|
Dunleavy JEM, O'Connor AE, Okuda H, Merriner DJ, O'Bryan MK. KATNB1 is a master regulator of multiple katanin enzymes in male meiosis and haploid germ cell development. Development 2021; 148:273717. [PMID: 34822718 DOI: 10.1242/dev.199922] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Katanin microtubule-severing enzymes are crucial executers of microtubule regulation. Here, we have created an allelic loss-of-function series of the katanin regulatory B-subunit KATNB1 in mice. We reveal that KATNB1 is the master regulator of all katanin enzymatic A-subunits during mammalian spermatogenesis, wherein it is required to maintain katanin A-subunit abundance. Our data shows that complete loss of KATNB1 from germ cells is incompatible with sperm production, and we reveal multiple new spermatogenesis functions for KATNB1, including essential roles in male meiosis, acrosome formation, sperm tail assembly, regulation of both the Sertoli and germ cell cytoskeletons during sperm nuclear remodelling, and maintenance of seminiferous epithelium integrity. Collectively, our findings reveal that katanins are able to differentially regulate almost all key microtubule-based structures during mammalian male germ cell development, through the complexing of one master controller, KATNB1, with a 'toolbox' of neofunctionalised katanin A-subunits.
Collapse
Affiliation(s)
- Jessica E M Dunleavy
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Anne E O'Connor
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Hidenobu Okuda
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia
| | - D Jo Merriner
- School of Biological Sciences, Faculty of Science, Monash University, Clayton, VIC, 3800, Australia.,School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Moira K O'Bryan
- School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
22
|
p53 inactivation unmasks histone methylation-independent WDR5 functions that drive self-renewal and differentiation of pluripotent stem cells. Stem Cell Reports 2021; 16:2642-2658. [PMID: 34715053 PMCID: PMC8581203 DOI: 10.1016/j.stemcr.2021.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 11/23/2022] Open
Abstract
p53 alterations occur during culture of pluripotent stem cells (PSCs), but the significance of these events on epigenetic control of PSC fate determination remains poorly understood. Wdr5 deletion in p53-null (DKO) mouse ESCs (mESCs) leads to impaired self-renewal, defective retinal neuroectoderm differentiation, and de-repression of germ cell/meiosis (GCM)-specific genes. Re-introduction of a WDR5 mutant with defective H3K4 methylation activity into DKO ESCs restored self-renewal and suppressed GCM gene expression but failed to induce retinal neuroectoderm differentiation. Mechanistically, mutant WDR5 targets chromatin that is largely devoid of H3K4me3 and regulates gene expression in p53-null mESCs. Furthermore, MAX and WDR5 co-target lineage-specifying chromatin and regulate chromatin accessibility of GCM-related genes. Importantly, MAX and WDR5 are core subunits of a non-canonical polycomb repressor complex 1 responsible for gene silencing. This function, together with canonical, pro-transcriptional WDR5-dependent MLL complex H3K4 methyltransferase activity, highlight how WDR5 mediates crosstalk between transcription and repression during mESC fate choice. H3K4me defective WDR5 supports self-renewal and GCM differentiation in p53-null mESCs WDR5 regulates H3K4me-independent stemness and GCM gene expression in p53-null mESCs MAX and WDR5 repress GCM-related gene chromatin accessibility upon differentiation
Collapse
|
23
|
Bauer J, Poupart V, Goupil E, Nguyen KCQ, Hall DH, Labbé JC. The initial expansion of the C. elegans syncytial germ line is coupled to incomplete primordial germ cell cytokinesis. Development 2021; 148:dev199633. [PMID: 34195824 PMCID: PMC8327289 DOI: 10.1242/dev.199633] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/25/2021] [Indexed: 01/06/2023]
Abstract
The C. elegans germline is organized as a syncytium in which each germ cell possesses an intercellular bridge that is maintained by a stable actomyosin ring and connected to a common pool of cytoplasm, termed the rachis. How germ cells undergo cytokinesis while maintaining this syncytial architecture is not completely understood. Here, we use live imaging to characterize primordial germ cell (PGC) division in C. elegans first-stage larvae. We show that each PGC possesses a stable intercellular bridge that connects it to a common pool of cytoplasm, which we term the proto-rachis. We further show that the first PGC cytokinesis is incomplete and that the stabilized cytokinetic ring progressively moves towards the proto-rachis and eventually integrates with it. Our results support a model in which the initial expansion of the C. elegans syncytial germline occurs by incomplete cytokinesis, where one daughter germ cell inherits the actomyosin ring that was newly formed by stabilization of the cytokinetic ring, while the other inherits the pre-existing stable actomyosin ring. We propose that such a mechanism of iterative cytokinesis incompletion underpins C. elegans germline expansion and maintenance.
Collapse
Affiliation(s)
- Jack Bauer
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Vincent Poupart
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Eugénie Goupil
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Ken C. Q. Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David H. Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jean-Claude Labbé
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, C.P. 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
- Department of Pathology and Cell Biology, Université de Montréal, C.P. 6128, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
24
|
Iwamori T, Iwamori N, Matsumoto M, Imai H, Ono E. Novel localizations and interactions of intercellular bridge proteins revealed by proteomic profiling†. Biol Reprod 2021; 102:1134-1144. [PMID: 31995159 DOI: 10.1093/biolre/ioaa017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/17/2019] [Accepted: 01/28/2020] [Indexed: 11/14/2022] Open
Abstract
Intercellular bridges (ICBs) connecting germ cells are essential for spermatogenesis, and their deletion causes male infertility. However, the functions and component factors of ICBs are still unknown. We previously identified novel ICB-associated proteins by proteomics analysis using ICB enrichment. Here, we performed immunoprecipitation-proteomics analyses using antibodies specific to known ICB proteins MKLP1, RBM44, and ectoplasmic specialization-associated protein KIAA1210 and predicted protein complexes in the ICB cores. KIAA1210, its binding protein topoisomerase2B (TOP2B), and tight junction protein ZO1 were identified as novel ICB proteins. On the other hand, as well as KIAA1210 and TOP2B, MKLP1 and RBM44, but not TEX14, were localized at the XY body of spermatocytes, suggesting that there is a relationship between ICB proteins and meiotic chromosomes. Moreover, small RNAs interacted with an ICB protein complex that included KIAA1210, RBM44, and MKLP1. These results indicate dynamic movements of ICB proteins and suggest that ICB proteins could be involved not only in the communication between germ cells but also in their epigenetic regulation. Our results provide a novel perspective on the function of ICBs and could be helpful in revealing the biological function of the ICB.
