1
|
Harris DE, Kim JJ, Stern SR, Vicars HM, Matias NR, Gallicchio L, Baker CC, Fuller MT. An RNA binding regulatory cascade controls the switch from proliferation to differentiation in the Drosophila male germ line stem cell lineage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611673. [PMID: 39282418 PMCID: PMC11398533 DOI: 10.1101/2024.09.06.611673] [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: 09/21/2024]
Abstract
The switch from precursor cell proliferation to onset of differentiation in adult stem cell lineages must be carefully regulated to produce sufficient progeny to maintain and repair tissues, yet prevent overproliferation that may enable oncogenesis. In the Drosophila male germ cell lineage, spermatogonia produced by germ line stem cells undergo a limited number of transit amplifying mitotic divisions before switching to the spermatocyte program that sets up meiosis and eventual spermatid differentiation. The number of transit amplifying divisions is set by accumulation of the bag-of-marbles (Bam) protein to a critical threshold. In bam mutants, spermatogonia proliferate through several extra rounds of mitosis then die without becoming spermatocytes. Here we show that the key role of Bam for the mitosis to differentiation switch is repressing expression of Held Out Wings (how), homolog of mammalian Quaking. Knock down of how in germ cells was sufficient to allow spermatogonia mutant for bam or its partner benign gonial cell neoplasm (bgcn) to differentiate, while forced expression of nuclear-targeted How protein in spermatogonia wild-type for bam resulted in continued proliferation at the expense of differentiation. Our findings suggest that Bam targets how RNA for degradation by acting as an adapter to recruit the CCR4-NOT deadenylation complex via binding its subunit, Caf40. As How is itself an RNA binding protein with roles in RNA processing, our findings reveal that the switch from proliferation to meiosis and differentiation in the Drosophila male germ line adult stem cell lineage is regulated by a cascade of RNA-binding proteins.
Collapse
Affiliation(s)
- Devon E. Harris
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jongmin J. Kim
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Current address: Department of Biomedical Sciences, Cornell University, Ithaca NY, 14853, USA
| | - Sarah R. Stern
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannah M. Vicars
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Neuza R. Matias
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lorenzo Gallicchio
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Catherine C. Baker
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Margaret T. Fuller
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
2
|
Mahadevaraju S, Pal S, Bhaskar P, McDonald BD, Benner L, Denti L, Cozzi D, Bonizzoni P, Przytycka TM, Oliver B. Diverse somatic Transformer and sex chromosome karyotype pathways regulate gene expression in Drosophila gonad development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607556. [PMID: 39372789 PMCID: PMC11451611 DOI: 10.1101/2024.08.12.607556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The somatic sex determination gene transformer (tra) is required for the highly sexually dimorphic development of most somatic cells, including those of the gonads. In addition, somatic tra is required for the germline development even though it is not required for sex determination within germ cells. Germ cell autonomous gene expression is also necessary for their sex determination. To understand the interplay between these signals, we compared the phenotype and gene expression of larval wild-type gonads and the sex-transformed tra gonads. XX larval ovaries transformed into testes were dramatically smaller than wild-type, with significant reductions in germ cell number, likely due to altered geometry of the stem cell niche. Additionally, there was a defect in progression into spermatocyte stages. XY larval testes transformed into ovaries had excessive germ cells, possibly due to the earlier onset of cell division. We suggest that germ cells are neither fully female nor male following somatic sex transformation, with certain pathways characteristic of each sex expressed in tra mutants. We found multiple patterns of somatic and germline gene expression control exclusively due to tra, exclusively due to sex chromosome karyotype, but usually due to a combination of these factors showing tra and sex chromosome karyotype pathways regulate gene expression during Drosophila gonad development.
Collapse
Affiliation(s)
- Sharvani Mahadevaraju
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Biology. St. Mary’s College of Maryland, St. Mary’s City, Maryland, USA
| | - Soumitra Pal
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
- Neurobiology Neurodegeneration and Repair Lab, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Pradeep Bhaskar
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Brennan D. McDonald
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Biology, Stanford University, Stanford, California, USA
| | - Leif Benner
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Luca Denti
- Department of Informatics, Systems, and Communication, University of Milano - Bicocca, Milan, Italy
| | - Davide Cozzi
- Department of Informatics, Systems, and Communication, University of Milano - Bicocca, Milan, Italy
| | - Paola Bonizzoni
- Department of Informatics, Systems, and Communication, University of Milano - Bicocca, Milan, Italy
| | - Teresa M. Przytycka
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian Oliver
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
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: 5.0] [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
|
4
|
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: 0] [Impact Index Per Article: 0] [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
|
5
|
Rosales-Nieves AE, Marín-Menguiano M, Campoy-Lopez A, González-Reyes A. Visualization and Quantification of Drosophila Larval Ovaries. Methods Mol Biol 2023; 2626:37-47. [PMID: 36715898 DOI: 10.1007/978-1-0716-2970-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The morphogenesis of the ovarian germline stem cell (GSC) niche during larval stages in Drosophila provides the initial cellular and molecular basis for female gamete production in the adult. During larval instars, the Drosophila female gonad matures gradually from a round structure enclosing primordial germ cells (PGCs) and somatic cells into a functional organ containing GSC populations in their niches that later in adult stages support oogenesis. In this chapter, we describe a technique for dissecting, staining, and analyzing gonads from female Drosophila larvae and early pupae, offering the possibility of a direct view of the morphogenesis of an ovarian niche.
Collapse
Affiliation(s)
- Alicia E Rosales-Nieves
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain.
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
| | - Miriam Marín-Menguiano
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain
| | - Alejandro Campoy-Lopez
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain
| | - Acaimo González-Reyes
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Sevilla, Spain.
| |
Collapse
|
6
|
Khalid MZ, Sun Z, Zhang J, Zhang S, Zhong G. Cyromazine affects the ovarian germ cells of Drosophila via the ecdysone signaling pathway. Front Physiol 2022; 13:992306. [PMID: 36246127 PMCID: PMC9557234 DOI: 10.3389/fphys.2022.992306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cyromazine, an insect growth regulator, has been extensively used against the insect pests of livestock and households. Previously, it was observed that the continuous selection of cyromazine from the larval to the adult stage decreased the number of germline stem cells (GSCs) and cystoblasts (CBs) in the adult ovary. In addition, in this study, we observed that the number of primordial germ cells (PGCs) was also decreased in the larval ovary after treatment with cyromazine. However, the mechanism by which it affects the germ cells is yet to be explored. Consequently, to deeply investigate the effects of cyromazine on the germ cells, we performed tissue-specific RNA sequencing. Bioinformatics analysis revealed that the ecdysone signaling pathway was significantly influenced under cyromazine stress. Based on that, we screened and selected 14 ecdysone signaling responsive genes and silenced their expression in the germ cells only. Results of that showed a considerable reduction in the number of germ cells. Furthermore, we mixed exogenous 20E with the cyromazine-containing diet to rescue the ecdysone signaling. Our results supported that the application of exogenous 20E significantly rescued the germ cells in the transgenic lines. Therefore, this implies that the cyromazine decreased the number of germ cells by affecting the ecdysone signaling pathway.
Collapse
|
7
|
Szarka-Kovács AB, Takács Z, Bence M, Erdélyi M, Jankovics F. Drosophila MESR4 Gene Ensures Germline Stem Cell Differentiation by Promoting the Transcription of bag of marbles. Cells 2022; 11:cells11132056. [PMID: 35805140 PMCID: PMC9265997 DOI: 10.3390/cells11132056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/02/2022] Open
Abstract
Ovarian germline stem cells (GSCs) of Drosophila melanogaster provide a valuable in vivo model to investigate how the adult stem cell identity is maintained and the differentiation of the daughter cells is regulated. GSCs are embedded into a specialized cellular microenvironment, the so-called stem cell niche. Besides the complex signaling interactions between the germ cells and the niche cells, the germ cell intrinsic mechanisms, such as chromatin regulation and transcriptional control, are also crucial in the decision about self-renewal and differentiation. The key differentiation regulator gene is the bag of marbles (bam), which is transcriptionally repressed in the GSCs and de-repressed in the differentiating daughter cell. Here, we show that the transcription factor MESR4 functions in the germline to promote GSC daughter differentiation. We find that the loss of MESR4 results in the accumulation of GSC daughter cells which fail to transit from the pre-cystoblast (pre-CB) to the differentiated cystoblast (CB) stage. The forced expression of bam can rescue this differentiation defect. By a series of epistasis experiments and a transcriptional analysis, we demonstrate that MESR4 positively regulates the transcription of bam. Our results suggest that lack of repression alone is not sufficient, but MESR4-mediated transcriptional activation is also required for bam expression.
Collapse
Affiliation(s)
- Alexandra Brigitta Szarka-Kovács
- Institute of Genetics, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.B.S.-K.); (Z.T.); (M.B.)
