1
|
Lenartowski R, Ostrowski J, Suwińska A, Richert A, Zakrzewski P, Izdebska M, Arendt W, Miller KG, Lenartowska M. Myosin VI is expressed in developing ovarian follicles in Drosophila but is not essential for effective oogenesis. Front Cell Dev Biol 2025; 13:1535117. [PMID: 40530337 PMCID: PMC12171261 DOI: 10.3389/fcell.2025.1535117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/22/2025] [Indexed: 06/20/2025] Open
Abstract
Myosin VI is the only actin-based motor known to move toward the minus end of actin filaments. This protein is involved in many different cellular processes, such as endocytosis, autophagy, secretion, and regulation of actin organization and dynamics. Myosin VI has also been suggested to play an important role in collective migration of border cells and egg chamber development during Drosophila oogenesis. Here we show for the first time that myosin VI is expressed in Drosophila germarium as well as in early ovarian follicles, especially in the developing oocyte. As oogenesis progresses, the level of myosin VI in maturing egg chambers decreases, but this protein is present both in the nascent border cell cluster, during its delamination from the epithelium, and then during the early stages of border cell migration. However, we demonstrate that myosin VI deficiency in border cells, or even complete lack of this protein in myosin VI mutant do not inhibit border cell migration. Moreover, deficiency/lack of myosin VI does not cause any serious defects in ovarian morphology, egg chamber morphogenesis, oogenesis, and egg development. Thus we conclude that myosin VI is not a key player in Drosophila oogenesis.
Collapse
Affiliation(s)
- Robert Lenartowski
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Jakub Ostrowski
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Anna Suwińska
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Anna Richert
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Przemysław Zakrzewski
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Magdalena Izdebska
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Wioletta Arendt
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Torun, Poland
| | - Kathryn G. Miller
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Marta Lenartowska
- Department of Cellular and Molecular Biology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Torun, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Toruń, Torun, Poland
| |
Collapse
|
2
|
Kotb NM, Ulukaya G, Ramamoorthy A, Park LS, Tang J, Hasson D, Rangan P. TORC1-driven translation of Nucleoporin44A promotes chromatin remodeling and germ cell-to-maternal transition in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643309. [PMID: 40161787 PMCID: PMC11952567 DOI: 10.1101/2025.03.14.643309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Oocyte specification is a critical developmental transition that requires the coordinated repression of germ cell-specific genes and activation of the maternal program to support embryogenesis. In Drosophila, the timely repression of germ cell and early oogenesis genes is essential for this transition, yet the mechanisms that coordinate this process remain unclear. Here, we uncover an unexpected translation-chromatin axis, where transient Target of Rapamycin Complex 1 (TORC1)-driven translation triggers chromatin remodeling, ensuring irreversible oocyte fate commitment. Through a screen, we identified ribosome biogenesis regulators, including Zinc finger protein RP-8 (Zfrp8) and TORC1 components, as key mediators of gene silencing. We show that TORC1 activity increases during oocyte specification, and disrupting ribosome biogenesis, translation, or TORC1 function prevents proper heterochromatin formation, leading to epigenetic instability. Polysome-seq analysis of zfrp8-depleted ovaries revealed that Zfrp8 is required for the translation of Nucleoporin 44A (Nup44A), a key nuclear pore complex (NPC) component. Given the role of the NPC in chromatin organization, independent disruption of Nup44A results in defective silencing of the germ cell and early oogenesis genes. Our findings reveal a mechanism in which translation-driven NPC remodeling coordinates heterochromatin establishment, facilitating the germ cell-to-maternal transition and ensuring proper oocyte fate commitment.
Collapse
Affiliation(s)
- Noor M. Kotb
- Department of Biomedical Sciences/Wadsworth Center, University at Albany State University of New York (SUNY), Albany, New York 12202, USA
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NewYork 10029, USA
- Current address, Hologic Diagenode, 400 Morris Avenue, Suite 101, Denville, New Jersey 07834, USA
| | - Gulay Ulukaya
- Bioinformatics for Next-Generation Sequencing (BiNGS) Core, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Anupriya Ramamoorthy
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NewYork 10029, USA
| | - Lina Seojin Park
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NewYork 10029, USA
| | - Julia Tang
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NewYork 10029, USA
| | - Dan Hasson
- Bioinformatics for Next-Generation Sequencing (BiNGS) Core, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Prashanth Rangan
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NewYork 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
3
|
Das S, Hegde S, Wagh N, Sudhakaran J, Roy AE, Deshpande G, Ratnaparkhi GS. Caspar specifies primordial germ cell count and identity in Drosophila melanogaster. eLife 2024; 13:RP98584. [PMID: 39671304 PMCID: PMC11643641 DOI: 10.7554/elife.98584] [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] [Indexed: 12/15/2024] Open
Abstract
Repurposing of pleiotropic factors during execution of diverse cellular processes has emerged as a regulatory paradigm. Embryonic development in metazoans is controlled by maternal factors deposited in the egg during oogenesis. Here, we explore maternal role(s) of Caspar (Casp), the Drosophila orthologue of human Fas-associated factor-1 (FAF1) originally implicated in host-defense as a negative regulator of NF-κB signaling. Maternal loss of either Casp or it's protein partner, transitional endoplasmic reticulum 94 (TER94) leads to partial embryonic lethality correlated with aberrant centrosome behavior, cytoskeletal abnormalities, and defective gastrulation. Although ubiquitously distributed, both proteins are enriched in the primordial germ cells (PGCs), and in keeping with the centrosome problems, mutant embryos display a significant reduction in the PGC count. Moreover, the total number of pole buds is directly proportional to the level of Casp. Consistently, it's 'loss' and 'gain' results in respective reduction and increase in the Oskar protein levels, the master determinant of PGC fate. To elucidate this regulatory loop, we analyzed several known components of mid-blastula transition and identify the translational repressor Smaug, a zygotic regulator of germ cell specification, as a potential critical target. We present a detailed structure-function analysis of Casp aimed at understanding its novel involvement during PGC development.
Collapse
Affiliation(s)
- Subhradip Das
- Department of Biology, Indian Institute of Science Education & ResearchPuneIndia
| | - Sushmitha Hegde
- Department of Biology, Indian Institute of Science Education & ResearchPuneIndia
| | - Neel Wagh
- Department of Biology, Indian Institute of Science Education & ResearchPuneIndia
| | - Jyothish Sudhakaran
- Department of Biology, Indian Institute of Science Education & ResearchPuneIndia
| | - Adheena Elsa Roy
- Department of Biology, Indian Institute of Science Education & ResearchPuneIndia
| | - Girish Deshpande
- Department of Biology, Indian Institute of Science Education & ResearchPuneIndia
- Department of Molecular Biology, Princeton UniversityPrincetonUnited States
| | - Girish S Ratnaparkhi
- Department of Biology, Indian Institute of Science Education & ResearchPuneIndia
| |
Collapse
|
4
|
Vallés AM, Rubin T, Macaisne N, Dal Toe L, Molla-Herman A, Antoniewski C, Huynh JR. Transcriptomic analysis of meiotic genes during the mitosis-to-meiosis transition in Drosophila females. Genetics 2024; 228:iyae130. [PMID: 39225982 DOI: 10.1093/genetics/iyae130] [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: 12/22/2023] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Germline cells produce gametes, which are specialized cells essential for sexual reproduction. Germline cells first amplify through several rounds of mitosis before switching to the meiotic program, which requires specific sets of proteins for DNA recombination, chromosome pairing, and segregation. Surprisingly, we previously found that some proteins of the synaptonemal complex, a prophase I meiotic structure, are already expressed and required in the mitotic region of Drosophila females. Here, to assess if additional meiotic genes were expressed earlier than expected, we isolated mitotic and meiotic cell populations to compare their RNA content. Our transcriptomic analysis reveals that all known meiosis I genes are already expressed in the mitotic region; however, only some of them are translated. As a case study, we focused on mei-W68, the Drosophila homolog of Spo11, to assess its expression at both the mRNA and protein levels and used different mutant alleles to assay for a premeiotic function. We could not detect any functional role for Mei-W68 during homologous chromosome pairing in dividing germ cells. Our study paves the way for further functional analysis of meiotic genes expressed in the mitotic region.
Collapse
Affiliation(s)
- Ana Maria Vallés
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Thomas Rubin
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Nicolas Macaisne
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Laurine Dal Toe
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Anahi Molla-Herman
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Christophe Antoniewski
- ARTbio Bioinformatics Analysis Facility, IBPS, CNRS, Sorbonne Université, Institut Français de Bioinformatique, 75005 Paris, France
| | - Jean-René Huynh
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| |
Collapse
|
5
|
Jaglarz MK, Kuziak A, Jankowska W. The pattern of the follicle cell diversification in ovarian follicles of the true fruit flies, Tephritidae. J Anat 2024; 245:643-657. [PMID: 38817113 PMCID: PMC11424825 DOI: 10.1111/joa.14065] [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: 03/14/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024] Open
Abstract
In flies (Diptera), the ovary displays several distinct patterns of the follicular epithelium formation and diversification. Two main patterns have been identified in the true flies or Brachycera, namely the Rhagio type and the Drosophila type. These patterns align with the traditional division of Brachycera into Orthorrhapha and Cyclorrhapha. However, studies of the follicular epithelium morphogenesis in cyclorrhaphans other than Drosophila are scarce. We characterise the developmental changes associated with the emergence of follicle cell (FC) diversity in two cyclorrhaphans belonging to the family Tephritidae (Brachycera, Cyclorrhapha). Our analysis revealed that the diversification of FCs in these species shows characteristics of both the Rhagio and Drosophila types. First, a distinct cluster of FCs, consisting of polar cells and border-like cells, differentiates at the posterior pole of the ovarian follicle. This feature is unique to the Rhagio type and has only been reported in species representing the Orthorrhapha group. Second, morphological criteria have identified a significantly smaller number of subpopulations of FCs than in Drosophila. Furthermore, while the general pattern of FC migration is similar to that of Drosophila, the distinctive migration of the anterior-dorsal FCs is absent. In the studied tephritids, the migration of the anterior polar cell/border cell cluster towards the anterior pole of the oocyte is followed by the posterior migration of the main body cuboidal FCs to cover the expanding oocyte. Finally, during the onset of vitellogenesis, a distinct subset of FCs migrates towards the centre of the ovarian follicle to cover the oocyte's anterior pole. Our study also highlights specific actions of some FCs that accompany the migration process, which has not been previously documented in cyclorrhaphans. These results support the hypothesis that the posterior and centripetal migrations of morphologically unique FC subsets arose in the common ancestor of Cyclorrhapha. These events appear to have occurred fairly recently in the evolutionary timeline of Diptera.