Collapse
Affiliation(s)
- Tokuko Iwamori
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Iwamori
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan and
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Imai
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Etsuro Ono
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
25
|
Soygur B, Laird DJ. Ovary Development: Insights From a Three-Dimensional Imaging Revolution. Front Cell Dev Biol 2021; 9:698315. [PMID: 34381780 PMCID: PMC8351467 DOI: 10.3389/fcell.2021.698315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022] Open
Abstract
The ovary is an indispensable unit of female reproduction and health. However, the study of ovarian function in mammals is hindered by unique challenges, which include the desynchronized development of oocytes, irregular distribution and vast size discrepancy of follicles, and dynamic tissue remodeling during each hormonal cycle. Overcoming the limitations of traditional histology, recent advances in optical tissue clearing and three-dimensional (3D) visualization offer an advanced platform to explore the architecture of intact organs at a single cell level and reveal new relationships and levels of organization. Here we summarize the development and function of ovarian compartments that have been delineated by conventional two-dimensional (2D) methods and the limits of what can be learned by these approaches. We compare types of optical tissue clearing, 3D analysis technologies, and their application to the mammalian ovary. We discuss how 3D modeling of the ovary has extended our knowledge and propose future directions to unravel ovarian structure toward therapeutic applications for ovarian disease and extending female reproductive lifespan.
Collapse
Affiliation(s)
| | - Diana J. Laird
- Department of Obstetrics, Gynecology & Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
26
|
Ikami K, Nuzhat N, Abbott H, Pandoy R, Haky L, Spradling AC, Tanner H, Lei L. Altered germline cyst formation and oogenesis in Tex14 mutant mice. Biol Open 2021; 10:269245. [PMID: 34156079 PMCID: PMC8249907 DOI: 10.1242/bio.058807] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/24/2022] Open
Abstract
During oocyte differentiation in mouse fetal ovaries, sister germ cells are connected by intercellular bridges, forming germline cysts. Within the cyst, primary oocytes form via gaining cytoplasm and organelles from sister germ cells through germ cell connectivity. To uncover the role of intercellular bridges in oocyte differentiation, we analyzed mutant female mice lacking testis-expressed 14 (TEX14), a protein involved in intercellular bridge formation and stabilization. In Tex14 homozygous mutant fetal ovaries, germ cells divide to form a reduced number of cysts in which germ cells remained connected via syncytia or fragmented cell membranes, rather than normal intercellular bridges. Compared with wild-type cysts, homozygous mutant cysts fragmented at a higher frequency and produced a greatly reduced number of primary oocytes with precocious cytoplasmic enrichment and enlarged volume. By contrast, Tex14 heterozygous mutant germline cysts were less fragmented and generate primary oocytes at a reduced size. Moreover, enlarged primary oocytes in homozygous mutants were used more efficiently to sustain folliculogenesis than undersized heterozygous mutant primary oocytes. Our observations directly link the nature of fetal germline cysts to oocyte differentiation and development. Summary: Altered germline cyst formation and fragmentation due to defective germ cell connectivity leads to changes in oocyte differentiation and development in Tex14 mutant mice.
Collapse
Affiliation(s)
- Kanako Ikami
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Nafisa Nuzhat
- Department of Cell and Developmental Biology, University of Michigan Medical School, 48109, Ann Arbor, MI, USA
| | - Haley Abbott
- Department of Cell and Developmental Biology, University of Michigan Medical School, 48109, Ann Arbor, MI, USA
| | - Ronald Pandoy
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Lauren Haky
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Allan C Spradling
- Department of Embryology, Carnegie Institution for Science, 21218, Baltimore, MD, USA
| | - Heather Tanner
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Lei Lei
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| |
Collapse
|
27
|
Mytlis A, Elkouby YM. Live and Time-Lapse Imaging of Early Oogenesis and Meiotic Chromosomal Dynamics in Cultured Juvenile Zebrafish Ovaries. Methods Mol Biol 2021; 2218:137-155. [PMID: 33606229 DOI: 10.1007/978-1-0716-0970-5_12] [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] [Indexed: 02/21/2023]
Abstract
Oocyte production is crucial for sexual reproduction. Recent findings in zebrafish and other established model organisms emphasize that the early steps of oogenesis involve the coordination of simultaneous and tightly sequential processes across cellular compartments and between sister cells. To fully understand the mechanistic framework of these coordinated processes, cellular and morphological analysis in high temporal resolution is required. Here, we provide a protocol for four-dimensional live time-lapse analysis of cultured juvenile zebrafish ovaries. We describe how multiple-stage oocytes can be simultaneously analyzed in single ovaries, and several ovaries can be processed in single experiments. In addition, we detail adequate conditions for quantitative image acquisition. Finally, we demonstrate that using this protocol, we successfully capture rapid meiotic chromosomal movements in early prophase for the first time in zebrafish oocytes, in four dimensions and in vivo. Our protocol expands the use of the zebrafish as a model system to understand germ cell and ovarian development in postembryonic stages.