- Doctoral School in Biology, University of Szeged, H-6720 Szeged, Hungary
| | - Zsanett Takács
- Institute of Genetics, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.B.S.-K.); (Z.T.); (M.B.)
| | - Melinda Bence
- Institute of Genetics, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.B.S.-K.); (Z.T.); (M.B.)
| | - Miklós Erdélyi
- Institute of Genetics, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.B.S.-K.); (Z.T.); (M.B.)
- Correspondence: (M.E.); (F.J.)
| | - Ferenc Jankovics
- Institute of Genetics, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary; (A.B.S.-K.); (Z.T.); (M.B.)
- Department of Medical Biology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- Correspondence: (M.E.); (F.J.)
| |
Collapse
|
8
|
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: 2] [Impact Index Per Article: 1.0] [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
|
9
|
Durkina VB, Chapman JW, Demchenko NL. First observations of ovary regeneration in an amphipod, Ampelisca eschrichtii Krøyer, 1842. PeerJ 2022; 10:e12950. [PMID: 35291485 PMCID: PMC8918206 DOI: 10.7717/peerj.12950] [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: 08/27/2021] [Accepted: 01/26/2022] [Indexed: 01/11/2023] Open
Abstract
Background Females of the gammaridean amphipod Ampelisca eschrichtii with signs of regenerating, previously atrophied ovaries were recovered from the northeastern shelf of Sakhalin Island (Okhotsk Sea, Russia). Ovarian regeneration was previously unknown for any amphipod species. A. eschrichtii have a predominantly 2-year life cycle (from embryo to adult death) and reproduce once between late winter or early spring at the age of 2 years. Occasionally, females survive to a third year. An adaptive value of extended survival among these females is likely to require that they are also reproductive. Methods Histological sections from a second-year female with ovarian atrophy, a female with normal ovaries, a third-year female with ovarian regeneration, as well as testes of an immature and a sexually mature male were compared to determine the sources of cells of the germinal and somatic lines necessary for ovarian regeneration. Results Ovarian regeneration in the third-year female began with the formation of a new germinal zone from germ cells preserved in the atrophied ovaries and eosinophilic cells of the previously starving second-year female. Eosinophilic cells form the mesodermal component of the germinal zone. A mass of these cells appeared in the second-year female that had atrophied ovaries and in large numbers on the intestine wall of the third-year female with regenerating ovaries. These eosinophilic cells appear to migrate into the regenerating ovaries. Conclusions All germ cells of the second-year female are not lost during ovarian atrophy and can be involved in subsequent ovarian regeneration. Eosinophilic cells involved in ovarian regeneration are of mesodermal origin. The eosinophilic cell morphologies are similar to those of quiescence cells (cells in a reversible state that do not divide but retain the ability to re-enter cell division and participate in regeneration). These histological data thus indicate that eosinophilic and germ cells of third-year females can participate in the regeneration of the ovaries to reproduce a second brood. The precursors of these third-year females (a small number the second-year females with an asynchronous [summer] breeding period and ovaries that have atrophied due to seasonal starvation) appear to possess sources of somatic and germ cells that are sufficient for ovarian regeneration and that may be adaptations to starvation stress.
Collapse
Affiliation(s)
- Valentina B. Durkina
- Laboratory of Physiology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Primorsky Krai, Russia
| | - John W. Chapman
- Department of Fisheries, Wildlife and Conservation, Oregon State University, Newport, Oregon, United States of America
| | - Natalia L. Demchenko
- Laboratory of Marine Ecosystem Dynamics, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Primorsky Krai, Russia
| |
Collapse
|
10
|
Ho CH, Paolantoni C, Bawankar P, Tang Z, Brown S, Roignant J, Treisman JE. An exon junction complex-independent function of Barentsz in neuromuscular synapse growth. EMBO Rep 2022; 23:e53231. [PMID: 34726300 PMCID: PMC8728599 DOI: 10.15252/embr.202153231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023] Open
Abstract
The exon junction complex controls the translation, degradation, and localization of spliced mRNAs, and three of its core subunits also play a role in splicing. Here, we show that a fourth subunit, Barentsz, has distinct functions within and separate from the exon junction complex in Drosophila neuromuscular development. The distribution of mitochondria in larval muscles requires Barentsz as well as other exon junction complex subunits and is not rescued by a Barentsz transgene in which residues required for binding to the core subunit eIF4AIII are mutated. In contrast, interactions with the exon junction complex are not required for Barentsz to promote the growth of neuromuscular synapses. We find that the Activin ligand Dawdle shows reduced expression in barentsz mutants and acts downstream of Barentsz to control synapse growth. Both barentsz and dawdle are required in motor neurons, muscles, and glia for normal synapse growth, and exogenous Dawdle can rescue synapse growth in the absence of barentsz. These results identify a biological function for Barentsz that is independent of the exon junction complex.
Collapse
Affiliation(s)
- Cheuk Hei Ho
- Skirball Institute for Biomolecular Medicine and Department of Cell BiologyNYU School of MedicineNew YorkNYUSA
| | - Chiara Paolantoni
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
| | - Praveen Bawankar
- Institute of Pharmaceutical and Biomedical SciencesJohannes Gutenberg‐University MainzMainzGermany
| | - Zuojian Tang
- Center for Health Informatics and BioinformaticsNYU Langone Medical CenterNew YorkNYUSA
- Present address:
Computational Biology at Ridgefield US, Global Computational Biology and Digital ScienceBoehringer IngelheimRidgefieldCTUSA
| | - Stuart Brown
- Center for Health Informatics and BioinformaticsNYU Langone Medical CenterNew YorkNYUSA
- Present address:
ExxonMobil Corporate Strategic ResearchAnnandaleNJUSA
| | - Jean‐Yves Roignant
- Center for Integrative Genomics, Génopode Building, Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
- Institute of Pharmaceutical and Biomedical SciencesJohannes Gutenberg‐University MainzMainzGermany
| | - Jessica E Treisman
- Skirball Institute for Biomolecular Medicine and Department of Cell BiologyNYU School of MedicineNew YorkNYUSA
| |
Collapse
|
11
|
Banisch TU, Slaidina M, Gupta S, Ho M, Gilboa L, Lehmann R. A transitory signaling center controls timing of primordial germ cell differentiation. Dev Cell 2021; 56:1742-1755.e4. [PMID: 34081907 PMCID: PMC8330407 DOI: 10.1016/j.devcel.2021.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 03/07/2021] [Accepted: 05/10/2021] [Indexed: 12/16/2022]
Abstract
Organogenesis requires exquisite spatiotemporal coordination of cell morphogenesis, migration, proliferation, and differentiation of multiple cell types. For gonads, this involves complex interactions between somatic and germline tissues. During Drosophila ovary morphogenesis, primordial germ cells (PGCs) either are sequestered in stem cell niches and are maintained in an undifferentiated germline stem cell state or transition directly toward differentiation. Here, we identify a mechanism that links hormonal triggers of somatic tissue morphogenesis with PGC differentiation. An early ecdysone pulse initiates somatic swarm cell (SwC) migration, positioning these cells close to PGCs. A second hormone peak activates Torso-like signal in SwCs, which stimulates the Torso receptor tyrosine kinase (RTK) signaling pathway in PGCs promoting their differentiation by de-repression of the differentiation gene, bag of marbles. Thus, systemic temporal cues generate a transitory signaling center that coordinates ovarian morphogenesis with stem cell self-renewal and differentiation programs, highlighting a more general role for such centers in reproductive and developmental biology.
Collapse
Affiliation(s)
- Torsten U Banisch
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA.
| | - Maija Slaidina
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Selena Gupta
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Megan Ho
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Lilach Gilboa
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ruth Lehmann
- Department of Cell Biology, Howard Hughes Medical Institute, Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
12
|
Miscopein Saler L, Hauser V, Bartoletti M, Mallart C, Malartre M, Lebrun L, Pret AM, Théodore L, Chalvet F, Netter S. The Bric-à-Brac BTB/POZ transcription factors are necessary in niche cells for germline stem cells establishment and homeostasis through control of BMP/DPP signaling in the Drosophila melanogaster ovary. PLoS Genet 2020; 16:e1009128. [PMID: 33151937 PMCID: PMC7643948 DOI: 10.1371/journal.pgen.1009128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/22/2020] [Indexed: 11/26/2022] Open
Abstract
Many studies have focused on the mechanisms of stem cell maintenance via their interaction with a particular niche or microenvironment in adult tissues, but how formation of a functional niche is initiated, including how stem cells within a niche are established, is less well understood. Adult Drosophila melanogaster ovary Germline Stem Cell (GSC) niches are comprised of somatic cells forming a stack called a Terminal Filament (TF) and associated Cap and Escort Cells (CCs and ECs, respectively), which are in direct contact with GSCs. In the adult ovary, the transcription factor Engrailed is specifically expressed in niche cells where it directly controls expression of the decapentaplegic (dpp) gene encoding a member of the Bone Morphogenetic Protein (BMP) family of secreted signaling molecules, which are key factors for GSC maintenance. In larval ovaries, in response to BMP signaling from newly formed niches, adjacent primordial germ cells become GSCs. The bric-à-brac paralogs (bab1 and bab2) encode BTB/POZ domain-containing transcription factors that are expressed in developing niches of larval ovaries. We show here that their functions are necessary specifically within precursor cells for TF formation during these stages. We also identify a new function for Bab1 and Bab2 within developing niches for GSC establishment in the larval ovary and for robust GSC maintenance in the adult. Moreover, we show that the presence of Bab proteins in niche cells is necessary for activation of transgenes reporting dpp expression as of larval stages in otherwise correctly specified Cap Cells, independently of Engrailed and its paralog Invected (En/Inv). Moreover, strong reduction of engrailed/invected expression during larval stages does not impair TF formation and only partially reduces GSC numbers. In the adult ovary, Bab proteins are also required for dpp reporter expression in CCs. Finally, when bab2 was overexpressed at this stage in somatic cells outside of the niche, there were no detectable levels of ectopic En/Inv, but ectopic expression of a dpp transgene was found in these cells and BMP signaling activation was induced in adjacent germ cells, which produced GSC-like tumors. Together, these results indicate that Bab transcription factors are positive regulators of BMP signaling in niche cells for establishment and homeostasis of GSCs in the Drosophila ovary.