Collapse
Affiliation(s)
- Mariusz K Jaglarz
- Department of Developmental Biology and Invertebrate Morphology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Kraków, Poland
| | - Agata Kuziak
- Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Wladyslawa Jankowska
- Department of Developmental Biology and Invertebrate Morphology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Kraków, Poland
| |
Collapse
|
6
|
Mathieu J, Huynh JR. Incomplete divisions between sister germline cells require Usp8 function. C R Biol 2024; 347:109-117. [PMID: 39345214 DOI: 10.5802/crbiol.161] [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: 05/28/2024] [Revised: 07/16/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024]
Abstract
Cytokinetic abscission is the final step of cell division, resulting in two separate daughter cells. While abscission is typically complete across most cell types, germline cells, which produce sexual gametes, do not finish cytokinesis, maintaining connections between sister cells. These connections are essential for sharing cytoplasm as they differentiate into oocyte and sperm. First, we outline the molecular events of cytokinesis during both complete and delayed abscission, highlighting the role of the ESCRT-III proteins. We then focus on recent discoveries that reveal the molecular mechanisms blocking abscission in Drosophila germline cells. The enzyme Usp8 was identified as vital for ensuring incomplete cytokinesis through the regulation of ESCRT-III ubiquitination and localization. Finally, we explore how the processes of incomplete cytokinesis could hold evolutionary importance, suggesting additional studies into choanoflagellates to comprehend the origins of multicellularity.
Collapse
|
7
|
Barr J, Diegmiller R, Colonnetta MM, Ke W, Imran Alsous J, Stern T, Shvartsman SY, Schedl P. To be or not to be: orb, the fusome and oocyte specification in Drosophila. Genetics 2024; 226:iyae020. [PMID: 38345426 PMCID: PMC10990432 DOI: 10.1093/genetics/iyae020] [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: 12/17/2023] [Accepted: 01/21/2024] [Indexed: 04/05/2024] Open
Abstract
In the fruit fly Drosophila melanogaster, two cells in a cyst of 16 interconnected cells have the potential to become the oocyte, but only one of these will assume an oocyte fate as the cysts transition through regions 2a and 2b of the germarium. The mechanism of specification depends on a polarized microtubule network, a dynein dependent Egl:BicD mRNA cargo complex, a special membranous structure called the fusome and its associated proteins, and the translational regulator orb. In this work, we have investigated the role of orb and the fusome in oocyte specification. We show here that specification is a stepwise process. Initially, orb mRNAs accumulate in the two pro-oocytes in close association with the fusome. This association is accompanied by the activation of the orb autoregulatory loop, generating high levels of Orb. Subsequently, orb mRNAs become enriched in only one of the pro-oocytes, the presumptive oocyte, and this is followed, with a delay, by Orb localization to the oocyte. We find that fusome association of orb mRNAs is essential for oocyte specification in the germarium, is mediated by the orb 3' UTR, and requires Orb protein. We also show that the microtubule minus end binding protein Patronin functions downstream of orb in oocyte specification. Finally, in contrast to a previously proposed model for oocyte selection, we find that the choice of which pro-oocyte becomes the oocyte does not seem to be predetermined by the amount of fusome material in these two cells, but instead depends upon a competition for orb gene products.
Collapse
Affiliation(s)
- Justinn Barr
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Rocky Diegmiller
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Megan M Colonnetta
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Wenfan Ke
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jasmin Imran Alsous
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Flatiron Institute, Simons Foundation, New York, NY 10010, USA
| | - Tomer Stern
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Stanislav Y Shvartsman
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Flatiron Institute, Simons Foundation, New York, NY 10010, USA
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
8
|
Lepesant JA, Roland-Gosselin F, Guillemet C, Bernard F, Guichet A. The Importance of the Position of the Nucleus in Drosophila Oocyte Development. Cells 2024; 13:201. [PMID: 38275826 PMCID: PMC10814754 DOI: 10.3390/cells13020201] [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: 11/22/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Oogenesis is a developmental process leading to the formation of an oocyte, a haploid gamete, which upon fertilisation and sperm entry allows the male and the female pronuclei to fuse and give rise to a zygote. In addition to forming a haploid gamete, oogenesis builds up a store of proteins, mRNAs, and organelles in the oocyte needed for the development of the future embryo. In several species, such as Drosophila, the polarity axes determinants of the future embryo must be asymmetrically distributed prior to fertilisation. In the Drosophila oocyte, the correct positioning of the nucleus is essential for establishing the dorsoventral polarity axis of the future embryo and allowing the meiotic spindles to be positioned in close vicinity to the unique sperm entry point into the oocyte.
Collapse
Affiliation(s)
| | | | | | | | - Antoine Guichet
- Université Paris Cité, CNRS, Institut Jacques Monod, 75013 Paris, France; (J.-A.L.); (F.R.-G.); (C.G.); (F.B.)
| |
Collapse
|
9
|
Berg C, Sieber M, Sun J. Finishing the egg. Genetics 2024; 226:iyad183. [PMID: 38000906 PMCID: PMC10763546 DOI: 10.1093/genetics/iyad183] [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: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/26/2023] Open
Abstract
Gamete development is a fundamental process that is highly conserved from early eukaryotes to mammals. As germ cells develop, they must coordinate a dynamic series of cellular processes that support growth, cell specification, patterning, the loading of maternal factors (RNAs, proteins, and nutrients), differentiation of structures to enable fertilization and ensure embryonic survival, and other processes that make a functional oocyte. To achieve these goals, germ cells integrate a complex milieu of environmental and developmental signals to produce fertilizable eggs. Over the past 50 years, Drosophila oogenesis has risen to the forefront as a system to interrogate the sophisticated mechanisms that drive oocyte development. Studies in Drosophila have defined mechanisms in germ cells that control meiosis, protect genome integrity, facilitate mRNA trafficking, and support the maternal loading of nutrients. Work in this system has provided key insights into the mechanisms that establish egg chamber polarity and patterning as well as the mechanisms that drive ovulation and egg activation. Using the power of Drosophila genetics, the field has begun to define the molecular mechanisms that coordinate environmental stresses and nutrient availability with oocyte development. Importantly, the majority of these reproductive mechanisms are highly conserved throughout evolution, and many play critical roles in the development of somatic tissues as well. In this chapter, we summarize the recent progress in several key areas that impact egg chamber development and ovulation. First, we discuss the mechanisms that drive nutrient storage and trafficking during oocyte maturation and vitellogenesis. Second, we examine the processes that regulate follicle cell patterning and how that patterning impacts the construction of the egg shell and the establishment of embryonic polarity. Finally, we examine regulatory factors that control ovulation, egg activation, and successful fertilization.
Collapse
Affiliation(s)
- Celeste Berg
- Department of Genome Sciences, University of Washington, Seattle, WA 98195-5065 USA
| | - Matthew Sieber
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jianjun Sun
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269 USA
| |
Collapse
|
10
|
Tian Y, Liu L, Gao J, Wang R. Homologous chromosome pairing: The linchpin of accurate segregation in meiosis. J Cell Physiol 2024; 239:3-19. [PMID: 38032002 DOI: 10.1002/jcp.31166] [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: 09/09/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 12/01/2023]
Abstract
Meiosis is a specialized cell division that occurs in sexually reproducing organisms, generating haploid gametes containing half the chromosome number through two rounds of cell division. Homologous chromosomes pair and prepare for their proper segregation in subsequent divisions. How homologous chromosomes recognize each other and achieve pairing is an important question. Early studies showed that in most organisms, homologous pairing relies on homologous recombination. However, pairing mechanisms differ across species. Evidence indicates that chromosomes are dynamic and move during early meiotic stages, facilitating pairing. Recent studies in various model organisms suggest conserved mechanisms and key regulators of homologous chromosome pairing. This review summarizes these findings and compare similarities and differences in homologous chromosome pairing mechanisms across species.
Collapse
Affiliation(s)
- Yuqi Tian
- Center for Cell Structure and Function, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Shandong Normal University, Jinan, China
| | - Libo Liu
- Center for Cell Structure and Function, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Shandong Normal University, Jinan, China
| | - Jinmin Gao
- Center for Cell Structure and Function, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Shandong Normal University, Jinan, China
| | - Ruoxi Wang
- Center for Cell Structure and Function, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Shandong Normal University, Jinan, China
| |
Collapse
|
11
|
Sarkar K, Kotb NM, Lemus A, Martin ET, McCarthy A, Camacho J, Iqbal A, Valm AM, Sammons MA, Rangan P. A feedback loop between heterochromatin and the nucleopore complex controls germ-cell-to-oocyte transition during Drosophila oogenesis. Dev Cell 2023; 58:2580-2596.e6. [PMID: 37673064 PMCID: PMC11301765 DOI: 10.1016/j.devcel.2023.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 04/12/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023]
Abstract
Germ cells differentiate into oocytes that launch the next generation upon fertilization. How the highly specialized oocyte acquires this distinct cell fate is poorly understood. During Drosophila oogenesis, H3K9me3 histone methyltransferase SETDB1 translocates from the cytoplasm to the nucleus of germ cells concurrently with oocyte specification. Here, we discovered that nuclear SETDB1 is required for silencing a cohort of differentiation-promoting genes by mediating their heterochromatinization. Intriguingly, SETDB1 is also required for upregulating 18 of the ∼30 nucleoporins (Nups) that compose the nucleopore complex (NPC), promoting NPC formation. NPCs anchor SETDB1-dependent heterochromatin at the nuclear periphery to maintain H3K9me3 and gene silencing in the egg chambers. Aberrant gene expression due to the loss of SETDB1 or Nups results in the loss of oocyte identity, cell death, and sterility. Thus, a feedback loop between heterochromatin and NPCs promotes transcriptional reprogramming at the onset of oocyte specification, which is critical for establishing oocyte identity.