Collapse
Affiliation(s)
- Avishag Mytlis
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem, Faculty of Medicine, Institute for Medical Research - Israel-Canada (IMRIC), Jerusalem, Israel
| | - Yaniv M Elkouby
- Department of Developmental Biology and Cancer Research, The Hebrew University of Jerusalem, Faculty of Medicine, Institute for Medical Research - Israel-Canada (IMRIC), Jerusalem, Israel.
| |
Collapse
|
28
|
Gerdes JA, Mannix KM, Hudson AM, Cooley L. HtsRC-Mediated Accumulation of F-Actin Regulates Ring Canal Size During Drosophila melanogaster Oogenesis. Genetics 2020; 216:717-734. [PMID: 32883702 PMCID: PMC7648574 DOI: 10.1534/genetics.120.303629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/30/2020] [Indexed: 12/21/2022] Open
Abstract
Ring canals in the female germline of Drosophila melanogaster are supported by a robust filamentous actin (F-actin) cytoskeleton, setting them apart from ring canals in other species and tissues. Previous work has identified components required for the expansion of the ring canal actin cytoskeleton, but has not identified the proteins responsible for F-actin recruitment or accumulation. Using a combination of CRISPR-Cas9 mediated mutagenesis and UAS-Gal4 overexpression, we show that HtsRC-a component specific to female germline ring canals-is both necessary and sufficient to drive F-actin accumulation. Absence of HtsRC in the germline resulted in ring canals lacking inner rim F-actin, while overexpression of HtsRC led to larger ring canals. HtsRC functions in combination with Filamin to recruit F-actin to ectopic actin structures in somatic follicle cells. Finally, we present findings that indicate that HtsRC expression and robust female germline ring canal expansion are important for high fecundity in fruit flies but dispensable for their fertility-a result that is consistent with our understanding of HtsRC as a newly evolved gene specific to female germline ring canals.
Collapse
Affiliation(s)
- Julianne A Gerdes
- Department of Genetics, Yale University School of Medicine, New Haven, 06520 Connecticut
| | - Katelynn M Mannix
- Department of Genetics, Yale University School of Medicine, New Haven, 06520 Connecticut
| | - Andrew M Hudson
- Department of Genetics, Yale University School of Medicine, New Haven, 06520 Connecticut
| | - Lynn Cooley
- Department of Genetics, Yale University School of Medicine, New Haven, 06520 Connecticut
- Department of Cell Biology, Yale University School of Medicine, New Haven, 06520 Connecticut
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511 Connecticut
| |
Collapse
|
29
|
Umehara T, Tsujita N, Zhu Z, Ikedo M, Shimada M. A simple sperm-sexing method that activates TLR7/8 on X sperm for the efficient production of sexed mouse or cattle embryos. Nat Protoc 2020; 15:2645-2667. [DOI: 10.1038/s41596-020-0348-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/30/2020] [Indexed: 12/13/2022]
|
30
|
Bai X, Melesse M, Sorensen Turpin CG, Sloan DE, Chen CY, Wang WC, Lee PY, Simmons JR, Nebenfuehr B, Mitchell D, Klebanow LR, Mattson N, Betzig E, Chen BC, Cheerambathur D, Bembenek JN. Aurora B functions at the apical surface after specialized cytokinesis during morphogenesis in C. elegans. Development 2020; 147:dev.181099. [PMID: 31806662 PMCID: PMC6983721 DOI: 10.1242/dev.181099] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022]
Abstract
Although cytokinesis has been intensely studied, the way it is executed during development is not well understood, despite a long-standing appreciation that various aspects of cytokinesis vary across cell and tissue types. To address this, we investigated cytokinesis during the invariant Caenorhabditis elegans embryonic divisions and found several parameters that are altered at different stages in a reproducible manner. During early divisions, furrow ingression asymmetry and midbody inheritance is consistent, suggesting specific regulation of these events. During morphogenesis, we found several unexpected alterations to cytokinesis, including apical midbody migration in polarizing epithelial cells of the gut, pharynx and sensory neurons. Aurora B kinase, which is essential for several aspects of cytokinesis, remains apically localized in each of these tissues after internalization of midbody ring components. Aurora B inactivation disrupts cytokinesis and causes defects in apical structures, even if inactivated post-mitotically. Therefore, we demonstrate that cytokinesis is implemented in a specialized way during epithelial polarization and that Aurora B has a role in the formation of the apical surface.
Collapse
Affiliation(s)
- Xiaofei Bai
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Michael Melesse
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | | | - Dillon E. Sloan
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA,Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chin-Yi Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Cheng Wang
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Po-Yi Lee
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - James R. Simmons
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Benjamin Nebenfuehr
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Diana Mitchell
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindsey R. Klebanow
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Nicholas Mattson
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA
| | - Eric Betzig
- Janelia Research Campus, HHMI, Ashburn, VA 20147, USA
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan,Janelia Research Campus, HHMI, Ashburn, VA 20147, USA
| | - Dhanya Cheerambathur
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Joshua N. Bembenek
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996, USA,Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA,Author for correspondence ()
| |
Collapse
|
31
|
Mannix KM, Starble RM, Kaufman RS, Cooley L. Proximity labeling reveals novel interactomes in live Drosophila tissue. Development 2019; 146:dev.176644. [PMID: 31208963 DOI: 10.1242/dev.176644] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022]
Abstract
Gametogenesis is dependent on intercellular communication facilitated by stable intercellular bridges connecting developing germ cells. During Drosophila oogenesis, intercellular bridges (referred to as ring canals; RCs) have a dynamic actin cytoskeleton that drives their expansion to a diameter of 10 μm. Although multiple proteins have been identified as components of RCs, we lack a basic understanding of how RC proteins interact together to form and regulate the RC cytoskeleton. Thus, here, we optimized a procedure for proximity-dependent biotinylation in live tissue using the APEX enzyme to interrogate the RC interactome. APEX was fused to four different RC components (RC-APEX baits) and 55 unique high-confidence prey were identified. The RC-APEX baits produced almost entirely distinct interactomes that included both known RC proteins and uncharacterized proteins. A proximity ligation assay was used to validate close-proximity interactions between the RC-APEX baits and their respective prey. Furthermore, an RNA interference screen revealed functional roles for several high-confidence prey genes in RC biology. These findings highlight the utility of enzyme-catalyzed proximity labeling for protein interactome analysis in live tissue and expand our understanding of RC biology.