Collapse
Affiliation(s)
- Laurine Miscopein Saler
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Virginie Hauser
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Mathieu Bartoletti
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Charlotte Mallart
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Marianne Malartre
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Laura Lebrun
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Anne-Marie Pret
- Université Paris-Saclay, UVSQ, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Laurent Théodore
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Fabienne Chalvet
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Sophie Netter
- Université Paris-Saclay, UVSQ, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| |
Collapse
|
13
|
Blatt P, Martin ET, Breznak SM, Rangan P. Post-transcriptional gene regulation regulates germline stem cell to oocyte transition during Drosophila oogenesis. Curr Top Dev Biol 2019; 140:3-34. [PMID: 32591078 DOI: 10.1016/bs.ctdb.2019.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
During oogenesis, several developmental processes must be traversed to ensure effective completion of gametogenesis including, stem cell maintenance and asymmetric division, differentiation, mitosis and meiosis, and production of maternally contributed mRNAs, making the germline a salient model for understanding how cell fate transitions are mediated. Due to silencing of the genome during meiotic divisions, there is little instructive transcription, barring a few examples, to mediate these critical transitions. In Drosophila, several layers of post-transcriptional regulation ensure that the mRNAs required for these processes are expressed in a timely manner and as needed during germline differentiation. These layers of regulation include alternative splicing, RNA modification, ribosome production, and translational repression. Many of the molecules and pathways involved in these regulatory activities are conserved from Drosophila to humans making the Drosophila germline an elegant model for studying the role of post-transcriptional regulation during stem cell differentiation and meiosis.
Collapse
Affiliation(s)
- Patrick Blatt
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY, United States; University at Albany SUNY, Albany, NY, United States
| | - Elliot T Martin
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY, United States; University at Albany SUNY, Albany, NY, United States
| | - Shane M Breznak
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY, United States; University at Albany SUNY, Albany, NY, United States
| | - Prashanth Rangan
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY, United States; University at Albany SUNY, Albany, NY, United States.
| |
Collapse
|
14
|
Tiwari MD, Zeitler DM, Meister G, Wodarz A. Molecular profiling of stem cell-like female germ line cells in Drosophila delineates networks important for stemness and differentiation. Biol Open 2019; 8:bio.046789. [PMID: 31649115 PMCID: PMC6899027 DOI: 10.1242/bio.046789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Stem cells can self-renew and produce daughter cells destined for differentiation. The precise control of the balance between these two outcomes is essential to ensure tissue homeostasis and to prevent uncontrolled proliferation resulting in tumor formation. As self-renewal and differentiation are likely to be controlled by different gene expression programs, unraveling the underlying gene regulatory networks is crucial for understanding the molecular logic of this system. In this study, we have characterized by next generation RNA sequencing (RNA-seq) the transcriptome of germline stem cell (GSC)-like cells isolated from bag of marbles (bam) mutant Drosophila ovaries and compared it to the transcriptome of germ line cells isolated from wild-type ovaries. We have complemented this dataset by utilizing an RNA-immunoprecipitation strategy to identify transcripts bound to the master differentiation factor Bam. Protein complex enrichment analysis on these combined datasets allows us to delineate known and novel networks essential for GSC maintenance and differentiation. Further comparative transcriptomics illustrates similarities between GSCs and primordial germ cells and provides a molecular footprint of the stem cell state. Our study represents a useful resource for functional studies on stem cell maintenance and differentiation. Summary: Fruit fly germline stem cell differentiation is accompanied by major changes of the transcriptome that may be regulated at the post-transcriptional level.
Collapse
Affiliation(s)
- Manu D Tiwari
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Kerpener Str. 62, 50937 Köln, Germany .,Cluster of Excellence - Cellular stress response in aging-associated diseases (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany.,Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany
| | - Daniela M Zeitler
- Regensburg Center for Biochemistry (RCB), University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany
| | - Gunter Meister
- Regensburg Center for Biochemistry (RCB), University of Regensburg, Universitätsstr. 31, 93053 Regensburg, Germany
| | - Andreas Wodarz
- Molecular Cell Biology, Institute I for Anatomy, University of Cologne Medical School, Kerpener Str. 62, 50937 Köln, Germany .,Cluster of Excellence - Cellular stress response in aging-associated diseases (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany.,Stem Cell Biology, Institute for Anatomy and Cell Biology, Georg-August University Göttingen, Justus-von-Liebig-Weg 11, 37077 Göttingen, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| |
Collapse
|
15
|
Gao Y, Mao Y, Xu RG, Zhu R, Zhang M, Sun J, Shen D, Peng P, Xie T, Ni JQ. Defining gene networks controlling the maintenance and function of the differentiation niche by an in vivo systematic RNAi screen. J Genet Genomics 2019; 46:19-30. [PMID: 30745214 DOI: 10.1016/j.jgg.2018.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/02/2018] [Accepted: 10/23/2018] [Indexed: 01/13/2023]
Abstract
In the Drosophila ovary, escort cells (ECs) extrinsically control germline stem cell (GSC) maintenance and progeny differentiation. However, the underlying mechanisms remain poorly understood. In this study, we identified 173 EC genes for their roles in controlling GSC maintenance and progeny differentiation by using an in vivo systematic RNAi approach. Of the identified genes, 10 and 163 are required in ECs to promote GSC maintenance and progeny differentiation, respectively. The genes required for progeny differentiation fall into different functional categories, including transcription, mRNA splicing, protein degradation, signal transduction and cytoskeleton regulation. In addition, the GSC progeny differentiation defects caused by defective ECs are often associated with BMP signaling elevation, indicating that preventing BMP signaling is a general functional feature of the differentiation niche. Lastly, exon junction complex (EJC) components, which are essential for mRNA splicing, are required in ECs to promote GSC progeny differentiation by maintaining ECs and preventing BMP signaling. Therefore, this study has identified the major regulators of the differentiation niche, which provides important insights into how stem cell progeny differentiation is extrinsically controlled.
Collapse
Affiliation(s)
- Yuan Gao
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ying Mao
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Rong-Gang Xu
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China; Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ruibao Zhu
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China; Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ming Zhang
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jin Sun
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China; Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Da Shen
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China; Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ping Peng
- PKU-THU Joint Center for Life Sciences, College of Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China; Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ting Xie
- Stowers Institute for Medical Research, 1000 East 50(th) Street, Kansas City, MO, 64110, USA.
| | - Jian-Quan Ni
- Gene Regulatory Lab, School of Medicine, Tsinghua University, Beijing, 100084, China; Tsingdao Advanced Research Institute, Tongji University, Qingdao, 266000, China.
| |
Collapse
|
16
|
|
17
|
Xu R, Li Q, Yu H, Kong L. Oocyte maturation and origin of the germline as revealed by the expression of Nanos-like in the Pacific oyster Crassostrea gigas. Gene 2018; 663:41-50. [PMID: 29660519 DOI: 10.1016/j.gene.2018.04.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/07/2018] [Accepted: 04/09/2018] [Indexed: 01/19/2023]
Abstract
Nanos gene plays an important role in germline development in animals. However, the molecular mechanisms involved in germline development in Mollusca, the second largest animal phylum, are still poorly understood. Here we identified the Nanos orthologue from the Pacific oyster Crassostrea gigas (Cg-Nanos-like), and investigated the expression patterns of Nanos during gametogenesis and embryogenesis in C. gigas. Tissue expression analysis showed that Cg-Nanos-like was specifically expressed in female gonads. During the reproductive cycle, the expression of Cg-Nanos-like mRNA increased matching the seasonal development of the ovarian tissues in diploids, while the expression levels were significantly lower in the ovaries of sterile triploids compared to diploids. High expression of Cg-Nanos-like transcripts were detected in early embryonic stages, while the expression significantly dropped at gastrulation and was barely detectable in veliger stages. In situ hybridization showed that Cg-Nanos-like was expressed at different stages of developing oocytes, whereas positive signals were detected only in spermatogonia during the spermatogenic cycle. These findings indicated that Cg-Nanos-like was involved in the development of germ cells, and maintenance of oocyte maturation. In early embryogenesis, the transcripts were broadly expressed; following gastrulation, the expression was restricted to two cell clumps, which might be the putative primordial germ cells (PGCs) or their precursors. Based on the results, the formation of the PGCs in C. gigas was consistent with the model of transition from epigenesis to preformation.