Collapse
Affiliation(s)
- Kahini Sarkar
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Noor M Kotb
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA; Department of Biomedical Sciences, School of Public Health, University at Albany SUNY, Albany, NY 12222, USA
| | - Alex Lemus
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Elliot T Martin
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Alicia McCarthy
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Justin Camacho
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Ayman Iqbal
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Alex M Valm
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Prashanth Rangan
- Department of Biological Sciences and RNA Institute, University at Albany SUNY, Albany, NY 12222, USA.
| |
Collapse
|
12
|
Kotb NM, Ulukaya G, Chavan A, Nguyen SC, Proskauer L, Joyce E, Hasson D, Jagannathan M, Rangan P. Genome organization regulates nuclear pore complex formation and promotes differentiation during Drosophila oogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.15.567233. [PMID: 38014330 PMCID: PMC10680722 DOI: 10.1101/2023.11.15.567233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Genome organization can regulate gene expression and promote cell fate transitions. The differentiation of germline stem cells (GSCs) to oocytes in Drosophila involves changes in genome organization mediated by heterochromatin and the nuclear pore complex (NPC). Heterochromatin represses germ-cell genes during differentiation and NPCs anchor these silenced genes to the nuclear periphery, maintaining silencing to allow for oocyte development. Surprisingly, we find that genome organization also contributes to NPC formation, mediated by the transcription factor Stonewall (Stwl). As GSCs differentiate, Stwl accumulates at boundaries between silenced and active gene compartments. Stwl at these boundaries plays a pivotal role in transitioning germ-cell genes into a silenced state and activating a group of oocyte genes and Nucleoporins (Nups). The upregulation of these Nups during differentiation is crucial for NPC formation and further genome organization. Thus, crosstalk between genome architecture and NPCs is essential for successful cell fate transitions.
Collapse
Affiliation(s)
- Noor M. Kotb
- Department of Biomedical Sciences/Wadsworth Center, University at Albany SUNY, Albany, NY 12202
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12202
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Gulay Ulukaya
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) core
| | - Ankita Chavan
- Department of Biology, Institute of Biochemistry, ETH Zurich, 8092 Zurich
| | - Son C. Nguyen
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104
| | - Lydia Proskauer
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12202
- Current address: Biochemistry and Molecular Biology Department, University of Massachusetts Amherst, Amherst, MA 01003
| | - Eric Joyce
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104
| | - Dan Hasson
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Tisch Cancer Institute Bioinformatics for Next Generation Sequencing (BiNGS) core
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhav Jagannathan
- Department of Biology, Institute of Biochemistry, ETH Zurich, 8092 Zurich
| | - Prashanth Rangan
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
13
|
Jung GI, Londoño-Vásquez D, Park S, Skop AR, Balboula AZ, Schindler K. An oocyte meiotic midbody cap is required for developmental competence in mice. Nat Commun 2023; 14:7419. [PMID: 37973997 PMCID: PMC10654508 DOI: 10.1038/s41467-023-43288-x] [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: 07/25/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
Embryo development depends upon maternally derived materials. Mammalian oocytes undergo extreme asymmetric cytokinesis events, producing one large egg and two small polar bodies. During cytokinesis in somatic cells, the midbody and subsequent assembly of the midbody remnant, a signaling organelle containing RNAs, transcription factors and translation machinery, is thought to influence cellular function or fate. The role of the midbody and midbody remnant in gametes, in particular, oocytes, remains unclear. Here, we examined the formation and function of meiotic midbodies (mMB) and mMB remnants using mouse oocytes and demonstrate that mMBs have a specialized cap structure that is orientated toward polar bodies. We show that that mMBs are translationally active, and that mMB caps are required to retain nascent proteins in eggs. We propose that this specialized mMB cap maintains genetic factors in eggs allowing for full developmental competency.
Collapse
Affiliation(s)
- Gyu Ik Jung
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Human Genetics Institute of New Jersey, Piscataway, NJ, USA
| | | | - Sungjin Park
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Ahna R Skop
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Ahmed Z Balboula
- Animal Sciences Research Center, University of Missouri, Columbia, MO, USA
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
- Human Genetics Institute of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
14
|
Smart M, Shvartsman SY, Nunley H. A model of replicating coupled oscillators generates naturally occurring cell networks. Development 2023; 150:dev202187. [PMID: 37823332 PMCID: PMC10690053 DOI: 10.1242/dev.202187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
When a founder cell and its progeny divide with incomplete cytokinesis, a network forms in which each intercellular bridge corresponds to a past mitotic event. Such networks are required for gamete production in many animals, and different species have evolved diverse final network topologies. Although mechanisms regulating network assembly have been identified in particular organisms, we lack a quantitative framework to understand network assembly and inter-species variability. Motivated by cell networks responsible for oocyte production in invertebrates, where the final topology is typically invariant within each species, we devised a mathematical model for generating cell networks, in which each node is an oscillator and, after a full cycle, the node produces a daughter to which it remains connected. These cell cycle oscillations are transient and coupled via diffusion over the edges of the network. By variation of three biologically motivated parameters, our model generates nearly all such networks currently reported across invertebrates. Furthermore, small parameter variations can rationalize cases of intra-species variation. Because cell networks outside of the ovary often form less deterministically, we propose model generalizations to account for sources of stochasticity.
Collapse
Affiliation(s)
- Matthew Smart
- Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
| | - Stanislav Y. Shvartsman
- Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
- Department of Molecular Biology and Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Hayden Nunley
- Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA
| |
Collapse
|
15
|
Wang ZH, Zhao W, Combs CA, Zhang F, Knutson JR, Lilly MA, Xu H. Mechanical stimulation from the surrounding tissue activates mitochondrial energy metabolism in Drosophila differentiating germ cells. Dev Cell 2023; 58:2249-2260.e9. [PMID: 37647895 PMCID: PMC10843713 DOI: 10.1016/j.devcel.2023.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/24/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
In multicellular lives, the differentiation of stem cells and progenitor cells is often accompanied by a transition from glycolysis to mitochondrial oxidative phosphorylation (OXPHOS). However, the underlying mechanism of this metabolic transition remains largely unknown. In this study, we investigate the role of mechanical stress in activating OXPHOS during differentiation of the female germline cyst in Drosophila. We demonstrate that the surrounding somatic cells flatten the 16-cell differentiating cyst, resulting in an increase of the membrane tension of germ cells inside the cyst. This mechanical stress is necessary to maintain cytosolic Ca2+ concentration in germ cells through a mechanically activated channel, transmembrane channel-like. The sustained cytosolic Ca2+ triggers a CaMKI-Fray-JNK signaling relay, leading to the transcriptional activation of OXPHOS in differentiating cysts. Our findings demonstrate a molecular link between cell mechanics and mitochondrial energy metabolism, with implications for other developmentally orchestrated metabolic transitions in mammals.
Collapse
Affiliation(s)
- Zong-Heng Wang
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wenjing Zhao
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christian A Combs
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fan Zhang
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jay R Knutson
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mary A Lilly
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hong Xu
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Schindler K, Jung GI, Londoño-Vásquez D, Park S, Skop A, Balboula A. An oocyte meiotic midbody cap is required for developmental competence in mice. RESEARCH SQUARE 2023:rs.3.rs-3399188. [PMID: 37886573 PMCID: PMC10602078 DOI: 10.21203/rs.3.rs-3399188/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Embryo development depends upon maternally derived materials. Mammalian oocytes undergo extreme asymmetric cytokinesis events, producing one large egg and two small polar bodies (PB). During cytokinesis in somatic cells, the midbody (MB) and subsequent assembly of the midbody remnant (MBR), a signaling organelle containing RNAs, transcription factors and translation machinery, is thought to influence cellular function or fate. The role of the MB and MBR in gametes, in particular, oocytes, remains unclear. Here, we examined the formation and function of meiotic MBs (mMB) and mMB remnants (mMBRs) using mouse oocytes and demonstrate that mMBs have a specialized meiotic mMB cap structure that is orientated toward PBs. We show that that mMBs are translationally active, and that mMB caps are required to retain nascent proteins in eggs. We propose that this specialized mMB cap maintains genetic factors in eggs allowing for full developmental competency.
Collapse
|
17
|
Breznak SM, Peng Y, Deng L, Kotb NM, Flamholz Z, Rapisarda IT, Martin ET, LaBarge KA, Fabris D, Gavis ER, Rangan P. H/ACA snRNP-dependent ribosome biogenesis regulates translation of polyglutamine proteins. SCIENCE ADVANCES 2023; 9:eade5492. [PMID: 37343092 PMCID: PMC10284551 DOI: 10.1126/sciadv.ade5492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 05/17/2023] [Indexed: 06/23/2023]
Abstract
Stem cells in many systems, including Drosophila germline stem cells (GSCs), increase ribosome biogenesis and translation during terminal differentiation. Here, we show that the H/ACA small nuclear ribonucleoprotein (snRNP) complex that promotes pseudouridylation of ribosomal RNA (rRNA) and ribosome biogenesis is required for oocyte specification. Reducing ribosome levels during differentiation decreased the translation of a subset of messenger RNAs that are enriched for CAG trinucleotide repeats and encode polyglutamine-containing proteins, including differentiation factors such as RNA-binding Fox protein 1. Moreover, ribosomes were enriched at CAG repeats within transcripts during oogenesis. Increasing target of rapamycin (TOR) activity to elevate ribosome levels in H/ACA snRNP complex-depleted germlines suppressed the GSC differentiation defects, whereas germlines treated with the TOR inhibitor rapamycin had reduced levels of polyglutamine-containing proteins. Thus, ribosome biogenesis and ribosome levels can control stem cell differentiation via selective translation of CAG repeat-containing transcripts.