Collapse
Affiliation(s)
- Katelynn M Mannix
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Rebecca M Starble
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Ronit S Kaufman
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lynn Cooley
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA .,Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
32
|
Huang G, Kaufman AJ, Ryan RJH, Romin Y, Huryn L, Bains S, Manova-Todorova K, Morris PL, Hunnicutt GR, Adelman CA, Petrini JHJ, Ramanathan Y, Singh B. Mouse DCUN1D1 (SCCRO) is required for spermatogenetic individualization. PLoS One 2019; 14:e0209995. [PMID: 30653527 PMCID: PMC6336273 DOI: 10.1371/journal.pone.0209995] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/15/2018] [Indexed: 11/19/2022] Open
Abstract
Squamous cell carcinoma–related oncogene (SCCRO, also known as DCUN1D1) is a component of the E3 for neddylation. As such, DCUN1D1 regulates the neddylation of cullin family members. Targeted inactivation of DCUN1D1 in mice results in male-specific infertility. Infertility in DCUN1D1-/- mice is secondary to primary defects in spermatogenesis. Time-dam experiments mapped the onset of the defect in spermatogenesis to 5.5 to 6 weeks of age, which temporally corresponds to defects in spermiogenesis. Although the first round of spermatogenesis progressed normally, the number of spermatozoa released into the seminiferous lumen and epididymis of DCUN1D1-/- mice was significantly reduced. Spermatozoa in DCUN1D1-/- mice had multiple abnormalities, including globozoospermia, macrocephaly, and multiple flagella. Many of the malformed spermatozoa in DCUN1D1-/- mice were multinucleated, with supernumerary and malpositioned centrioles, suggesting a defect in the resolution of intercellular bridges. The onset of the defect in spermatogenesis in DCUN1D1-/- mice corresponds to an increase in DCUN1D1 expression observed during normal spermatogenesis. Moreover, consistent with its known function as a component of the E3 in neddylation, the pattern of DCUN1D1 expression temporally correlates with an increase in the neddylated cullin fraction and stage-specific increases in the total ubiquitinated protein pool in wild-type mice. Levels of neddylated Cul3 were decreased in DCUN1D1-/- mice, and ubiquitinated proteins did not accumulate during the stages in which DCUN1D1 expression peaks during spermatogenesis in wild-type mice. Combined, these findings suggest that DCUN1D1-/- mice fail to release mature spermatozoa into the seminiferous lumen, possibly due to unresolved intercellular bridges. Furthermore, the effects of DCUN1D1 on spermatogenesis likely involve its regulation of cullin-RING-ligase (CRL)–type ubiquitin E3 activity during spermiogenesis through its role in promoting Cul3 neddylation. The specific CRLs required for spermiogenesis and their protein targets require identification.
Collapse
Affiliation(s)
- Guochang Huang
- Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Andrew J. Kaufman
- Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Russell J. H. Ryan
- Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Yevgeniy Romin
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Laryssa Huryn
- Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Sarina Bains
- Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Katia Manova-Todorova
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Patricia L. Morris
- Population Council and The Rockefeller University, New York, New York, United States of America
| | - Gary R. Hunnicutt
- Population Council and The Rockefeller University, New York, New York, United States of America
| | - Carrie A. Adelman
- Department of Molecular Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - John H. J. Petrini
- Department of Molecular Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Y. Ramanathan
- Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Bhuvanesh Singh
- Department of Surgery, Laboratory of Epithelial Cancer Biology, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
33
|
Jamieson-Lucy A, Mullins MC. The vertebrate Balbiani body, germ plasm, and oocyte polarity. Curr Top Dev Biol 2019; 135:1-34. [DOI: 10.1016/bs.ctdb.2019.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
34
|
Yoshida S. Heterogeneous, dynamic, and stochastic nature of mammalian spermatogenic stem cells. Curr Top Dev Biol 2019; 135:245-285. [DOI: 10.1016/bs.ctdb.2019.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
35
|
Lee KY, Green RA, Gutierrez E, Gomez-Cavazos JS, Kolotuev I, Wang S, Desai A, Groisman A, Oegema K. CYK-4 functions independently of its centralspindlin partner ZEN-4 to cellularize oocytes in germline syncytia. eLife 2018; 7:36919. [PMID: 29989548 PMCID: PMC6056237 DOI: 10.7554/elife.36919] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022] Open
Abstract
Throughout metazoans, germ cells undergo incomplete cytokinesis to form syncytia connected by intercellular bridges. Gamete formation ultimately requires bridge closure, yet how bridges are reactivated to close is not known. The most conserved bridge component is centralspindlin, a complex of the Rho family GTPase-activating protein (GAP) CYK-4/MgcRacGAP and the microtubule motor ZEN-4/kinesin-6. Here, we show that oocyte production by the syncytial Caenorhabditis elegans germline requires CYK-4 but not ZEN-4, which contrasts with cytokinesis, where both are essential. Longitudinal imaging after conditional inactivation revealed that CYK-4 activity is important for oocyte cellularization, but not for the cytokinesis-like events that generate syncytial compartments. CYK-4’s lipid-binding C1 domain and the GTPase-binding interface of its GAP domain were both required to target CYK-4 to intercellular bridges and to cellularize oocytes. These results suggest that the conserved C1-GAP region of CYK-4 constitutes a targeting module required for closure of intercellular bridges in germline syncytia.