Collapse
Affiliation(s)
- Rui Xu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, China.
| | - Hong Yu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China.
| | - Lingfeng Kong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
18
|
Spermiogenesis and Male Fertility Require the Function of Suppressor of Hairy-Wing in Somatic Cyst Cells of Drosophila. Genetics 2018; 209:757-772. [PMID: 29739818 DOI: 10.1534/genetics.118.301088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023] Open
Abstract
Drosophila Suppressor of Hairy-wing [Su(Hw)] protein is an example of a multivalent transcription factor. Although best known for its role in establishing the chromatin insulator of the gypsy retrotransposon, Su(Hw) functions as an activator and repressor at non-gypsy genomic sites. It remains unclear how the different regulatory activities of Su(Hw) are utilized during development. Motivated from observations of spatially restricted expression of Su(Hw) in the testis, we investigated the role of Su(Hw) in spermatogenesis to advance an understanding of its developmental contributions as an insulator, repressor, and activator protein. We discovered that Su(Hw) is required for sustained male fertility. Although dynamics of Su(Hw) expression coincide with changes in nuclear architecture and activation of coregulated testis-specific gene clusters, we show that loss of Su(Hw) does not disrupt meiotic chromosome pairing or transcription of testis-specific genes, suggesting that Su(Hw) has minor architectural or insulator functions in the testis. Instead, Su(Hw) has a prominent role as a repressor of neuronal genes, consistent with suggestions that Su(Hw) is a functional homolog of mammalian REST, a repressor of neuronal genes in non-neuronal tissues. We show that Su(Hw) regulates transcription in both germline and somatic cells. Surprisingly, the essential spermatogenesis function of Su(Hw) resides in somatic cyst cells, implying context-specific consequences due to loss of this transcription factor. Together, our studies highlight that Su(Hw) has a major developmental function as a transcriptional repressor, with the effect of its loss dependent upon the cell-specific factors.
Collapse
|
19
|
Coux RX, Teixeira FK, Lehmann R. L(3)mbt and the LINT complex safeguard cellular identity in the Drosophila ovary. Development 2018; 145:dev.160721. [PMID: 29511022 PMCID: PMC5963868 DOI: 10.1242/dev.160721] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/19/2018] [Indexed: 01/10/2023]
Abstract
Maintenance of cellular identity is essential for tissue development and homeostasis. At the molecular level, cell identity is determined by the coordinated activation and repression of defined sets of genes. The tumor suppressor L(3)mbt has been shown to secure cellular identity in Drosophila larval brains by repressing germline-specific genes. Here, we interrogate the temporal and spatial requirements for L(3)mbt in the Drosophila ovary, and show that it safeguards the integrity of both somatic and germline tissues. l(3)mbt mutant ovaries exhibit multiple developmental defects, which we find to be largely caused by the inappropriate expression of a single gene, nanos, a key regulator of germline fate, in the somatic ovarian cells. In the female germline, we find that L(3)mbt represses testis-specific and neuronal genes. At the molecular level, we show that L(3)mbt function in the ovary is mediated through its co-factor Lint-1 but independently of the dREAM complex. Together, our work uncovers a more complex role for L(3)mbt than previously understood and demonstrates that L(3)mbt secures tissue identity by preventing the simultaneous expression of original identity markers and tissue-specific misexpression signatures. Highlighted Article: Characterization of the developmental defects of l(3)mbt mutant ovaries shows that L(3)mbt regulates tissue-specific gene signatures to secure the identity of somatic ovarian and germline tissues.
Collapse
Affiliation(s)
- Rémi-Xavier Coux
- Howard Hughes Medical Institute (HHMI) and Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Felipe Karam Teixeira
- Howard Hughes Medical Institute (HHMI) and Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA.,Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Ruth Lehmann
- Howard Hughes Medical Institute (HHMI) and Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
20
|
Upadhyay M, Kuna M, Tudor S, Martino Cortez Y, Rangan P. A switch in the mode of Wnt signaling orchestrates the formation of germline stem cell differentiation niche in Drosophila. PLoS Genet 2018; 14:e1007154. [PMID: 29370168 PMCID: PMC5811049 DOI: 10.1371/journal.pgen.1007154] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 02/13/2018] [Accepted: 12/13/2017] [Indexed: 01/12/2023] Open
Abstract
Germline stem cell (GSC) self-renewal and differentiation into gametes is regulated by both intrinsic factors in the germ line as well as extrinsic factors from the surrounding somatic niche. dWnt4, in the escort cells of the adult somatic niche promotes GSC differentiation using the canonical β-catenin-dependent transcriptional pathway to regulate escort cell survival, adhesion to the germ line and downregulation of self-renewal signaling. Here, we show that in addition to the β-catenin-dependent canonical pathway, dWnt4 also uses downstream components of the Wnt non-canonical pathway to promote escort cell function earlier in development. We find that the downstream non-canonical components, RhoA, Rac1 and cdc42, are expressed at high levels and are active in escort cell precursors of the female larval gonad compared to the adult somatic niche. Consistent with this expression pattern, we find that the non-canonical pathway components function in the larval stages but not in adults to regulate GSC differentiation. In the larval gonad, dWnt4, RhoA, Rac1 and cdc42 are required to promote intermingling of escort cell precursors, a function that then promotes proper escort cell function in the adults. We find that dWnt4 acts by modulating the activity of RhoA, Rac1 and cdc42, but not their protein levels. Together, our results indicate that at different points of development, dWnt4 switches from using the non-canonical pathway components to using a β-catenin-dependent canonical pathway in the escort cells to facilitate the proper differentiation of GSCs. Germ line association with the somatic cells is critical for various aspects of germ cell biology, including migration, self-renewal and differentiation. In Drosophila females, soma–germ line association begins during embryogenesis and continues until the mature egg is formed. In the adult, the somatic escort cells promote differentiation of the germline stem cell daughter using Wnt signaling. dWnt4, a Wnt ligand, acts in an autocrine manner in these escort cells, using the canonical pathway to regulate survival, division and encapsulation of the stem cell daughter, a function critical for differentiation. Here, we show at an earlier stage, in the larvae, the same ligand uses components of Wnt non-canonical pathway, RhoA, Rac1 and cdc42, to regulate proper mingling of escort cell precursors between the germ cells. Thus, dWnt4 uses different modules of signaling at different points in development to promote cell movement and control cytoplasmic protrusions. As Wnts have been associated with cancers, understanding how Wnts modulate cell movement by switching on and off different modules may lead to insights into the etiology and progression of cancers.
Collapse
Affiliation(s)
- Maitreyi Upadhyay
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, New York, United States of America
| | - Michael Kuna
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, New York, United States of America
- Albany Medical College, Albany, New York, United States of America
| | - Sara Tudor
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, New York, United States of America
- Albany Medical College, Albany, New York, United States of America
| | - Yesenia Martino Cortez
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, New York, United States of America
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Prashanth Rangan
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Chartier A, Joly W, Simonelig M. Measurement of mRNA Poly(A) Tail Lengths in Drosophila Female Germ Cells and Germ-Line Stem Cells. Methods Mol Biol 2018; 1463:93-102. [PMID: 27734350 DOI: 10.1007/978-1-4939-4017-2_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
mRNA regulation by poly(A) tail length variations plays an important role in many developmental processes. Recent advances have shown that, in particular, deadenylation (the shortening of mRNA poly(A) tails) is essential for germ-line stem cell biology in the Drosophila ovary. Therefore, a rapid and accurate method to analyze poly(A) tail lengths of specific mRNAs in this tissue is valuable. Several methods of poly(A) test (PAT) assays have been reported to measure mRNA poly(A) tail lengths in vivo. Here, we describe two of these methods (PAT and ePAT) that we have adapted for Drosophila ovarian germ cells and germ-line stem cells.