Collapse
Affiliation(s)
- Shane M. Breznak
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
| | - Yingshi Peng
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Limin Deng
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
- Department of Chemistry, University of Connecticut, 55N Eagleville Rd, Storrs, CT 06269, USA
| | - Noor M. Kotb
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
- Department of Biomedical Sciences, University at Albany School of Public Health, Albany, NY 12144, USA
| | - Zachary Flamholz
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Ian T. Rapisarda
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
- Lake Erie College of Osteopathic Medicine, College of Medicine, 1858 W Grandview Blvd, Erie, PA 16509, USA
| | - Elliot T. Martin
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
| | - Kara A. LaBarge
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
| | - Dan Fabris
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
- Department of Chemistry, University of Connecticut, 55N Eagleville Rd, Storrs, CT 06269, USA
| | - Elizabeth R. Gavis
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Prashanth Rangan
- Department of Biological Sciences, RNA Institute, University at Albany, 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222, USA
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
18
|
Neiswender H, Baker FC, Veeranan-Karmegam R, Allen P, Gonsalvez GB. dTtc1, a conserved tetratricopeptide repeat protein, is required for maturation of Drosophila egg chambers via its role in stabilizing electron transport chain components. Front Cell Dev Biol 2023; 11:1148773. [PMID: 37333987 PMCID: PMC10272552 DOI: 10.3389/fcell.2023.1148773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
We recently identified the Drosophila ortholog of TTC1 (dTtc1) as an interacting partner of Egalitarian, an RNA adaptor of the Dynein motor. In order to better understand the function of this relatively uncharacterized protein, we depleted dTtc1 in the Drosophila female germline. Depletion of dTtc1 resulted in defective oogenesis and no mature eggs were produced. A closer examination revealed that mRNA cargoes normally transported by Dynein were relatively unaffected. However, mitochondria in dTtc1 depleted egg chambers displayed an extremely swollen phenotype. Ultrastructural analysis revealed a lack of cristae. These phenotypes were not observed upon disruption of Dynein. Thus, this function of dTtc1 is likely to be Dynein independent. Consistent with a role for dTtc1 in mitochondrial biology, a published proteomics screen revealed that dTtc1 interacts with numerous components of electron transport chain (ETC) complexes. Our results indicate that the expression level of several of these ETC components was significantly reduced upon depletion of dTtc1. Importantly, this phenotype was completely rescued upon expression of wild-type GFP-dTtc1 in the depleted background. Lastly, we demonstrate that the mitochondrial phenotype caused by a lack of dTtc1 is not restricted to the germline but is also observed in somatic tissues. Our model suggests that dTtc1, likely in combination with cytoplasmic chaperones, is required for stabilizing ETC components.
Collapse
|
19
|
Cabrita B, Martinho RG. Genetic and Epigenetic Regulation of Drosophila Oocyte Determination. J Dev Biol 2023; 11:21. [PMID: 37367475 DOI: 10.3390/jdb11020021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023] Open
Abstract
Primary oocyte determination occurs in many organisms within a germ line cyst, a multicellular structure composed of interconnected germ cells. However, the structure of the cyst is itself highly diverse, which raises intriguing questions about the benefits of this stereotypical multicellular environment for female gametogenesis. Drosophila melanogaster is a well-studied model for female gametogenesis, and numerous genes and pathways critical for the determination and differentiation of a viable female gamete have been identified. This review provides an up-to-date overview of Drosophila oocyte determination, with a particular emphasis on the mechanisms that regulate germ line gene expression.
Collapse
Affiliation(s)
- Brigite Cabrita
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| | - Rui Gonçalo Martinho
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Agra do Crasto, Edifício 30, 3810-193 Aveiro, Portugal
| |
Collapse
|
20
|
Lovegrove MR, Dearden PK, Duncan EJ. Honeybee queen mandibular pheromone induces a starvation response in Drosophila melanogaster. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 154:103908. [PMID: 36657589 DOI: 10.1016/j.ibmb.2023.103908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/27/2022] [Accepted: 01/13/2023] [Indexed: 06/17/2023]
Abstract
Eusocial insect societies are defined by the reproductive division of labour, a social structure that is generally enforced by the reproductive dominant(s) or 'queen(s)'. Reproductive dominance is maintained through behavioural dominance or production of queen pheromones, or a mixture of both. Queen mandibular pheromone (QMP) is a queen pheromone produced by queen honeybees (Apis mellifera) which represses reproduction in worker honeybees. How QMP acts to repress worker reproduction, the mechanisms by which this repression is induced, and how it has evolved this activity, remain poorly understood. Surprisingly, QMP is capable of repressing reproduction in non-target arthropods. Here we show that in Drosophila melanogaster QMP treatment mimics the starvation response, disrupting reproduction. QMP exposure induces an increase in food consumption and activation of checkpoints in the ovary that reduce fecundity and depresses insulin signalling. The magnitude of these effects is indistinguishable between QMP-treated and starved individuals. As QMP triggers a starvation response in an insect diverged from honeybees, we propose that QMP originally evolved by co-opting nutrition signalling pathways to regulate reproduction.
Collapse
Affiliation(s)
- Mackenzie R Lovegrove
- Genomics Aotearoa and Biochemistry Department, University of Otago, P.O. Box 56, Dunedin, Aotearoa, New Zealand; School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Peter K Dearden
- Genomics Aotearoa and Biochemistry Department, University of Otago, P.O. Box 56, Dunedin, Aotearoa, New Zealand.
| | - Elizabeth J Duncan
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
21
|
Abstract
In this chapter, we highlight examples of the diverse array of developmental, cellular, and biochemical insights that can be gained by using Drosophila melanogaster oogenesis as a model tissue. We begin with an overview of ovary development and adult oogenesis. Then we summarize how the adult Drosophila ovary continues to advance our understanding of stem cells, cell cycle, cell migration, cytoplasmic streaming, nurse cell dumping, and cell death. We also review emerging areas of study, including the roles of lipid droplets, ribosomes, and nuclear actin in egg development. Finally, we conclude by discussing the growing conservation of processes and signaling pathways that regulate oogenesis and female reproduction from flies to humans.
Collapse
|
22
|
Saha B, Acharjee S, Ghosh G, Dasgupta P, Prasad M. Germline protein, Cup, non-cell autonomously limits migratory cell fate in Drosophila oogenesis. PLoS Genet 2023; 19:e1010631. [PMID: 36791149 PMCID: PMC9974129 DOI: 10.1371/journal.pgen.1010631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/28/2023] [Accepted: 01/22/2023] [Indexed: 02/16/2023] Open
Abstract
Specification of migratory cell fate from a stationary population is complex and indispensable both for metazoan development as well for the progression of the pathological condition like tumor metastasis. Though this cell fate transformation is widely prevalent, the molecular understanding of this phenomenon remains largely elusive. We have employed the model of border cells (BC) in Drosophila oogenesis and identified germline activity of an RNA binding protein, Cup that limits acquisition of migratory cell fate from the neighbouring follicle epithelial cells. As activation of JAK-STAT in the follicle cells is critical for BC specification, our data suggest that Cup, non-cell autonomously restricts the domain of JAK-STAT by activating Notch in the follicle cells. Employing genetics and Delta endocytosis assay, we demonstrate that Cup regulates Delta recycling in the nurse cells through Rab11GTPase thus facilitating Notch activation in the adjacent follicle cells. Since Notch and JAK-STAT are antagonistic, we propose that germline Cup functions through Notch and JAK-STAT to modulate BC fate specification from their static epithelial progenitors.
Collapse
Affiliation(s)
- Banhisikha Saha
- Department of Biological Sciences Indian Institute of Science Education & Research- Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Maryland, United States of America
| | - Sayan Acharjee
- Department of Biological Sciences Indian Institute of Science Education & Research- Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
| | - Gaurab Ghosh
- Department of Biological Sciences Indian Institute of Science Education & Research- Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
| | - Purbasa Dasgupta
- Department of Biological Sciences Indian Institute of Science Education & Research- Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
| | - Mohit Prasad
- Department of Biological Sciences Indian Institute of Science Education & Research- Kolkata Mohanpur Campus Mohanpur, Nadia, West Bengal, India
| |
Collapse
|
23
|
Diegmiller R, Imran Alsous J, Li D, Yamashita YM, Shvartsman SY. Fusome topology and inheritance during insect gametogenesis. PLoS Comput Biol 2023; 19:e1010875. [PMID: 36821548 PMCID: PMC9949678 DOI: 10.1371/journal.pcbi.1010875] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/16/2023] [Indexed: 02/24/2023] Open
Abstract
From insects to mammals, oocytes and sperm develop within germline cysts comprising cells connected by intercellular bridges (ICBs). In numerous insects, formation of the cyst is accompanied by growth of the fusome-a membranous organelle that permeates the cyst. Fusome composition and function are best understood in Drosophila melanogaster: during oogenesis, the fusome dictates cyst topology and size and facilitates oocyte selection, while during spermatogenesis, the fusome synchronizes the cyst's response to DNA damage. Despite its distinct and sex-specific roles during insect gametogenesis, elucidating fusome growth and inheritance in females and its structure and connectivity in males has remained challenging. Here, we take advantage of advances in three-dimensional (3D) confocal microscopy and computational image processing tools to reconstruct the topology, growth, and distribution of the fusome in both sexes. In females, our experimental findings inform a theoretical model for fusome assembly and inheritance and suggest that oocyte selection proceeds through an 'equivalency with a bias' mechanism. In males, we find that cell divisions can deviate from the maximally branched pattern observed in females, leading to greater topological variability. Our work consolidates existing disjointed experimental observations and contributes a readily generalizable computational approach for quantitative studies of gametogenesis within and across species.
Collapse
Affiliation(s)
- Rocky Diegmiller
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
| | - Jasmin Imran Alsous
- Flatiron Institute, Simons Foundation, New York, New York, United States of America
| | - Duojia Li
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Yukiko M. Yamashita
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute, Cambridge, Massachusetts, United States of America
| | - Stanislav Y. Shvartsman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Flatiron Institute, Simons Foundation, New York, New York, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| |
Collapse
|
24
|
Abstract
By the time a Drosophila egg is laid, both major body axes have already been defined and it contains all the nutrients needed to develop into a free-living larva in 24 h. By contrast, it takes almost a week to make an egg from a female germline stem cell, during the complex process of oogenesis. This review will discuss key symmetry-breaking steps in Drosophila oogenesis that lead to the polarisation of both body axes: the asymmetric divisions of the germline stem cells; the selection of the oocyte from the 16-cell germline cyst; the positioning of the oocyte at the posterior of the cyst; Gurken signalling from the oocyte to polarise the anterior-posterior axis of the somatic follicle cell epithelium around the developing germline cyst; the signalling back from the posterior follicle cells to polarise the anterior-posterior axis of the oocyte; and the migration of the oocyte nucleus that specifies the dorsal-ventral axis. Since each event creates the preconditions for the next, I will focus on the mechanisms that drive these symmetry-breaking steps, how they are linked and the outstanding questions that remain to be answered.