Collapse
Affiliation(s)
- Kian-Yong Lee
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Rebecca A Green
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Edgar Gutierrez
- Department of Physics, University of California, San Diego, La Jolla, United States
| | - J Sebastian Gomez-Cavazos
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Irina Kolotuev
- Microscopy Rennes Imaging Center and Biosit, University of Rennes 1, Rennes, France
| | - Shaohe Wang
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Arshad Desai
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| | - Alex Groisman
- Department of Physics, University of California, San Diego, La Jolla, United States
| | - Karen Oegema
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, United States
| |
Collapse
|
36
|
Iwamori T, Iwamori N, Matsumoto M, Ono E, Matzuk MM. Identification of KIAA1210 as a novel X-chromosome-linked protein that localizes to the acrosome and associates with the ectoplasmic specialization in testes. Biol Reprod 2018; 96:469-477. [PMID: 28203736 DOI: 10.1095/biolreprod.116.145458] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/21/2016] [Accepted: 12/15/2016] [Indexed: 11/01/2022] Open
Abstract
Cell junctions are necessary for spermatogenesis, and there are numerous types of junctions in testis, such as blood–testis barrier, intercellular bridge, and ectoplasmic specialization (ES). The details of their functions and construction are still unknown. To identify a novel protein essential to the function of a cell junction, we enriched testis membrane protein and analyzed it using a proteomics approach. Here, we report a novel ES protein, which is encoded on the X chromosome and an ortholog of hypothetical human protein KIAA1210. KIAA1210 is expressed in testis predominantly, localized to the sex body in spermatocyte, acrosome, and near ES. Moreover, KIAA1210 possesses a topoisomerase 2 (TOP2)-associated protein PAT1 domain, a herpes simplex virus 1 (HSV-1) large tegument protein UL36 hypothetical domain, and a provisional DNA translocase FtsK domain. Using IP-proteomics with specific antibody to KIAA1210, we identified proteins including TOP2 isoforms as components of a complex with KIAA1210, in cell junctions in testis. The interaction between KIAA1210 and TOP2 was confirmed by two different proteomic analyses. Furthermore, immunofluorescence showed that KIAA1210 and TOP2B co-localize around the sex body in spermatocyte, apical ES, and residual bodies in elongated spermatids. Our findings suggest that KIAA1210 may be essential cell junction protein that interacts with TOP2B to regulate the dynamic change of chromatin structures during spermiogenesis.
Collapse
Affiliation(s)
- Tokuko Iwamori
- Department of Biomedicine, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Iwamori
- Department of Biomedicine, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Matsumoto
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Etsuro Ono
- Department of Biomedicine, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Martin M Matzuk
- Departments of Pathology and Immunology, Molecular and Cellular Biology, Molecular and Human Genetics, and Pharmacology, Center for Drug Discovery, and Center for Reproductive Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
37
|
Abstract
Male infertility is a major and growing problem and, in most cases, the specific root cause is unknown. Here we show that the transcription factor SOX30 plays a critical role in mouse spermatogenesis. Sox30-null mice are healthy and females are fertile, but males are sterile. In the absence of Sox30 meiosis initiates normally in both sexes but, in males, germ cell development arrests during the post-meiotic round spermatid period. In the mutant testis, acrosome and axoneme development are aberrant, multinucleated germ cells (symplasts) form and round spermatids unable to process beyond step 3 of spermiogenesis. No elongated spermatids nor spermatozoa are produced. Thus, Sox30 represents a rare example of a gene for which loss of function results in a complete arrest of spermatogenesis at the onset of spermiogenesis. Our results suggest that SOX30 mutations may underlie some instances of unexplained non-obstructive azoospermia in humans.
Collapse
|
38
|
Goupil E, Amini R, Hall DH, Labbé JC. Actomyosin contractility regulators stabilize the cytoplasmic bridge between the two primordial germ cells during Caenorhabditis elegans embryogenesis. Mol Biol Cell 2017; 28:3789-3800. [PMID: 29074566 PMCID: PMC5739295 DOI: 10.1091/mbc.e17-08-0502] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 01/08/2023] Open
Abstract
The Caenorhabditis elegans germline is syncytial but its formation is not completely understood. During embryogenesis, the germline precursor blastomere does not complete cytokinesis and maintains a stable cytoplasmic bridge between the two primordial germ cells that is enriched in actomyosin contractility regulators. Stable cytoplasmic bridges arise from failed cytokinesis, the last step of cell division, and are a key feature of syncytial architectures in the germline of most metazoans. Whereas the Caenorhabditis elegans germline is syncytial, its formation remains poorly understood. We found that the germline precursor blastomere, P4, fails cytokinesis, leaving a stable cytoplasmic bridge between the two daughter cells, Z2 and Z3. Depletion of several regulators of actomyosin contractility resulted in a regression of the membrane partition between Z2 and Z3, indicating that they are required to stabilize the cytoplasmic bridge. Epistatic analysis revealed a pathway in which Rho regulators promote accumulation of the noncannonical anillin ANI-2 at the stable cytoplasmic bridge, which in turns promotes the accumulation of the nonmuscle myosin II NMY-2 and the midbody component CYK-7 at the bridge, in part by limiting the accumulation of canonical anillin ANI-1. Our results uncover key steps in C. elegans germline formation and define a set of conserved regulators that are enriched at the primordial germ cell cytoplasmic bridge to ensure its stability during embryonic development.