Collapse
Affiliation(s)
- Aymeric Chartier
- mRNA Regulation and Development, Institut de Génétique Humaine, CNRS UPR1142 and University of Montpellier, 141 rue de la Cardonille, 34396, Montpellier Cedex 5, France
| | - Willy Joly
- mRNA Regulation and Development, Institut de Génétique Humaine, CNRS UPR1142 and University of Montpellier, 141 rue de la Cardonille, 34396, Montpellier Cedex 5, France
| | - Martine Simonelig
- mRNA Regulation and Development, Institut de Génétique Humaine, CNRS UPR1142 and University of Montpellier, 141 rue de la Cardonille, 34396, Montpellier Cedex 5, France
| |
Collapse
|
22
|
Bohn JA, Van Etten JL, Schagat TL, Bowman BM, McEachin RC, Freddolino PL, Goldstrohm AC. Identification of diverse target RNAs that are functionally regulated by human Pumilio proteins. Nucleic Acids Res 2018; 46:362-386. [PMID: 29165587 PMCID: PMC5758885 DOI: 10.1093/nar/gkx1120] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/21/2017] [Accepted: 10/25/2017] [Indexed: 12/20/2022] Open
Abstract
Human Pumilio proteins, PUM1 and PUM2, are sequence specific RNA-binding proteins that regulate protein expression. We used RNA-seq, rigorous statistical testing and an experimentally derived fold change cut-off to identify nearly 1000 target RNAs-including mRNAs and non-coding RNAs-that are functionally regulated by PUMs. Bioinformatic analysis defined a PUM Response Element (PRE) that was significantly enriched in transcripts that increased in abundance and matches the PUM RNA-binding consensus. We created a computational model that incorporates PRE position and frequency within an RNA relative to the magnitude of regulation. The model reveals significant correlation of PUM regulation with PREs in 3' untranslated regions (UTRs), coding sequences and non-coding RNAs, but not 5' UTRs. To define direct, high confidence PUM targets, we cross-referenced PUM-regulated RNAs with all PRE-containing RNAs and experimentally defined PUM-bound RNAs. The results define nearly 300 direct targets that include both PUM-repressed and, surprisingly, PUM-activated target RNAs. Annotation enrichment analysis reveal that PUMs regulate genes from multiple signaling pathways and developmental and neurological processes. Moreover, PUM target mRNAs impinge on human disease genes linked to cancer, neurological disorders and cardiovascular disease. These discoveries pave the way for determining how the PUM-dependent regulatory network impacts biological functions and disease states.
Collapse
Affiliation(s)
- Jennifer A Bohn
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jamie L Van Etten
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Trista L Schagat
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
- Promega Corporation, Madison, WI 53711, USA
| | - Brittany M Bowman
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard C McEachin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter L Freddolino
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aaron C Goldstrohm
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
23
|
Salz HK, Dawson EP, Heaney JD. Germ cell tumors: Insights from the Drosophila ovary and the mouse testis. Mol Reprod Dev 2017; 84:200-211. [PMID: 28079292 DOI: 10.1002/mrd.22779] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/10/2017] [Indexed: 12/14/2022]
Abstract
Ovarian and testicular germ cell tumors of young adults are thought to arise from defects in germ cell development, but the molecular mechanisms underlying malignant transformation are poorly understood. In this review, we focus on the biology of germ cell tumor formation in the Drosophila ovary and the mouse testis, for which evidence supports common underlying mechanisms, such as blocking initiation into the differentiation pathway, impaired lineage progression, and sexual identity instability. We then discuss how these concepts inform our understanding of the disease in humans. Mol. Reprod. Dev. 84: 200-211, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Helen K Salz
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Emily P Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
24
|
Flora P, Schowalter S, Wong-Deyrup S, DeGennaro M, Nasrallah MA, Rangan P. Transient transcriptional silencing alters the cell cycle to promote germline stem cell differentiation in Drosophila. Dev Biol 2017; 434:84-95. [PMID: 29198563 PMCID: PMC5830152 DOI: 10.1016/j.ydbio.2017.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/18/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022]
Abstract
Transcriptional silencing is a conserved process used by embryonic germ cells to repress somatic fate and maintain totipotency and immortality. In Drosophila, this transcriptional silencing is mediated by polar granule component (pgc). Here, we show that in the adult ovary, pgc is required for timely germline stem cell (GSC) differentiation. Pgc is expressed transiently in the immediate GSC daughter (pre-cystoblast), where it mediates a pulse of transcriptional silencing. This transcriptional silencing mediated by pgc indirectly promotes the accumulation of Cyclin B (CycB) and cell cycle progression into late-G2 phase, when the differentiation factor bag of marbles (bam) is expressed. Pgc mediated accumulation of CycB is also required for heterochromatin deposition, which protects the germ line genome against selfish DNA elements. Our results suggest that transient transcriptional silencing in the pre-cystoblast “re-programs” it away from self-renewal and toward the gamete differentiation program.
Collapse
Affiliation(s)
- Pooja Flora
- Department of Biological Sciences/The RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Sean Schowalter
- Department of Biological Sciences/The RNA Institute, University at Albany SUNY, Albany, NY 12222, USA; Boston University School of Medicine, 815 Albany Street, MA 02119, USA
| | - SiuWah Wong-Deyrup
- Department of Biological Sciences/The RNA Institute, University at Albany SUNY, Albany, NY 12222, USA
| | - Matthew DeGennaro
- Biomolecular Sciences Institute, Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Mohamad Ali Nasrallah
- Department of Biological Sciences/The RNA Institute, University at Albany SUNY, Albany, NY 12222, USA; University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Prashanth Rangan
- Department of Biological Sciences/The RNA Institute, University at Albany SUNY, Albany, NY 12222, USA.
| |
Collapse
|
25
|
Romero-Pozuelo J, Demetriades C, Schroeder P, Teleman AA. CycD/Cdk4 and Discontinuities in Dpp Signaling Activate TORC1 in the Drosophila Wing Disc. Dev Cell 2017; 42:376-387.e5. [PMID: 28829945 DOI: 10.1016/j.devcel.2017.07.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 06/19/2017] [Accepted: 07/23/2017] [Indexed: 01/08/2023]
Abstract
The molecular mechanisms regulating animal tissue size during development are unclear. This question has been extensively studied in the Drosophila wing disc. Although cell growth is regulated by the kinase TORC1, no readout exists to visualize TORC1 activity in situ in Drosophila. Both the cell cycle and the morphogen Dpp are linked to tissue growth, but whether they regulate TORC1 activity is not known. We develop here an anti-phospho-dRpS6 antibody that detects TORC1 activity in situ. We find, unexpectedly, that TORC1 activity in the wing disc is patchy. This is caused by elevated TORC1 activity at the cell cycle G1/S transition due to CycD/Cdk4 phosphorylating TSC1/2. We find that TORC1 is also activated independently of CycD/Cdk4 when cells with different levels of Dpp signaling or Brinker protein are juxtaposed. We thereby characterize the spatial distribution of TORC1 activity in a developing organ.
Collapse
Affiliation(s)
- Jesús Romero-Pozuelo
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | - Constantinos Demetriades
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | - Phillip Schroeder
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany
| | - Aurelio A Teleman
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
26
|
Bone Morphogenetic Protein (BMP) signaling in animal reproductive system development and function. Dev Biol 2017; 427:258-269. [DOI: 10.1016/j.ydbio.2017.03.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 12/15/2022]
|
27
|
Oulhen N, Swartz SZ, Laird J, Mascaro A, Wessel GM. Transient translational quiescence in primordial germ cells. Development 2017; 144:1201-1210. [PMID: 28235822 PMCID: PMC5399625 DOI: 10.1242/dev.144170] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 02/01/2017] [Indexed: 01/07/2023]
Abstract
Stem cells in animals often exhibit a slow cell cycle and/or low transcriptional activity referred to as quiescence. Here, we report that the translational activity in the primordial germ cells (PGCs) of the sea urchin embryo (Strongylocentrotus purpuratus) is quiescent. We measured new protein synthesis with O-propargyl-puromycin and L-homopropargylglycine Click-iT technologies, and determined that these cells synthesize protein at only 6% the level of their adjacent somatic cells. Knockdown of translation of the RNA-binding protein Nanos2 by morpholino antisense oligonucleotides, or knockout of the Nanos2 gene by CRISPR/Cas9 resulted in a significant, but partial, increase (47%) in general translation specifically in the PGCs. We found that the mRNA of the translation factor eEF1A is excluded from the PGCs in a Nanos2-dependent manner, a consequence of a Nanos/Pumilio response element (PRE) in its 3'UTR. In addition to eEF1A, the cytoplasmic pH of the PGCs appears to repress translation and simply increasing the pH also significantly restores translation selectively in the PGCs. We conclude that the PGCs of this sea urchin institute parallel pathways to quiesce translation thoroughly but transiently.