Collapse
|
25
|
Koury S. Mitotic exchange in female germline stem cells is the major source of Sex Ratio chromosome recombination in Drosophila pseudoobscura. G3 (BETHESDA, MD.) 2022; 12:jkac264. [PMID: 36194019 PMCID: PMC9713450 DOI: 10.1093/g3journal/jkac264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022]
Abstract
Sex Ratio chromosomes in Drosophila pseudoobscura are selfish X chromosome variants associated with 3 nonoverlapping inversions. In the male germline, Sex Ratio chromosomes distort the segregation of X and Y chromosomes (99:1), thereby skewing progeny sex ratio. In the female germline, segregation of Sex Ratio chromosomes is mendelian (50:50), but nonoverlapping inversions strongly suppress recombination establishing a 26-Mb haplotype (constituting ∼20% of the haploid genome). Rare crossover events located between nonoverlapping inversions can disrupt this haplotype, and recombinants have sometimes been found in natural populations. We recently reported on the first lab-generated Sex Ratio recombinants occurring at a rate of 0.0012 crossovers per female meiosis. An improved experimental design presented here reveals that these recombination events were at least 4 times more frequent than previously estimated. Furthermore, recombination events were strongly clustered, indicating that the majority arose from mitotic exchange in female germline stem cells and not from meiotic crossing-over in primary oocytes. Finally, asymmetric recovery of complementary recombinants was consistent with unequal exchange causing the recombination-induced viability defects. Incorporating these experimental results into population models for Sex Ratio chromosome evolution provided a substantially better fit to natural population frequencies and allowed maintenance of the highly differentiated 26-Mb Sex Ratio haplotype without invoking strong epistatic selection. This study provides the first estimate of spontaneous mitotic exchange for naturally occurring chromosomes in Drosophila female germline stem cells, reveals a much higher Sex Ratio chromosome recombination rate, and develops a mathematical model that accurately predicts the rarity of recombinant Sex Ratio chromosomes in natural populations.
Collapse
Affiliation(s)
- Spencer Koury
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
26
|
Riparbelli MG, Persico V, Callaini G. Cell-to-Cell Interactions during Early Drosophila Oogenesis: An Ultrastructural Analysis. Cells 2022; 11:cells11172658. [PMID: 36078066 PMCID: PMC9454453 DOI: 10.3390/cells11172658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/27/2022] Open
Abstract
Drosophila oogenesis requires the subsequent growth of distinct egg chambers each containing a group of sixteen germline cells surrounded by a simple epithelium of follicle cells. The oocyte occupies a posterior position within the germ cells, thus giving a distinct asymmetry to the egg chamber. Although this disposition is critical for the formation of the anterior–posterior axis of the embryo, the interplay between somatic and germ cells during the early stages of oogenesis remains an open question. We uncover by stage 2, when the egg chambers leaved the germarium, some unique spatial interactions between the posterior follicle cells and the oocyte. These interactions are restricted to the surface of the oocyte over the centriole cluster that formed during early oogenesis. Moreover, the posterior follicle cells in front of the oocyte display a convoluted apical membrane with extensive contacts, whereas the other follicle cells have a flat apical surface without obvious surface protrusions. In addition, the germ cells located at the posterior end of the egg chamber have very elongated protrusions that come into contact with each other or with facing follicle cells. These observations point to distinct polarization events during early oogenesis supporting previous molecular data of an inherent asymmetry between the anterior and the posterior regions of the egg chambers.
Collapse
|
27
|
Diegmiller R, Nunley H, Shvartsman SY, Imran Alsous J. Quantitative models for building and growing fated small cell networks. Interface Focus 2022; 12:20210082. [PMID: 35865502 PMCID: PMC9184967 DOI: 10.1098/rsfs.2021.0082] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
Small cell clusters exhibit numerous phenomena typically associated with complex systems, such as division of labour and programmed cell death. A conserved class of such clusters occurs during oogenesis in the form of germline cysts that give rise to oocytes. Germline cysts form through cell divisions with incomplete cytokinesis, leaving cells intimately connected through intercellular bridges that facilitate cyst generation, cell fate determination and collective growth dynamics. Using the well-characterized Drosophila melanogaster female germline cyst as a foundation, we present mathematical models rooted in the dynamics of cell cycle proteins and their interactions to explain the generation of germline cell lineage trees (CLTs) and highlight the diversity of observed CLT sizes and topologies across species. We analyse competing models of symmetry breaking in CLTs to rationalize the observed dynamics and robustness of oocyte fate specification, and highlight remaining gaps in knowledge. We also explore how CLT topology affects cell cycle dynamics and synchronization and highlight mechanisms of intercellular coupling that underlie the observed collective growth patterns during oogenesis. Throughout, we point to similarities across organisms that warrant further investigation and comment on the extent to which experimental and theoretical findings made in model systems extend to other species.
Collapse
Affiliation(s)
- Rocky Diegmiller
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Hayden Nunley
- Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Stanislav Y. Shvartsman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Flatiron Institute, Simons Foundation, New York, NY, USA
| | | |
Collapse
|
28
|
Mathieu J, Michel-Hissier P, Boucherit V, Huynh JR. The deubiquitinase USP8 targets ESCRT-III to promote incomplete cell division. Science 2022; 376:818-823. [PMID: 35587967 DOI: 10.1126/science.abg2653] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In many vertebrate and invertebrate organisms, gametes develop within groups of interconnected cells called germline cysts formed by several rounds of incomplete divisions. We found that loss of the deubiquitinase USP8 gene in Drosophila can transform incomplete divisions of germline cells into complete divisions. Conversely, overexpression of USP8 in germline stem cells is sufficient for the reverse transformation from complete to incomplete cytokinesis. The ESCRT-III proteins CHMP2B and Shrub/CHMP4 are targets of USP8 deubiquitinating activity. In Usp8 mutant sister cells, ectopic recruitment of ESCRT proteins at intercellular bridges causes cysts to break apart. A Shrub/CHMP4 variant that cannot be ubiquitinated does not localize at abscission bridges and cannot complete abscission. Our results uncover ubiquitination of ESCRT-III as a major switch between two types of cell division.
Collapse
Affiliation(s)
- Juliette Mathieu
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS, Inserm, Paris, France
| | - Pascale Michel-Hissier
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS, Inserm, Paris, France
| | - Virginie Boucherit
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS, Inserm, Paris, France
| | - Jean-René Huynh
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS, Inserm, Paris, France
| |
Collapse
|
29
|
Niu W, Spradling AC. Mouse oocytes develop in cysts with the help of nurse cells. Cell 2022; 185:2576-2590.e12. [PMID: 35623357 DOI: 10.1016/j.cell.2022.05.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/07/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Mouse germline cysts, on average, develop into six oocytes supported by 24 nurse cells that transfer cytoplasm and organelles to generate a Balbiani body. We showed that between E14.5 and P5, cysts periodically activate some nurse cells to begin cytoplasmic transfer, which causes them to shrink and turnover within 2 days. Nurse cells die by a programmed cell death (PCD) pathway involving acidification, similar to Drosophila nurse cells, and only infrequently by apoptosis. Prior to initiating transfer, nurse cells co-cluster by scRNA-seq with their pro-oocyte sisters, but during their final 2 days, they cluster separately. The genes promoting oocyte development and nurse cell PCD are upregulated, whereas the genes that repress transfer, such as Tex14, and oocyte factors, such as Nobox and Lhx8, are under-expressed. The transferred nurse cell centrosomes build a cytocentrum that establishes a large microtubule aster in the primordial oocyte that organizes the Balbiani body, defining the earliest oocyte polarity.
Collapse
Affiliation(s)
- Wanbao Niu
- Howard Hughes Medical Institute Research Laboratories, Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
| | - Allan C Spradling
- Howard Hughes Medical Institute Research Laboratories, Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA.
| |
Collapse
|
30
|
Martin ET, Sarkar K, McCarthy A, Rangan P. Oo-site: A dashboard to visualize gene expression during Drosophila oogenesis suggests meiotic entry is regulated post-transcriptionally. Biol Open 2022; 11:bio059286. [PMID: 35579517 PMCID: PMC9148541 DOI: 10.1242/bio.059286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
Determining how stem cell differentiation is controlled has important implications for understanding the etiology of degenerative disease and designing regenerative therapies. In vivo analyses of stem cell model systems have revealed regulatory paradigms for stem cell self-renewal and differentiation. The germarium of the female Drosophila gonad, which houses both germline and somatic stem cells, is one such model system. Bulk mRNA sequencing (RNA-seq), single-cell RNA-seq (scRNA-seq), and bulk translation efficiency (polysome-seq) of mRNAs are available for stem cells and their differentiating progeny within the Drosophila germarium. However, visualizing those data is hampered by the lack of a tool to spatially map gene expression and translational data in the germarium. Here, we have developed Oo-site (https://www.ranganlab.com/Oo-site), a tool for visualizing bulk RNA-seq, scRNA-seq, and translational efficiency data during different stages of germline differentiation, which makes these data accessible to non-bioinformaticians. Using this tool, we recapitulated previously reported expression patterns of developmentally regulated genes and discovered that meiotic genes, such as those that regulate the synaptonemal complex, are regulated at the level of translation.
Collapse
Affiliation(s)
- Elliot T. Martin
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12202, USA
| | - Kahini Sarkar
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12202, USA
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Alicia McCarthy
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12202, USA
| | - Prashanth Rangan
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY 12202, USA
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
31
|
Milas A, Telley IA. Polarity Events in the Drosophila melanogaster Oocyte. Front Cell Dev Biol 2022; 10:895876. [PMID: 35602591 PMCID: PMC9117655 DOI: 10.3389/fcell.2022.895876] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Cell polarity is a pre-requirement for many fundamental processes in animal cells, such as asymmetric cell division, axon specification, morphogenesis and epithelial tissue formation. For all these different processes, polarization is established by the same set of proteins, called partitioning defective (Par) proteins. During development in Drosophila melanogaster, decision making on the cellular and organism level is achieved with temporally controlled cell polarization events. The initial polarization of Par proteins occurs as early as in the germline cyst, when one of the 16 cells becomes the oocyte. Another marked event occurs when the anterior–posterior axis of the future organism is defined by Par redistribution in the oocyte, requiring external signaling from somatic cells. Here, we review the current literature on cell polarity events that constitute the oogenesis from the stem cell to the mature egg.