Collapse
Affiliation(s)
| | - Rana Amini
- Institute of Research in Immunology and Cancer and
| | - David H Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jean-Claude Labbé
- Institute of Research in Immunology and Cancer and .,Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
39
|
Lu K, Jensen L, Lei L, Yamashita YM. Stay Connected: A Germ Cell Strategy. Trends Genet 2017; 33:971-978. [PMID: 28947158 DOI: 10.1016/j.tig.2017.09.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/24/2017] [Accepted: 09/05/2017] [Indexed: 01/20/2023]
Abstract
Germ cells develop as a cyst of interconnected sibling cells in a broad range of organisms in both sexes. A well-established function of intercellular connectivity is to transport cytoplasmic materials from 'nurse' cells to oocytes, a critical process for developing functional oocytes in ovaries of many species. However, there are situations where connectivity exists without a nursing mechanism, and the biological meaning of such connectivity remains obscure. In this review, we summarize current knowledge on the formation of intercellular connectivity, and discuss its meaning by visiting multiple examples of germ cell connectivity observed in evolutionarily distant species.
Collapse
Affiliation(s)
- Kevin Lu
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindy Jensen
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lei Lei
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yukiko M Yamashita
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA; Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
40
|
Katsumata O, Mori M, Sawane Y, Niimura T, Ito A, Okamoto H, Fukaya M, Sakagami H. Cellular and subcellular localization of ADP-ribosylation factor 6 in mouse peripheral tissues. Histochem Cell Biol 2017; 148:577-596. [PMID: 28748255 DOI: 10.1007/s00418-017-1599-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2017] [Indexed: 01/30/2023]
Abstract
ADP-ribosylation factor 6 (Arf6) is a small GTPase that regulates endosomal trafficking and actin cytoskeleton remodeling. In the present study, we comprehensively examined the cellular and subcellular localization of Arf6 in adult mouse peripheral tissues by immunofluorescence and immunoelectron microscopy using the heat-induced antigen retrieval method with Tris-EDTA buffer (pH 9.0). Marked immunolabeling of Arf6 was observed particularly in epithelial cells of several tissues including the esophagus, stomach, small and large intestines, trachea, kidney, epididymis, oviduct, and uterus. In most epithelial cells of simple or pseudostratified epithelia, Arf6 exhibited predominant localization to the basolateral membrane and a subpopulation of endosomes. At an electron microscopic level, Arf6 was localized along the basolateral membrane, with dense accumulation at interdigitating processes and infoldings. Arf6 was present in a ring-like appearance at intercellular bridges in spermatogonia and spermatocytes in the testis and at the Flemming body of cytokinetic somatic cells in the ovarian follicle, thymus, and spleen. The present study provides anatomical clues to help understand the physiological roles of Arf6 at the whole animal level.
Collapse
Affiliation(s)
- Osamu Katsumata
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Momoko Mori
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yusuke Sawane
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Tomoko Niimura
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Akiko Ito
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.,Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hirotsugu Okamoto
- Department of Anesthesiology, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Masahiro Fukaya
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hiroyuki Sakagami
- Department of Anatomy, Kitasato University School of Medicine, Sagamihara, Kanagawa, 252-0374, Japan.
| |
Collapse
|
41
|
Elkouby YM, Mullins MC. Coordination of cellular differentiation, polarity, mitosis and meiosis - New findings from early vertebrate oogenesis. Dev Biol 2017; 430:275-287. [PMID: 28666956 DOI: 10.1016/j.ydbio.2017.06.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 12/21/2022]
Abstract
A mechanistic dissection of early oocyte differentiation in vertebrates is key to advancing our knowledge of germline development, reproductive biology, the regulation of meiosis, and all of their associated disorders. Recent advances in the field include breakthroughs in the identification of germline stem cells in Medaka, in the cellular architecture of the germline cyst in mice, in a mechanistic dissection of chromosomal pairing and bouquet formation in meiosis in mice, in tracing oocyte symmetry breaking to the chromosomal bouquet of meiosis in zebrafish, and in the biology of the Balbiani body, a universal oocyte granule. Many of the major events in early oogenesis are universally conserved, and some are co-opted for species-specific needs. The chromosomal events of meiosis are of tremendous consequence to gamete formation and have been extensively studied. New light is now being shed on other aspects of early oocyte differentiation, which were traditionally considered outside the scope of meiosis, and their coordination with meiotic events. The emerging theme is of meiosis as a common groundwork for coordinating multifaceted processes of oocyte differentiation. In an accompanying manuscript we describe methods that allowed for investigations in the zebrafish ovary to contribute to these breakthroughs. Here, we review these advances mostly from the zebrafish and mouse. We discuss oogenesis concepts across established model organisms, and construct an inclusive paradigm for early oocyte differentiation in vertebrates.
Collapse
Affiliation(s)
- Yaniv M Elkouby
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
42
|
Ikami K, Nuzhat N, Lei L. Organelle transport during mouse oocyte differentiation in germline cysts. Curr Opin Cell Biol 2017; 44:14-19. [PMID: 28038435 DOI: 10.1016/j.ceb.2016.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/25/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
Abstract
During mammalian oogenesis, germ cells undergo oocyte differentiation and oocyte development to form mature oocytes that contain essential components for supporting early embryogenesis. However, only a small fraction of germ cells become mature oocytes and the mechanism of this massive germ cell loss has been unclear. Our recent studies suggested that the formation of functional oocytes and germ cell loss are interlinked by a 'nursing' process in germline cysts during oocyte differentiation in mouse fetal ovaries. 80% of the fetal germ cells sacrifice themselves by donating their cytoplasmic contents to the remaining sister germ cells that differentiate into primary oocytes with augmented developmental potential. In this review, we will summarize the process of mouse oocyte differentiation with a particular focus on organelle transport in germline cysts.