Collapse
Affiliation(s)
- Nathalie Oulhen
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - S Zachary Swartz
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Whitehead Institute for Biomedical Research, MIT, Nine Cambridge Center, Cambridge, MA 02142, USA
| | - Jessica Laird
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - Alexandra Mascaro
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - Gary M Wessel
- Department of Molecular and Cell Biology and Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| |
Collapse
|
28
|
Hitrik A, Popliker M, Gancz D, Mukamel Z, Lifshitz A, Schwartzman O, Tanay A, Gilboa L. Combgap Promotes Ovarian Niche Development and Chromatin Association of EcR-Binding Regions in BR-C. PLoS Genet 2016; 12:e1006330. [PMID: 27846223 PMCID: PMC5147775 DOI: 10.1371/journal.pgen.1006330] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 08/29/2016] [Indexed: 01/26/2023] Open
Abstract
The development of niches for tissue-specific stem cells is an important aspect of stem cell biology. Determination of niche size and niche numbers during organogenesis involves precise control of gene expression. How this is achieved in the context of a complex chromatin landscape is largely unknown. Here we show that the nuclear protein Combgap (Cg) supports correct ovarian niche formation in Drosophila by controlling ecdysone-Receptor (EcR)- mediated transcription and long-range chromatin contacts in the broad locus (BR-C). Both cg and BR-C promote ovarian growth and the development of niches for germ line stem cells. BR-C levels were lower when Combgap was either reduced or over-expressed, indicating an intricate regulation of the BR-C locus by Combgap. Polytene chromosome stains showed that Cg co-localizes with EcR, the major regulator of BR-C, at the BR-C locus and that EcR binding to chromatin was sensitive to changes in Cg levels. Proximity ligation assay indicated that the two proteins could reside in the same complex. Finally, chromatin conformation analysis revealed that EcR-bound regions within BR-C, which span ~30 KBs, contacted each other. Significantly, these contacts were stabilized in an ecdysone- and Combgap-dependent manner. Together, these results highlight Combgap as a novel regulator of chromatin structure that promotes transcription of ecdysone target genes and ovarian niche formation. Germ line stem cells (GSCs) supply either eggs or sperm throughout the life- time of many organisms, including mammals. For their function, GSCs require input from somatic niche cells. Understanding how niches form during development is an important initial step in understanding how stem cell units form, and by extension, how they may regenerate. In this work we describe a new function for the chromatin binding protein Combgap in ovarian niche formation of the model organism Drosophila melanogaster. Combgap is required for the correct expression of another factor, BR-C, in somatic ovarian cells. BR-C is one of the central target genes of the steroid hormone ecdysone, and its expression is controlled by the ecdysone receptor (EcR). Interestingly, EcR-enriched regions within the BR-C locus are engaged in long-range contacts that are stabilized by ecdysone in a Combgap-depended manner. We also found that EcR binding to chromatin depends on WT levels of Combgap. BR-C regulates GSC unit establishment, intestinal stem cells, immune responses, and many other processes. Understanding Combgaps’ function in shaping the BR-C chromatin landscape is a first step towards better appreciation of how this important locus is controlled, and the general machinery coupling gene expression to 3D chromatin structure.
Collapse
Affiliation(s)
- Anna Hitrik
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Malka Popliker
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Dana Gancz
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Zohar Mukamel
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Aviezer Lifshitz
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Omer Schwartzman
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Mol. Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Childhood Leukemia Research Institute, Sheba Medical Center, Ramat Gan, Israel
| | - Amos Tanay
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
| | - Lilach Gilboa
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
29
|
Xin T, Greco V, Myung P. Hardwiring Stem Cell Communication through Tissue Structure. Cell 2016; 164:1212-1225. [PMID: 26967287 DOI: 10.1016/j.cell.2016.02.041] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Indexed: 12/24/2022]
Abstract
Adult stem cells across diverse organs self-renew and differentiate to maintain tissue homeostasis. How stem cells receive input to preserve tissue structure and function largely relies on their communication with surrounding cellular and non-cellular elements. As such, how tissues are organized and patterned not only reflects organ function, but also inherently hardwires networks of communication between stem cells and their environment to direct tissue homeostasis and injury repair. This review highlights how different methods of stem cell communication reflect the unique organization and function of diverse tissues.
Collapse
Affiliation(s)
- Tianchi Xin
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06510, USA; Department of Dermatology, Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06510, USA; Department of Cell Biology, Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut 06510, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06510, USA.
| | - Peggy Myung
- Department of Dermatology, Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06510, USA; Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut 06510, USA.
| |
Collapse
|
30
|
Yang H, Li M, Hu X, Xin T, Zhang S, Zhao G, Xuan T, Li M. MicroRNA-dependent roles of Drosha and Pasha in the Drosophila larval ovary morphogenesis. Dev Biol 2016; 416:312-23. [DOI: 10.1016/j.ydbio.2016.06.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/17/2016] [Accepted: 06/17/2016] [Indexed: 01/04/2023]
|
31
|
Barton LJ, Lovander KE, Pinto BS, Geyer PK. Drosophila male and female germline stem cell niches require the nuclear lamina protein Otefin. Dev Biol 2016; 415:75-86. [PMID: 27174470 DOI: 10.1016/j.ydbio.2016.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 04/28/2016] [Accepted: 05/02/2016] [Indexed: 12/11/2022]
Abstract
The nuclear lamina is an extensive protein network that underlies the inner nuclear envelope. This network includes the LAP2-emerin-MAN1-domain (LEM-D) protein family, proteins that share an association with the chromatin binding protein Barrier-to-autointegration factor (BAF). Loss of individual LEM-D proteins causes progressive, tissue-restricted diseases, known as laminopathies. Mechanisms associated with laminopathies are not yet understood. Here we present our studies of one of the Drosophila nuclear lamina LEM-D proteins, Otefin (Ote), a homologue of emerin. Previous studies have shown that Ote is autonomously required for the survival of female germline stem cells (GSCs). We demonstrate that Ote is also required for survival of somatic cells in the ovarian niche, with loss of Ote causing a decrease in cap cell number and altered signal transduction. We show germ cell-restricted expression of Ote rescues these defects, revealing a non-autonomous function for Ote in niche maintenance and emphasizing that GSCs contribute to the maintenance of their own niches. Further, we investigate the requirement of Ote in the male fertility. We show that ote mutant males become prematurely sterile as they age. Parallel to observations in females, this sterility is associated with GSC loss and changes in somatic cells of the niche, phenotypes that are largely rescued by germ cell-restricted Ote expression. Taken together, our studies demonstrate that Ote is required autonomously for survival of two stem cell populations, as well as non-autonomously for maintenance of two somatic niches. Finally, our data add to growing evidence that LEM-D proteins have critical roles in stem cell survival and tissue homeostasis.
Collapse
Affiliation(s)
- Lacy J Barton
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Kaylee E Lovander
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Belinda S Pinto
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Pamela K Geyer
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
32
|
Banerjee A, Saha SK. Histological and ultrastructural investigation of the female reproductive system of Argulus bengalensis Ramakrishna, 1951 (Crustacea: Branchiura). J Morphol 2016; 277:707-16. [PMID: 26991011 DOI: 10.1002/jmor.20528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 02/11/2016] [Accepted: 02/21/2016] [Indexed: 11/08/2022]
Abstract
In order to understand branchiuran reproductive biology, it is imperative to know the sites of oogenesis and oocyte maturation, locate the accessory reproductive glands, and identify the fertilization site with the present knowledge of the sperm transfer mechanism of the genus Argulus. With these objectives, we attempted to describe the female reproductive system of Argulus bengalensis using serial histological sections through the ovaries and associated ducts in the transverse, longitudinal, and sagittal planes. The reproductive organs include a median ovary, one pair of ovarian lumina, a median oviduct, and a pair of collateral accessory glands. A duct from each of the collateral accessory glands leads into the proximal part of the median oviduct, which opens to the exterior through a genital opening at the distal end. The glandular secretion presumably contributes to the jelly coat of the egg. The ovary is bound with a tunica propria which extends further diametrically inside the ovary forming the paired lumina. The lumina are confluent into the median oviduct. Two distinct areas, the germarium and differentiating zones, are clearly distinguishable within the ovary. The tunica propria itself houses the oogonia within a matrix, serving as the germarium. Transmission electron micrograph reveals that the matrix is made of collagen. The collagen matrix confers elasticity to the tunica propria to accommodate the postvitellogenic oocytes within the ovarian lumen. The differentiating zone is situated in between the germarium: dorsally it is covered with a chromatophore layer. The ovary is ensheathed by a circum ovarian striated muscle. The presence of spermatophores in the ovarian lumen indicates the fertilization site. J. Morphol. 277:707-716, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anirban Banerjee
- Department of Zoology, Fish Biology Research Unit, School of Life Sciences, Visva-Bharati University, Santiniketan, West Bengal, 731235, India
| | - Samar Kumar Saha
- Department of Zoology, Fish Biology Research Unit, School of Life Sciences, Visva-Bharati University, Santiniketan, West Bengal, 731235, India
| |
Collapse
|
33
|
Fu Z, Geng C, Wang H, Yang Z, Weng C, Li H, Deng L, Liu L, Liu N, Ni J, Xie T. Twin Promotes the Maintenance and Differentiation of Germline Stem Cell Lineage through Modulation of Multiple Pathways. Cell Rep 2015; 13:1366-1379. [DOI: 10.1016/j.celrep.2015.10.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 08/12/2015] [Accepted: 10/05/2015] [Indexed: 11/28/2022] Open
|
34
|
Shapiro-Kulnane L, Smolko AE, Salz HK. Maintenance of Drosophila germline stem cell sexual identity in oogenesis and tumorigenesis. Development 2015; 142:1073-82. [PMID: 25758221 DOI: 10.1242/dev.116590] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adult stem cells maintain tissue homeostasis by balancing self-renewal and differentiation. In Drosophila females, germline stem cells (GSCs) require Sex lethal (Sxl) to exit the stem cell state and to enter the differentiation pathway. Without Sxl GSCs do not differentiate and instead form tumors. Previous studies have shown that these tumors are not caused by a failure in the self-renewal/differentiation switch. Here, we show that Sxl is also necessary for the cell-autonomous maintenance of germ cell female identity and demonstrate that tumors are caused by the acquisition of male characteristics. Germ cells without Sxl protein exhibit a global derepression of testis genes, including Phf7, a male germline sexual identity gene. Phf7 is a key effector of the tumor-forming pathway, as it is both necessary and sufficient for tumor formation. In the absence of Sxl protein, inappropriate Phf7 expression drives tumor formation through a cell-autonomous mechanism that includes sex-inappropriate activation of Jak/Stat signaling. Remarkably, tumor formation requires a novel response to external signals emanating from the GSC niche, highlighting the importance of interactions between mutant cells and the surrounding normal cells that make up the tumor microenvironment. Derepression of testis genes, and inappropriate Phf7 expression, is also observed in germ cell tumors arising from the loss of bag of marbles (bam), demonstrating that maintenance of female sexual identity requires the concerted actions of Sxl and bam. Our work reveals that GSCs must maintain their sexual identity as they are reprogrammed into a differentiated cell, or risk tumorigenesis.