Collapse
Affiliation(s)
- Ana Milas
- *Correspondence: Ana Milas, ; Ivo A. Telley,
| | | |
Collapse
|
32
|
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]
|
33
|
Urbisz AZ, Chajec Ł, Małota K, Student S, Sawadro MK, Śliwińska MA, Świątek P. All for one - changes in mitochondrial morphology and activity during syncytial oogenesis. Biol Reprod 2022; 106:1232-1253. [PMID: 35156116 DOI: 10.1093/biolre/ioac035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/23/2021] [Accepted: 02/10/2022] [Indexed: 11/14/2022] Open
Abstract
The syncytial groups of germ cells (germ-line cysts) forming in ovaries of clitellate annelids are an attractive model to study mitochondrial stage-specific changes. Using transmission electron microscopy, serial block-face scanning electron microscopy, and fluorescent microscopy, we analyzed the mitochondria distribution and morphology and the state of membrane potential in female cysts in Enchytraeus albidus. We visualized in 3D at the ultrastructural level mitochondria in cysts at successive stages: 2-celled, 4-celled, 16-celled cysts, and cyst in advanced oogenesis. We found that mitochondria form extensive aggregates - they are fused and connected into large and branched mitochondrial networks. The most extensive networks are formed with up to 10,000 fused mitochondria, whereas individual organelles represent up to 2% of the total mitochondrial volume. We classify such morphology of mitochondria as a dynamic hyperfusion state, and suggest that it can maintain their high activity and intensifies the process of cellular respiration within the syncytial cysts. We found some individual mitochondria undergoing degradation, which implies that damaged mitochondria are removed from networks for their final elimination. As it was shown that growing oocytes possess less active mitochondria than the nurse cells, it suggests that the high activity of mitochondria in the nurse cells and their dynamic hyperfusion state serve the needs of the growing oocyte. Additionally, we measured by calorimetry the total antioxidant capacity of germ-line cysts in comparison to somatic tissue, and it suggests that antioxidative defense systems, together with mitochondrial networks, can effectively protect germ-line mitochondria from damage.
Collapse
Affiliation(s)
- Anna Z Urbisz
- Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Łukasz Chajec
- Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Karol Małota
- Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Sebastian Student
- Institute of Automatic Control, Silesian University of Technology, Gliwice, Poland
| | - Marta K Sawadro
- Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Małgorzata A Śliwińska
- Nencki Institute of Experimental Biology of Polish Academy of Sciences, Laboratory of Imaging Tissue Structure and Function, Warsaw, Poland
| | - Piotr Świątek
- Faculty of Natural Sciences, Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
34
|
Świątek P, Rodriguez P, Małota K, Urbisz AZ. Ovary micromorphology and oogenesis in a rhyacodriline oligochaete (Clitellata: Naididae, Rhyacodrilinae). J Morphol 2022; 283:605-617. [PMID: 35150164 DOI: 10.1002/jmor.21461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 11/07/2022]
Abstract
The main goal of the paper is to describe the ovary organization and oogenesis in Peristodrilus montanus, an aquatic oligochaete of the subfamily Rhyacodrilinae. The presented analysis will not only enrich the knowledge about how eggs are formed but, because of the suggested conservatism of ovary organization in clitellate annelids, can contribute to disentangling the complex phylogenetic relationships of the rhyacodrilines within Naididae. The paired, conically shaped ovaries are located in segment XI. They are composed of a dozen or so syncytial germ-line cysts, which are associated with somatic cells. Each germ cell in a cyst has one intercellular bridge that joins it to a central and anuclear cytoplasmic mass, the cytophore. This pattern of cyst organization is typical for all clitellates that have been studied to date. Initially, the germ cells in a cyst undergo a synchronous development, however, there is no synchrony between cysts, and therefore there is a developmental gradient (oogonia, pre-diplotene germ cells, germ cells in diplotene) of oogenesis along the long ovary axis. The cysts are composed of a maximum of 32 cells. Cysts with cells in diplotene detach from the ovaries and the extraovarian phase of oogenesis begins. The developmental synchrony is lost, one cell (an oocyte) per cyst starts to gather cell components and yolk and grows considerably. The remaining cells grow to some extent and function as nurse cells. Like in other microdriles, P. montanus oocytes are rich in yolk; other features of oogenesis are also similar to those that are known from other microdrile taxa. The system of ovary organization found in the studied species is broadly similar to the corresponding features known from Naidinae and Phreodrilidae and, to some extent, in Enchytraeidae. However, this system is different from the one that is known in Tubificinae, Limnodriloidinae and Branchiurinae. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Piotr Świątek
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Bankowa 9, Katowice, Poland
| | - Pilar Rodriguez
- Department of Zoology and Animal Cell Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Box 644, Bilbao, Spain
| | - Karol Małota
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Bankowa 9, Katowice, Poland
| | - Anna Z Urbisz
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Bankowa 9, Katowice, Poland
| |
Collapse
|
35
|
Li Y, Bagheri P, Chang P, Zeng A, Hao J, Fung A, Wu JY, Shi L. Direct Imaging of Lipid Metabolic Changes in Drosophila Ovary During Aging Using DO-SRS Microscopy. FRONTIERS IN AGING 2022; 2:819903. [PMID: 35822015 PMCID: PMC9261447 DOI: 10.3389/fragi.2021.819903] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/29/2021] [Indexed: 01/09/2023]
Abstract
Emerging studies have shown that lipids and proteins play versatile roles in various aspects of aging. High-resolution in situ optical imaging provides a powerful approach to study the metabolic dynamics of lipids and proteins during aging. Here, we integrated D2O probing and stimulated Raman scattering (DO-SRS) microscopy to directly visualize metabolic changes in aging Drosophila ovary. The subcellular spatial distribution of de novo protein synthesis and lipogenesis in ovary was quantitatively imaged and examined. Our Raman spectra showed that early stages follicles were protein-enriched whereas mature eggs were lipid-enriched. DO-SRS imaging showed a higher protein synthesis in the earlier developing stages and an increased lipid turned over at the late stage. Aged (35 days) flies exhibited a dramatic decrease in metabolic turnover activities of both proteins and lipids, particularly, in the germ stem cell niche of germarium. We found an accumulation of unsaturated lipids in the nurse cells and oocytes in old flies, suggesting that unsaturated lipids may play an important role in the processes of oocyte maturation. We further detected changes in mitochondrial morphology and accumulation of Cytochrome c during aging. To our knowledge, this is the first study that directly visualizes spatiotemporal changes in lipid and protein metabolism in Drosophila ovary during development and aging processes. Our study not only demonstrates the application of a new imaging platform in visualizing metabolic dynamics of lipids and proteins in situ but also unravels how the metabolic activity and lipid distribution change in Drosophila ovary during aging.
Collapse
Affiliation(s)
- Yajuan Li
- The Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Pegah Bagheri
- The Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Phyllis Chang
- The Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Audrey Zeng
- The Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Jie Hao
- The Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Anthony Fung
- The Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Jane Y. Wu
- Department of Neurology, Northwestern University, Chicago, IL, United States
| | - Lingyan Shi
- The Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
- *Correspondence: Lingyan Shi,
| |
Collapse
|
36
|
Abstract
In vitro systems capable of reconstituting the process of mouse oogenesis are now being established to help develop further understanding of the mechanisms underlying oocyte/follicle development and differentiation. These systems could also help increase the production of useful livestock or genetically modified animals, and aid in identifying the causes of infertility in humans. Recently, we revealed, using an in vitro system for recapitulating oogenesis, that the activation of the estrogen signaling pathway induces abnormal follicle formation, that blocking estrogen-induced expression of anti-Müllerian hormone is crucial for normal follicle formation, and that the production of α-fetoprotein in fetal liver tissue is involved in normal in vivo follicle formation. In mouse fetuses, follicle formation is not carried out by factors within the ovaries but is instead orchestrated by distal endocrine factors. This review outlines findings from genetics, endocrinology, and in vitro studies regarding the factors that can affect the formation of primordial follicles in mammals.
Collapse
|
37
|
Abstract
In mammals and flies, only one cell in a multicellular female germline cyst becomes an oocyte, but how symmetry is broken to select the oocyte is unknown. Here, we show that the microtubule (MT) minus end-stabilizing protein Patronin/CAMSAP marks the future Drosophila oocyte and is required for oocyte specification. The spectraplakin Shot recruits Patronin to the fusome, a branched structure extending into all cyst cells. Patronin stabilizes more MTs in the cell with the most fusome material. Our data suggest that this weak asymmetry is amplified by Dynein-dependent transport of Patronin-stabilized MTs. This forms a polarized MT network, along which Dynein transports oocyte determinants into the presumptive oocyte. Thus, Patronin amplifies a weak fusome anisotropy to break symmetry and select one cell to become the oocyte.
Collapse
Affiliation(s)
- D. Nashchekin
- The Gurdon Institute and the Department of Genetics, University of Cambridge; Tennis Court Road, Cambridge CB2 1QN, United Kingdom,Corresponding author. ,
| | - L. Busby
- The Gurdon Institute and the Department of Genetics, University of Cambridge; Tennis Court Road, Cambridge CB2 1QN, United Kingdom
| | - M. Jakobs
- The Department of Physiology, Development and Neuroscience, University of Cambridge; Cambridge CB2 3DY, United Kingdom
| | - I. Squires
- The Gurdon Institute and the Department of Genetics, University of Cambridge; Tennis Court Road, Cambridge CB2 1QN, United Kingdom
| | - D. Johnston
- The Gurdon Institute and the Department of Genetics, University of Cambridge; Tennis Court Road, Cambridge CB2 1QN, United Kingdom,Corresponding author. ,
| |
Collapse
|
38
|
Early Drosophila Oogenesis: A Tale of Centriolar Asymmetry. Cells 2021; 10:cells10081997. [PMID: 34440763 PMCID: PMC8391878 DOI: 10.3390/cells10081997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 11/17/2022] Open
Abstract
Among the morphological processes that characterize the early stages of Drosophila oogenesis, the dynamic of the centrioles deserves particular attention. We re-examined the architecture and the distribution of the centrioles within the germarium and early stages of the vitellarium. We found that most of the germ cell centrioles diverge from the canonical model and display notable variations in size. Moreover, duplication events were frequently observed within the germarium in the absence of DNA replication. Finally, we report the presence of an unusually long centriole that is first detected in the cystoblast and is always associated with the developing oocyte. This centriole is directly inherited after the asymmetric division of the germline stem cells and persists during the process of oocyte selection, thus already representing a marker for oocyte identification at the beginning of its formation and during the ensuing developmental stages.