Collapse
Affiliation(s)
- Kanako Ikami
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States
| | - Nafisa Nuzhat
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States
| | - Lei Lei
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States.
| |
Collapse
|
43
|
Kim HJ, Kim H, Chang R, Yu YG, Lee HH. Biochemical and Molecular Modeling Studies of the Interaction between Human CEP55 and TEX14. B KOREAN CHEM SOC 2016. [DOI: 10.1002/bkcs.10785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hee Jung Kim
- Department of Bio & Nano Chemistry; Kookmin University; Seoul 02707 Korea
| | - Hyunook Kim
- Department of Chemistry; Kwangwoon University; Seoul 139-701 Korea
| | - Rakwoo Chang
- Department of Chemistry; Kwangwoon University; Seoul 139-701 Korea
| | - Yeon Gyu Yu
- Department of Bio & Nano Chemistry; Kookmin University; Seoul 02707 Korea
| | - Hyung Ho Lee
- Department of Chemistry, College of Natural Sciences; Seoul National University; Seoul 151-742 Korea
| |
Collapse
|
44
|
Gadella BM, Ferraz MA. A Review of New Technologies that may Become Useful for in vitro Production of Boar Sperm. Reprod Domest Anim 2016; 50 Suppl 2:61-70. [PMID: 26174921 DOI: 10.1111/rda.12571] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/20/2015] [Indexed: 01/07/2023]
Abstract
Making sperm cells outside the original testicular environment in a culture dish has been considered for a long time as impossible due to the very complicated process of spermatogenesis and sperm maturation, which altogether, encompasses a 2-month period. However, new approaches in complex three-dimensional co-cell cultures, micro-perfusion and micro-fluidics technologies, new knowledge in the functioning, culturing and differentiation of spermatogonial stem cells (SSC) and their precursor cells have revolutionized this field. Furthermore, the use of better molecular markers as well as stimulatory factors has led to successful in vitro culture of stem cells either derived from germ line stem cells, from induced pluripotent stem cells (iPSC) or from embryonic stem cells (ESC). These stem cells when placed into small seminiferous tubule fragments are able to become SSC. The SSC beyond self-renewal can also be induced into haploid sperm-like cells under in vitro conditions. In mouse, this in vitro produced sperm can be injected into a mature oocyte and allow post-fertilization development into an early embryo in vitro. After transferring such obtained embryos into the uterus of a recipient mouse, they can further develop into healthy offspring. Recently, a similar approach has been performed with combining selected cells from testicular cell suspensions followed by a complete in vitro culture of seminiferous cords producing sperm-like cells. However, most of the techniques developed are laborious, time-consuming and have low efficiency, placing questionable that it will become useful used for setting up an efficient in vitro sperm production system for the boar. The benefits and drawbacks as well as the likeliness of in vitro pig sperm production to become applied in assisted technologies for swine reproduction are critically discussed. In this contribution, also the process of sperm production in the testis and sperm maturation is reviewed.
Collapse
Affiliation(s)
- B M Gadella
- Department of Farm Animal Health, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands.,Department of Biochemistry and Cell Biology, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| | - M A Ferraz
- Department of Farm Animal Health, Faculty of Veterinary Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
45
|
Spade DJ, Hall SJ, Wilson S, Boekelheide K. Di-n-Butyl Phthalate Induces Multinucleated Germ Cells in the Rat Fetal Testis Through a Nonproliferative Mechanism. Biol Reprod 2015; 93:110. [PMID: 26400400 DOI: 10.1095/biolreprod.115.131615] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/18/2015] [Indexed: 01/12/2023] Open
Abstract
In utero exposure to some phthalate esters adversely affects the development of the rat seminiferous cord, causing germ cell loss and increasing the number of multinucleated germ cells (MNGs). To understand the timing of MNG formation and determine whether it requires nuclear division, timed pregnant Sprague Dawley rats were exposed to 500 mg/kg di-n-butyl phthalate (DBP) or corn oil vehicle by oral gavage on Gestational Day (GD) 17 or 18 (0 h) and euthanized after 2, 4, 6, or 24 h or given a second dose at 24 h and euthanized 48 h after the initial dose. Dams were simultaneously exposed to 0.3 M 5-bromo-2'-deoxycitidine (BrdC; converted to 5-bromo-2'-deoxyuridylate [BrdU] in vivo) through a subcutaneous micro-osmotic pump implanted at -2 h. In the testes of male fetuses, DBP induced MNGs significantly beginning at 4-6 h and dramatically by 24 h when exposure began on GD 18 but not GD 17. Seminiferous cord diameter was significantly elevated in testes of rats treated with DBP at 24 and 48 h, and cell death, measured by TUNEL assay, was significantly elevated by DBP only at 48 h, when treatment began on GD 18. TUNEL-labeled MNGs were rare. Overall BrdU labeling rate in the testis was unaffected by DBP. Only one of 606 MNGs in BrdU-labeled sections had a strongly positive nucleus, confirming a nonproliferative mechanism of MNG formation, which is a degenerative process with the potential to adversely affect testis development.