Collapse
Affiliation(s)
- Laura Shapiro-Kulnane
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4955, USA
| | - Anne Elizabeth Smolko
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4955, USA
| | - Helen Karen Salz
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4955, USA
| |
Collapse
|
35
|
Abstract
Stem cells give rise to tissues and organs during development and maintain their integrity during adulthood. They have the potential to self-renew or differentiate at each division. To ensure proper organ growth and homeostasis, self-renewal versus differentiation decisions need to be tightly controlled. Systematic genetic studies in Drosophila melanogaster are revealing extensive regulatory networks that control the switch between stem cell self-renewal and differentiation in the germline. These networks, which are based primarily on mutual translational repression, act via interlocked feedback loops to provide robustness to this important fate decision.
Collapse
Affiliation(s)
- Maija Slaidina
- Howard Hughes Medical Institute and Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, NY 10016 Howard Hughes Medical Institute and Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| | - Ruth Lehmann
- Howard Hughes Medical Institute and Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, NY 10016 Howard Hughes Medical Institute and Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Cell Biology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
36
|
Gilboa L. Organizing stem cell units in the Drosophila ovary. Curr Opin Genet Dev 2015; 32:31-6. [PMID: 25703842 DOI: 10.1016/j.gde.2015.01.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/17/2015] [Accepted: 01/21/2015] [Indexed: 01/20/2023]
Abstract
Organogenesis utilizes processes fundamental to development: cell proliferation, cell differentiation and morphogenesis. Each of these processes is complex in itself; the challenge of studying organogenesis is to determine how all of them integrate to shape organs with recurring precision. This review focuses on the emerging understanding of how synchronized proliferation and differentiation of both somatic and germ cell lineages form 16-20 germ line stem cell (GSC) units in the ovary of Drosophila melanogaster. Recent work demonstrates that the Insulin, ecdysone, Epidermal Growth Factor, Decapentaplegic and Activin signaling pathways are used reiteratively for proliferation and differentiation in both somatic and germ cell lineages. This linkage underlies ovarian coordinated development and provides opportunity for correction mechanisms for stem cell unit numbers.
Collapse
Affiliation(s)
- Lilach Gilboa
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
37
|
Matias NR, Mathieu J, Huynh JR. Abscission is regulated by the ESCRT-III protein shrub in Drosophila germline stem cells. PLoS Genet 2015; 11:e1004653. [PMID: 25647097 PMCID: PMC4372032 DOI: 10.1371/journal.pgen.1004653] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 07/31/2014] [Indexed: 11/18/2022] Open
Abstract
Abscission is the final event of cytokinesis that leads to the physical separation of the two daughter cells. Recent technical advances have allowed a better understanding of the cellular and molecular events leading to abscission in isolated yeast or mammalian cells. However, how abscission is regulated in different cell types or in a developing organism remains poorly understood. Here, we characterized the function of the ESCRT-III protein Shrub during cytokinesis in germ cells undergoing a series of complete and incomplete divisions. We found that Shrub is required for complete abscission, and that levels of Shrub are critical for proper timing of abscission. Loss or gain of Shrub delays abscission in germline stem cells (GSCs), and leads to the formation of stem-cysts, where daughter cells share the same cytoplasm as the mother stem cell and cannot differentiate. In addition, our results indicate a negative regulation of Shrub by the Aurora B kinase during GSC abscission. Finally, we found that Lethal giant discs (lgd), known to be required for Shrub function in the endosomal pathway, also regulates the duration of abscission in GSCs. Abscission is the final step of cytokinesis which allows the physical separation of sister cells through the scission of a thin cytoplasmic bridge that links them at the end of mitosis. The duration of abscission varies depending on cell types, indicating that the event is developmentally regulated. Recently, we have identified two kinases, Aurora B and CycB/Cdk-1, which regulate the timing of abscission in germ cells and in mammalian cells. However, these kinases are upstream regulators and do not perform abscission per se. Here, we show that Shrub, a potential target of Aurora B and one of the most downstream effectors of abscission, is required for complete abscission in germline stem cells. In the absence of Shrub, the mother stem cell remains linked to its daughter cells, which then share the same cytoplasm and cannot differentiate. Loss of Shrub and Aurora B have opposite effects on abscission duration suggesting that Aurora B regulates negatively Shrub. We further show that Shrub acts together with its interactor Lethal giant disc to ensure proper abscission timing.
Collapse
Affiliation(s)
- Neuza Reis Matias
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
- CNRS UMR3215, Inserm U934, Paris, France
| | - Juliette Mathieu
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
- CNRS UMR3215, Inserm U934, Paris, France
- * E-mail: (JM); (JRH)
| | - Jean-René Huynh
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France
- CNRS UMR3215, Inserm U934, Paris, France
- * E-mail: (JM); (JRH)
| |
Collapse
|
38
|
Matsuoka S, Gupta S, Suzuki E, Hiromi Y, Asaoka M. gone early, a novel germline factor, ensures the proper size of the stem cell precursor pool in the Drosophila ovary. PLoS One 2014; 9:e113423. [PMID: 25420147 PMCID: PMC4242634 DOI: 10.1371/journal.pone.0113423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/27/2014] [Indexed: 01/06/2023] Open
Abstract
In order to sustain lifelong production of gametes, many animals have evolved a stem cell-based gametogenic program. In the Drosophila ovary, germline stem cells (GSCs) arise from a pool of primordial germ cells (PGCs) that remain undifferentiated even after gametogenesis has initiated. The decision of PGCs to differentiate or remain undifferentiated is regulated by somatic stromal cells: specifically, epidermal growth factor receptor (EGFR) signaling activated in the stromal cells determines the fraction of germ cells that remain undifferentiated by shaping a Decapentaplegic (Dpp) gradient that represses PGC differentiation. However, little is known about the contribution of germ cells to this process. Here we show that a novel germline factor, Gone early (Goe), limits the fraction of PGCs that initiate gametogenesis. goe encodes a non-peptidase homologue of the Neprilysin family metalloendopeptidases. At the onset of gametogenesis, Goe was localized on the germ cell membrane in the ovary, suggesting that it functions in a peptidase-independent manner in cell-cell communication at the cell surface. Overexpression of Goe in the germline decreased the number of PGCs that enter the gametogenic pathway, thereby increasing the proportion of undifferentiated PGCs. Inversely, depletion of Goe increased the number of PGCs initiating differentiation. Excess PGC differentiation in the goe mutant was augmented by halving the dose of argos, a somatically expressed inhibitor of EGFR signaling. This increase in PGC differentiation resulted in a massive decrease in the number of undifferentiated PGCs, and ultimately led to insufficient formation of GSCs. Thus, acting cooperatively with a somatic regulator of EGFR signaling, the germline factor goe plays a critical role in securing the proper size of the GSC precursor pool. Because goe can suppress EGFR signaling activity and is expressed in EGF-producing cells in various tissues, goe may function by attenuating EGFR signaling, and thereby affecting the stromal environment.