Collapse
|
39
|
Blatt P, Wong-Deyrup SW, McCarthy A, Breznak S, Hurton MD, Upadhyay M, Bennink B, Camacho J, Lee MT, Rangan P. RNA degradation is required for the germ-cell to maternal transition in Drosophila. Curr Biol 2021; 31:2984-2994.e7. [PMID: 33989522 PMCID: PMC8319052 DOI: 10.1016/j.cub.2021.04.052] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 03/26/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
In sexually reproducing animals, the oocyte contributes a large supply of RNAs that are essential to launch development upon fertilization. The mechanisms that regulate the composition of the maternal RNA contribution during oogenesis are unclear. Here, we show that a subset of RNAs expressed during the early stages of oogenesis is subjected to regulated degradation during oocyte specification. Failure to remove these RNAs results in oocyte dysfunction and death. We identify the RNA-degrading Super Killer complex and No-Go Decay factor Pelota as key regulators of oogenesis via targeted degradation of specific RNAs expressed in undifferentiated germ cells. These regulators target RNAs enriched for cytidine sequences that are bound by the polypyrimidine tract binding protein Half pint. Thus, RNA degradation helps orchestrate a germ cell-to-maternal transition that gives rise to the maternal contribution to the zygote.
Collapse
Affiliation(s)
- Patrick Blatt
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222
| | - Siu Wah Wong-Deyrup
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222
| | - Alicia McCarthy
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222; 10x Genomics, Inc., 6230 Stoneridge Mall Road, Pleasanton, CA, 94588
| | - Shane Breznak
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222
| | - Matthew D Hurton
- University of Pittsburgh, Department of Biological Sciences; 4249 Fifth Avenue, Pittsburgh, PA 15260
| | - Maitreyi Upadhyay
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222; Department of Stem Cell and Regenerative Biology, Sherman Fairchild 100, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138
| | - Benjamin Bennink
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222
| | - Justin Camacho
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222
| | - Miler T Lee
- University of Pittsburgh, Department of Biological Sciences; 4249 Fifth Avenue, Pittsburgh, PA 15260.
| | - Prashanth Rangan
- University at Albany, Department of Biological Sciences, RNA Institute; 1400 Washington Avenue, LSRB 2033D, Albany, NY 12222.
| |
Collapse
|
40
|
Neiswender H, Goldman CH, Veeranan-Karmegam R, Gonsalvez GB. Dynein light chain-dependent dimerization of Egalitarian is essential for maintaining oocyte fate in Drosophila. Dev Biol 2021; 478:76-88. [PMID: 34181915 DOI: 10.1016/j.ydbio.2021.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/02/2021] [Accepted: 06/17/2021] [Indexed: 11/28/2022]
Abstract
Egalitarian (Egl) is an RNA adaptor for the Dynein motor and is thought to link numerous, perhaps hundreds, of mRNAs with Dynein. Dynein, in turn, is responsible for the transport and localization of these mRNAs. Studies have shown that efficient mRNA binding by Egl requires the protein to dimerize. We recently demonstrated that Dynein light chain (Dlc) is responsible for facilitating the dimerization of Egl. Mutations in Egl that fail to interact with Dlc do not dimerize, and as such, are defective for mRNA binding. Consequently, this mutant does not efficiently associate with BicaudalD (BicD), the factor responsible for linking the Egl/mRNA complex with Dynein. In this report, we tested whether artificially dimerizing this Dlc-binding mutant using a leucine zipper would restore mRNA binding and rescue mutant phenotypes in vivo. Interestingly, we found that although artificial dimerization of Egl restored BicD binding, it only partially restored mRNA binding. As a result, Egl-dependent phenotypes, such as oocyte specification and mRNA localization, were only partially rescued. We hypothesize that Dlc-mediated dimerization of Egl results in a three-dimensional conformation of the Egl dimer that is best suited for mRNA binding. Although the leucine zipper restores Egl dimerization, it likely does not enable Egl to assemble into the conformation required for maximal mRNA binding activity.
Collapse
Affiliation(s)
- Hannah Neiswender
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Chandler H Goldman
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Rajalakshmi Veeranan-Karmegam
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Graydon B Gonsalvez
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
41
|
Goldman CH, Neiswender H, Baker F, Veeranan-Karmegam R, Misra S, Gonsalvez GB. Optimal RNA binding by Egalitarian, a Dynein cargo adaptor, is critical for maintaining oocyte fate in Drosophila. RNA Biol 2021; 18:2376-2389. [PMID: 33904382 DOI: 10.1080/15476286.2021.1914422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The Dynein motor is responsible for the localization of numerous mRNAs within Drosophila oocytes and embryos. The RNA binding protein, Egalitarian (Egl), is thought to link these various RNA cargoes with Dynein. Although numerous studies have shown that Egl is able to specifically associate with these RNAs, the nature of these interactions has remained elusive. Egl contains a central RNA binding domain that shares limited homology with an exonuclease, yet Egl binds to RNA without degrading it. Mutations have been identified within Egl that disrupt its association with its protein interaction partners, BicaudalD (BicD) and Dynein light chain (Dlc), but no mutants have been described that are specifically defective for RNA binding. In this report, we identified a series of positively charged residues within Egl that are required for RNA binding. Using corresponding RNA binding mutants, we demonstrate that specific RNA cargoes are more reliant on maximal Egl RNA biding activity for their correct localization in comparison to others. We also demonstrate that specification and maintenance of oocyte fate requires maximal Egl RNA binding activity. Even a subtle reduction in Egl's RNA binding activity completely disrupts this process. Our results show that efficient RNA localization at the earliest stages of oogenesis is required for specification of the oocyte and restriction of meiosis to a single cell.
Collapse
Affiliation(s)
- Chandler H Goldman
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA.,Department of Genetics, Davidson Life Sciences Complex, University of Georgia, Athens, GA, USA
| | - Hannah Neiswender
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Frederick Baker
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Saurav Misra
- Dept. Of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS,USA
| | - Graydon B Gonsalvez
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
42
|
Doherty CA, Diegmiller R, Kapasiawala M, Gavis ER, Shvartsman SY. Coupled oscillators coordinate collective germline growth. Dev Cell 2021; 56:860-870.e8. [PMID: 33689691 PMCID: PMC8265018 DOI: 10.1016/j.devcel.2021.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 12/03/2020] [Accepted: 02/16/2021] [Indexed: 12/20/2022]
Abstract
Developing oocytes need large supplies of macromolecules and organelles. A conserved strategy for accumulating these products is to pool resources of oocyte-associated germline nurse cells. In Drosophila, these cells grow more than 100-fold to boost their biosynthetic capacity. No previously known mechanism explains how nurse cells coordinate growth collectively. Here, we report a cell cycle-regulating mechanism that depends on bidirectional communication between the oocyte and nurse cells, revealing the oocyte as a critical regulator of germline cyst growth. Transcripts encoding the cyclin-dependent kinase inhibitor, Dacapo, are synthesized by the nurse cells and actively localized to the oocyte. Retrograde movement of the oocyte-synthesized Dacapo protein to the nurse cells generates a network of coupled oscillators that controls the cell cycle of the nurse cells to regulate cyst growth. We propose that bidirectional nurse cell-oocyte communication establishes a growth-sensing feedback mechanism that regulates the quantity of maternal resources loaded into the oocyte.
Collapse
Affiliation(s)
- Caroline A Doherty
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Rocky Diegmiller
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540, USA
| | - Manisha Kapasiawala
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540, USA
| | - Elizabeth R Gavis
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA.
| | - Stanislav Y Shvartsman
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540, USA; Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA.
| |
Collapse
|
43
|
Abouward R, Schiavo G. Walking the line: mechanisms underlying directional mRNA transport and localisation in neurons and beyond. Cell Mol Life Sci 2021; 78:2665-2681. [PMID: 33341920 PMCID: PMC8004493 DOI: 10.1007/s00018-020-03724-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/02/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022]
Abstract
Messenger RNA (mRNA) localisation enables a high degree of spatiotemporal control on protein synthesis, which contributes to establishing the asymmetric protein distribution required to set up and maintain cellular polarity. As such, a tight control of mRNA localisation is essential for many biological processes during development and in adulthood, such as body axes determination in Drosophila melanogaster and synaptic plasticity in neurons. The mechanisms controlling how mRNAs are localised, including diffusion and entrapment, local degradation and directed active transport, are largely conserved across evolution and have been under investigation for decades in different biological models. In this review, we will discuss the standing of the field regarding directional mRNA transport in light of the recent discovery that RNA can hitchhike on cytoplasmic organelles, such as endolysosomes, and the impact of these transport modalities on our understanding of neuronal function during development, adulthood and in neurodegeneration.
Collapse
Affiliation(s)
- Reem Abouward
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
44
|
Diegmiller R, Zhang L, Gameiro M, Barr J, Imran Alsous J, Schedl P, Shvartsman SY, Mischaikow K. Mapping parameter spaces of biological switches. PLoS Comput Biol 2021; 17:e1008711. [PMID: 33556054 PMCID: PMC7895388 DOI: 10.1371/journal.pcbi.1008711] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/19/2021] [Accepted: 01/15/2021] [Indexed: 01/02/2023] Open
Abstract
Since the seminal 1961 paper of Monod and Jacob, mathematical models of biomolecular circuits have guided our understanding of cell regulation. Model-based exploration of the functional capabilities of any given circuit requires systematic mapping of multidimensional spaces of model parameters. Despite significant advances in computational dynamical systems approaches, this analysis remains a nontrivial task. Here, we use a nonlinear system of ordinary differential equations to model oocyte selection in Drosophila, a robust symmetry-breaking event that relies on autoregulatory localization of oocyte-specification factors. By applying an algorithmic approach that implements symbolic computation and topological methods, we enumerate all phase portraits of stable steady states in the limit when nonlinear regulatory interactions become discrete switches. Leveraging this initial exact partitioning and further using numerical exploration, we locate parameter regions that are dense in purely asymmetric steady states when the nonlinearities are not infinitely sharp, enabling systematic identification of parameter regions that correspond to robust oocyte selection. This framework can be generalized to map the full parameter spaces in a broad class of models involving biological switches. Identification of qualitatively different regimes in models of biomolecular switches is essential for understanding dynamics of complex biological processes, including symmetry breaking in cells and cell networks. We demonstrate how topological methods, symbolic computation, and numerical simulations can be combined for systematic mapping of symmetry-broken states in a mathematical model of oocyte specification in Drosophila, a leading experimental system of animal oogenesis. Our algorithmic framework reveals global connectedness of parameter domains corresponding to robust oocyte specification and enables systematic navigation through multidimensional parameter spaces in a large class of biomolecular switches.