Collapse
Affiliation(s)
- Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - Shelby Wilson
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island
| |
Collapse
|
46
|
Structural and biochemical insights into the role of testis-expressed gene 14 (TEX14) in forming the stable intercellular bridges of germ cells. Proc Natl Acad Sci U S A 2015; 112:12372-7. [PMID: 26392564 DOI: 10.1073/pnas.1418606112] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Intercellular bridges are a conserved feature of spermatogenesis in mammalian germ cells and derive from arresting cell abscission at the final stage of cytokinesis. However, it remains to be fully understood how germ cell abscission is arrested in the presence of general cytokinesis components. The TEX14 (testis-expressed gene 14) protein is recruited to the midbody and plays a key role in the inactivation of germ cell abscission. To gain insights into the structural organization of TEX14 at the midbody, we have determined the crystal structures of the EABR [endosomal sorting complex required for transport (ESCRT) and ALIX-binding region] of CEP55 bound to the TEX14 peptide (or its chimeric peptides) and performed functional characterization of the CEP55-TEX14 interaction by multiexperiment analyses. We show that TEX14 interacts with CEP55-EABR via its AxGPPx3Y (Ala793, Gly795, Pro796, Pro797, and Tyr801) and PP (Pro803 and Pro804) sequences, which together form the AxGPPx3YxPP motif. TEX14 competitively binds to CEP55-EABR to prevent the recruitment of ALIX, which is a component of the ESCRT machinery with the AxGPPx3Y motif. We also demonstrate that a high affinity and a low dissociation rate of TEX14 to CEP55, and an increase in the local concentration of TEX14, cooperatively prevent ALIX from recruiting ESCRT complexes to the midbody. The action mechanism of TEX14 suggests a scheme of how to inactivate the abscission of abnormal cells, including cancer cells.
Collapse
|
47
|
Asano E, Hasegawa H, Hyodo T, Ito S, Maeda M, Chen D, Takahashi M, Hamaguchi M, Senga T. SHCBP1 is required for midbody organization and cytokinesis completion. Cell Cycle 2015; 13:2744-51. [PMID: 25486361 DOI: 10.4161/15384101.2015.945840] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The centralspindlin complex, which is composed of MKLP1 and MgcRacGAP, is one of the crucial factors involved in cytokinesis initiation. Centralspindlin is localized at the middle of the central spindle during anaphase and then concentrates at the midbody to control abscission. A number of proteins that associate with centralspindlin have been identified. These associating factors regulate furrowing and abscission in coordination with centralspindlin. A recent study identified a novel centralspindlin partner, called Nessun Dorma, which is essential for germ cell cytokinesis in Drosophila melanogaster. SHCBP1 is a human ortholog of Nessun Dorma that associates with human centralspindlin. In this report, we analyzed the interaction of SHCBP1 with centralspindlin in detail and determined the regions that are required for the interaction. In addition, we demonstrate that the central region is necessary for the SHCBP1 dimerization. Both MgcRacGAP and MKLP1 are degraded once cells exit mitosis. Similarly, endogenous and exogenous SHCBP1 were degraded with mitosis progression. Interestingly, SHCBP1 expression was significantly reduced in the absence of centralspindlin, whereas centralspindlin expression was not affected by SHCBP1 knockdown. Finally, we demonstrate that SHCBP1 depletion promotes midbody structure disruption and inhibits abscission, a final stage of cytokinesis. Our study gives novel insight into the role of SHCBP in cytokinesis completion.
Collapse
Affiliation(s)
- Eri Asano
- a Division of Cancer Biology ; Nagoya University Graduate School of Medicine ; Nagoya , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tormos AM, Taléns-Visconti R, Sastre J. Regulation of cytokinesis and its clinical significance. Crit Rev Clin Lab Sci 2015; 52:159-67. [DOI: 10.3109/10408363.2015.1012191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
49
|
Rabionet M, Bayerle A, Jennemann R, Heid H, Fuchser J, Marsching C, Porubsky S, Bolenz C, Guillou F, Gröne HJ, Gorgas K, Sandhoff R. Male meiotic cytokinesis requires ceramide synthase 3-dependent sphingolipids with unique membrane anchors. Hum Mol Genet 2015; 24:4792-808. [DOI: 10.1093/hmg/ddv204] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022] Open
|
50
|
Pennarossa G, Maffei S, Tettamanti G, Congiu T, deEguileor M, Gandolfi F, Brevini TAL. Intercellular bridges are essential for human parthenogenetic cell survival. Mech Dev 2015; 136:30-9. [PMID: 25700933 DOI: 10.1016/j.mod.2015.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 12/25/2022]
Abstract
Parthenogenetic cells, obtained from in vitro activated mammalian oocytes, display multipolar spindles, chromosome malsegregation and a high incidence of aneuploidy, probably due to the lack of paternal contribution. Despite this, parthenogenetic cells do not show high rates of apoptosis and are able to proliferate in a way comparable to their biparental counterpart. We hypothesize that a series of adaptive mechanisms are present in parthenogenetic cells, allowing a continuous proliferation and ordinate cell differentiation both in vitro and in vivo. Here we identify the presence of intercellular bridges that contribute to the establishment of a wide communication network among human parthenogenetic cells, providing a mutual exchange of missing products. Silencing of two molecules essential for intercellular bridge formation and maintenance demonstrates the key function played by these cytoplasmic passageways that ensure normal cell functions and survival, alleviating the unbalance in cellular component composition.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Laboratory of Biomedical Embryology, UniStem, Center For Stem Cell Research, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy
| | - Sara Maffei
- Laboratory of Biomedical Embryology, UniStem, Center For Stem Cell Research, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy
| | - Gianluca Tettamanti
- Department of Biotechnology and Life Sciences, Università degli Studi dell'Insubria, Varese, Italy
| | - Terenzio Congiu
- Department of Surgical and Morphological Science, Università degli Studi dell'Insubria, Varese, Italy
| | - Magda deEguileor
- Department of Biotechnology and Life Sciences, Università degli Studi dell'Insubria, Varese, Italy
| | - Fulvio Gandolfi
- Laboratory of Biomedical Embryology, UniStem, Center For Stem Cell Research, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy
| | - Tiziana A L Brevini
- Laboratory of Biomedical Embryology, UniStem, Center For Stem Cell Research, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|