Collapse
Affiliation(s)
- Rocky Diegmiller
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
| | - Lun Zhang
- Department of Mathematics, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Marcio Gameiro
- Department of Mathematics, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- Instituto de Ciências Matemáticas e de Computação, Universidade de São Paulo, São Carlos, São Paulo, Brazil
| | - Justinn Barr
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Jasmin Imran Alsous
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Stanislav Y. Shvartsman
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, United States of America
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- Flatiron Institute, Simons Foundation, New York, New York, United States of America
- * E-mail: (SYS); (KM)
| | - Konstantin Mischaikow
- Department of Mathematics, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- * E-mail: (SYS); (KM)
| |
Collapse
|
45
|
Rathore OS, Silva RD, Ascensão-Ferreira M, Matos R, Carvalho C, Marques B, Tiago MN, Prudêncio P, Andrade RP, Roignant JY, Barbosa-Morais NL, Martinho RG. NineTeen Complex-subunit Salsa is required for efficient splicing of a subset of introns and dorsal-ventral patterning. RNA (NEW YORK, N.Y.) 2020; 26:1935-1956. [PMID: 32963109 PMCID: PMC7668242 DOI: 10.1261/rna.077446.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/07/2020] [Indexed: 06/11/2023]
Abstract
The NineTeen Complex (NTC), also known as pre-mRNA-processing factor 19 (Prp19) complex, regulates distinct spliceosome conformational changes necessary for splicing. During Drosophila midblastula transition, splicing is particularly sensitive to mutations in NTC-subunit Fandango, which suggests differential requirements of NTC during development. We show that NTC-subunit Salsa, the Drosophila ortholog of human RNA helicase Aquarius, is rate-limiting for splicing of a subset of small first introns during oogenesis, including the first intron of gurken Germline depletion of Salsa and splice site mutations within gurken first intron impair both adult female fertility and oocyte dorsal-ventral patterning, due to an abnormal expression of Gurken. Supporting causality, the fertility and dorsal-ventral patterning defects observed after Salsa depletion could be suppressed by the expression of a gurken construct without its first intron. Altogether, our results suggest that one of the key rate-limiting functions of Salsa during oogenesis is to ensure the correct expression and efficient splicing of the first intron of gurken mRNA. Retention of gurken first intron compromises the function of this gene most likely because it undermines the correct structure and function of the transcript 5'UTR.
Collapse
Affiliation(s)
- Om Singh Rathore
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
| | - Rui D Silva
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
| | - Mariana Ascensão-Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ricardo Matos
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
| | - Célia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Bruno Marques
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
| | - Margarida N Tiago
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
| | - Pedro Prudêncio
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Raquel P Andrade
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
- Department of Medicine and Biomedical Sciences and Algarve Biomedical Center, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Jean-Yves Roignant
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015 Lausanne, Switzerland
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg-University Mainz, 55128 Mainz, Germany
| | - Nuno L Barbosa-Morais
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Rui Gonçalo Martinho
- Center for Biomedical Research (CBMR), Universidade do Algarve, Faro, 8005-139 Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Department of Medical Sciences and Institute for Biomedicine (iBiMED), Universidade de Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
46
|
Abstract
Model organisms are extensively used in research as accessible and convenient systems for studying a particular area or question in biology. Traditionally, only a limited number of organisms have been studied in detail, but modern genomic tools are enabling researchers to extend beyond the set of classical model organisms to include novel species from less-studied phylogenetic groups. This review focuses on model species for an important group of multicellular organisms, the brown algae. The development of genetic and genomic tools for the filamentous brown alga Ectocarpus has led to it emerging as a general model system for this group, but additional models, such as Fucus or Dictyota dichotoma, remain of interest for specific biological questions. In addition, Saccharina japonica has emerged as a model system to directly address applied questions related to algal aquaculture. We discuss the past, present, and future of brown algal model organisms in relation to the opportunities and challenges in brown algal research.
Collapse
Affiliation(s)
- Susana M Coelho
- Laboratory of Integrative Biology of Marine Models (LBI2M), Station Biologique de Roscoff (SBR), CNRS, Sorbonne Université, 29680 Roscoff, France;
- Current affiliation: Department of Algal Development and Evolution, Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany;
| | - J Mark Cock
- Laboratory of Integrative Biology of Marine Models (LBI2M), Station Biologique de Roscoff (SBR), CNRS, Sorbonne Université, 29680 Roscoff, France;
| |
Collapse
|
47
|
Kaufman RS, Price KL, Mannix KM, Ayers KM, Hudson AM, Cooley L. Drosophila sperm development and intercellular cytoplasm sharing through ring canals do not require an intact fusome. Development 2020; 147:dev.190140. [PMID: 33033119 DOI: 10.1242/dev.190140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022]
Abstract
Animal germ cells communicate directly with each other during gametogenesis through intercellular bridges, often called ring canals (RCs), that form as a consequence of incomplete cytokinesis during cell division. Developing germ cells in Drosophila have an additional specialized organelle connecting the cells called the fusome. Ring canals and the fusome are required for fertility in Drosophila females, but little is known about their roles during spermatogenesis. With live imaging, we directly observe the intercellular movement of GFP and a subset of endogenous proteins through RCs during spermatogenesis, from two-cell diploid spermatogonia to clusters of 64 post-meiotic haploid spermatids, demonstrating that RCs are stable and open to intercellular traffic throughout spermatogenesis. Disruption of the fusome, a large cytoplasmic structure that extends through RCs and is important during oogenesis, had no effect on spermatogenesis or male fertility under normal conditions. Our results reveal that male germline RCs allow the sharing of cytoplasmic information that might play a role in quality control surveillance during sperm development.
Collapse
Affiliation(s)
- Ronit S Kaufman
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kari L Price
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Katelynn M Mannix
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kathleen M Ayers
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Andrew M Hudson
- 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 & Developmental Biology, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
48
|
Chanet S, Huynh JR. Collective Cell Sorting Requires Contractile Cortical Waves in Germline Cells. Curr Biol 2020; 30:4213-4226.e4. [PMID: 32916115 DOI: 10.1016/j.cub.2020.08.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 07/01/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022]
Abstract
Encapsulation of germline cells by layers of somatic cells forms the basic unit of female reproduction called primordial follicles in mammals and egg chambers in Drosophila. How germline and somatic tissues are coordinated for the morphogenesis of each separated unit remains poorly understood. Here, using improved live imaging of Drosophila ovaries, we uncovered periodic actomyosin waves at the cortex of germ cells. These contractile waves are associated with pressure release blebs, which project from germ cells into somatic cells. We demonstrate that these cortical activities, together with cadherin-based adhesion, are required to sort each germline cyst as one collective unit. Genetic perturbations of cortical contractility, bleb protrusion, or adhesion between germline and somatic cells induced encapsulation defects resulting from failures to encapsulate any germ cells, or the inclusion of too many germ cells per egg chamber, or even the mechanical split of germline cysts. Live-imaging experiments revealed that reducing contractility or adhesion in the germline reduced the stiffness of germline cysts and their proper anchoring to the somatic cells. Germline cysts can then be squeezed and passively pushed by constricting surrounding somatic cells, resulting in cyst splitting and cyst collisions during encapsulation. Increasing germline cysts activity or blocking somatic cell constriction movements can reveal active forward migration of germline cysts. Our results show that germ cells play an active role in physical coupling with somatic cells to produce the female gamete.
Collapse
Affiliation(s)
- Soline Chanet
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS/UMR 7241 - INSERM U1050, 11 Place Marcelin Berthelot, 75005 Paris, France
| | - Jean-René Huynh
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS/UMR 7241 - INSERM U1050, 11 Place Marcelin Berthelot, 75005 Paris, France.
| |
Collapse
|
49
|
Morphology of Mitochondria in Syncytial Annelid Female Germ-Line Cyst Visualized by Serial Block-Face SEM. Int J Cell Biol 2020; 2020:7483467. [PMID: 32395131 PMCID: PMC7199535 DOI: 10.1155/2020/7483467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/11/2019] [Accepted: 08/04/2019] [Indexed: 11/23/2022] Open
Abstract
Mitochondria change their morphology and distribution depending on the metabolism and functional state of a cell. Here, we analyzed the mitochondria and selected structures in female germ-line cysts in a representative of clitellate annelids – the white worm Enchytraeus albidus in which each germ cell has one cytoplasmic bridge that connects it to a common cytoplasmic mass. Using serial block-face scanning electron microscopy (SBEM), we prepared three-dimensional ultrastructural reconstructions of the entire selected compartments of a cyst at the advanced stage of oogenesis, i.e. the nurse cell, cytophore, and cytoplasmic bridges of all 16 cells (15 nurse cells and oocyte). We revealed extensive mitochondrial networks in the nurse cells, cytophore and mitochondria that pass through the cytoplasmic bridges, which indicates that a mitochondrial network can extend throughout the entire cyst. The dynamic hyperfusion state was suggested for such mitochondrial aggregations. We measured the mitochondria distribution and revealed their polarized distribution in the nurse cells and more abundant accumulation within the cytophore compared to the nurse cell. A close association of mitochondrial networks with dispersed nuage material, which seems to be the structural equivalent of a Balbiani body, not described in clitellate annelids so far, was also revealed.
Collapse
|
50
|
Transposon Reactivation in the Germline May Be Useful for Both Transposons and Their Host Genomes. Cells 2020; 9:cells9051172. [PMID: 32397241 PMCID: PMC7290860 DOI: 10.3390/cells9051172] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/29/2022] Open
Abstract
Transposable elements (TEs) are long-term residents of eukaryotic genomes that make up a large portion of these genomes. They can be considered as perfectly fine members of genomes replicating with resident genes and being transmitted vertically to the next generation. However, unlike regular genes, TEs have the ability to send new copies to new sites. As such, they have been considered as parasitic members ensuring their own replication. In another view, TEs may also be considered as symbiotic sequences providing shared benefits after mutualistic interactions with their host genome. In this review, we recall the relationship between TEs and their host genome and discuss why transient relaxation of TE silencing within specific developmental windows may be useful for both.
Collapse
|