Collapse
Affiliation(s)
- Shinya Matsuoka
- Department of Developmental Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
- Department of Genetics, SOKENDAI, Mishima, Shizuoka, Japan
| | - Swati Gupta
- Department of Developmental Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Emiko Suzuki
- Department of Genetics, SOKENDAI, Mishima, Shizuoka, Japan
- Structural Biology Center, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Yasushi Hiromi
- Department of Developmental Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
- Department of Genetics, SOKENDAI, Mishima, Shizuoka, Japan
| | - Miho Asaoka
- Department of Developmental Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
- Department of Genetics, SOKENDAI, Mishima, Shizuoka, Japan
- * E-mail:
| |
Collapse
|
39
|
Abstract
It has long been established that germline stem cells (GSCs) are responsible for lifelong gametogenesis in males, and some female invertebrates (for example, Drosophila) and lower vertebrates (for example, teleost fish and some prosimians) also appear to rely on GSCs to replenish their oocyte reserve in adulthood. However, the presence of such cells in the majority of female mammals is controversial, and the idea of a fixed ovarian reserve determined at birth is the prevailing belief among reproductive biologists. However, accumulating evidence demonstrates the isolation and culture of putative GSCs from the ovaries of adult mice and humans. Live offspring have been reportedly produced from the culture of adult mouse GSCs, and human GSCs formed primordial follicles using a mouse xenograft model. If GSCs were present in adult female ovaries, it could be postulated that the occurrence of menopause is not due to the exhaustion of a fixed supply of oocytes but instead is a result of GSC and somatic cell aging. Alternatively, they may be benign under normal physiological conditions. If their existence were confirmed, female GSCs could have many potential applications in both basic science and clinical therapies. GSCs not only may provide a valuable model for germ cell development and maturation but may have a role in the field of fertility preservation, with women potentially being able to store GSCs or GSC-derived oocytes from their own ovaries prior to infertility-inducing treatments. Essential future work in this field will include further independent corroboration of the existence of GSCs in female mammals and the demonstration of the production of mature competent oocytes from GSCs cultured entirely in vitro.
Collapse
|
40
|
Functional analysis of the Drosophila embryonic germ cell transcriptome by RNA interference. PLoS One 2014; 9:e98579. [PMID: 24896584 PMCID: PMC4045815 DOI: 10.1371/journal.pone.0098579] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/05/2014] [Indexed: 11/19/2022] Open
Abstract
In Drosophila melanogaster, primordial germ cells are specified at the posterior pole of the very early embryo. This process is regulated by the posterior localized germ plasm that contains a large number of RNAs of maternal origin. Transcription in the primordial germ cells is actively down-regulated until germ cell fate is established. Bulk expression of the zygotic genes commences concomitantly with the degradation of the maternal transcripts. Thus, during embryogenesis, maternally provided and zygotically transcribed mRNAs determine germ cell development collectively. In an effort to identify novel genes involved in the regulation of germ cell behavior, we carried out a large-scale RNAi screen targeting both maternal and zygotic components of the embryonic germ line transcriptome. We identified 48 genes necessary for distinct stages in germ cell development. We found pebble and fascetto to be essential for germ cell migration and germ cell division, respectively. Our data uncover a previously unanticipated role of mei-P26 in maintenance of embryonic germ cell fate. We also performed systematic co-RNAi experiments, through which we found a low rate of functional redundancy among homologous gene pairs. As our data indicate a high degree of evolutionary conservation in genetic regulation of germ cell development, they are likely to provide valuable insights into the biology of the germ line in general.
Collapse
|
41
|
Herzig B, Yakulov TA, Klinge K, Günesdogan U, Jäckle H, Herzig A. Bällchen is required for self-renewal of germline stem cells in Drosophila melanogaster. Biol Open 2014; 3:510-21. [PMID: 24876388 PMCID: PMC4058086 DOI: 10.1242/bio.20147690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Self-renewing stem cells are pools of undifferentiated cells, which are maintained in cellular niche environments by distinct tissue-specific signalling pathways. In Drosophila melanogaster, female germline stem cells (GSCs) are maintained in a somatic niche of the gonads by BMP signalling. Here we report a novel function of the Drosophila kinase Bällchen (BALL), showing that its cell autonomous role is to maintain the self-renewing capacity of female GSCs independent of BMP signalling. ball mutant GSCs are eliminated from the niche and subsequently differentiate into mature eggs, indicating that BALL is largely dispensable for differentiation. Similar to female GSCs, BALL is required to maintain self-renewal of male GSCs, suggesting a tissue independent requirement of BALL for self-renewal of germline stem cells.
Collapse
Affiliation(s)
- Bettina Herzig
- Department of Molecular Developmental Biology, Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany Present address: Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany. Present address: Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK. Present address: Department of Cellular Microbiology, Max-Planck-Institut für Infektionsbiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Toma A Yakulov
- Present address: Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| | - Kathrin Klinge
- Department of Molecular Developmental Biology, Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany Present address: Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany. Present address: Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK. Present address: Department of Cellular Microbiology, Max-Planck-Institut für Infektionsbiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Ufuk Günesdogan
- Present address: Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Herbert Jäckle
- Department of Molecular Developmental Biology, Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany Present address: Renal Division, University Hospital Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany. Present address: Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK. Present address: Department of Cellular Microbiology, Max-Planck-Institut für Infektionsbiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Alf Herzig
- Present address: Department of Cellular Microbiology, Max-Planck-Institut für Infektionsbiologie, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
42
|
Deshpande G, Willis E, Chatterjee S, Fernandez R, Dias K, Schedl P. BMP signaling and the maintenance of primordial germ cell identity in Drosophila embryos. PLoS One 2014; 9:e88847. [PMID: 24551179 PMCID: PMC3925178 DOI: 10.1371/journal.pone.0088847] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 01/11/2014] [Indexed: 12/19/2022] Open
Abstract
The specification of primordial germ cells (PGCs) and subsequent maintenance of germ-line identity in Drosophila embryos has long been thought to occur solely under the control of cell-autonomous factors deposited in the posterior pole plasm during oogenesis. However, here we document a novel role for somatic BMP signaling in the maintenance of PGC fate during the period leading up to embryonic gonad coalescence. We find that PGCs fail to maintain their germline identity when BMP signaling is compromised. They initiate but are unable to properly assemble the germline stem cell-specific organelle, the spectrosome, and they lose expression of the germline-specific gene Vasa. BMP signaling must, however, be finely tuned as there are deleterious consequences to PGCs when the pathway is excessively active. We show that one mechanism used to calibrate the effects of BMP signals is dependent on the Ubc9 homolog Lesswright (Lwr).
Collapse
Affiliation(s)
- Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Elinor Willis
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Sandip Chatterjee
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Robert Fernandez
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Kristen Dias
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Paul Schedl
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- Institute of Gene Biology RAS, Moscow, Russian Federation
| |
Collapse
|
43
|
Durruthy Durruthy J, Ramathal C, Sukhwani M, Fang F, Cui J, Orwig KE, Reijo Pera RA. Fate of induced pluripotent stem cells following transplantation to murine seminiferous tubules. Hum Mol Genet 2014; 23:3071-84. [PMID: 24449759 PMCID: PMC4030765 DOI: 10.1093/hmg/ddu012] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Studies of human germ cell development are limited in large part by inaccessibility of germ cells during development. Moreover, although several studies have reported differentiation of mouse and human germ cells from pluripotent stem cells (PSCs) in vitro, differentiation of human germ cells from PSCs in vivo has not been reported. Here, we tested whether mRNA reprogramming in combination with xeno-transplantation may provide a viable system to probe the genetics of human germ cell development via use of induced pluripotent stem cells (iPSCs). For this purpose, we derived integration-free iPSCs via mRNA-based reprogramming with OCT3/4, SOX2, KLF4 and cMYC alone (OSKM) or in combination with the germ cell-specific mRNA, VASA (OSKMV). All iPSC lines met classic criteria of pluripotency. Moreover, global gene expression profiling did not distinguish large differences between undifferentiated OSKM and OSKMV iPSCs; however, some differences were observed in expression of pluripotency factors and germ cell-specific genes, and in epigenetic profiles and in vitro differentiation studies. In contrast, transplantation of undifferentiated iPSCs directly into the seminiferous tubules of germ cell-depleted immunodeficient mice revealed divergent fates of iPSCs produced with different factors. Transplantation resulted in morphologically and immunohistochemically recognizable germ cells in vivo, particularly in the case of OSKMV cells. Significantly, OSKMV cells also did not form tumors while OSKM cells that remained outside the seminiferous tubule proliferated extensively and formed tumors. Results indicate that mRNA reprogramming in combination with transplantation may contribute to tools for genetic analysis of human germ cell development.
Collapse
Affiliation(s)
- Jens Durruthy Durruthy
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Cyril Ramathal
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Fang Fang
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Jun Cui
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Renee A Reijo Pera
- Department of Genetics and Department of Obstetrics and Gynecology, Institute for Stem Cell Biology and Regenerative Medicine, Center for Reproductive and Stem Cell Biology, Stanford University, Stanford, CA 94305, USA and
| |
Collapse
|
44
|
Joly W, Chartier A, Rojas-Rios P, Busseau I, Simonelig M. The CCR4 deadenylase acts with Nanos and Pumilio in the fine-tuning of Mei-P26 expression to promote germline stem cell self-renewal. Stem Cell Reports 2013; 1:411-24. [PMID: 24286029 PMCID: PMC3841267 DOI: 10.1016/j.stemcr.2013.09.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 09/26/2013] [Accepted: 09/27/2013] [Indexed: 11/28/2022] Open
Abstract
Translational regulation plays an essential role in Drosophila ovarian germline stem cell (GSC) biology. GSC self-renewal requires two translational repressors, Nanos (Nos) and Pumilio (Pum), which repress the expression of differentiation factors in the stem cells. The molecular mechanisms underlying this translational repression remain unknown. Here, we show that the CCR4 deadenylase is required for GSC self-renewal and that Nos and Pum act through its recruitment onto specific mRNAs. We identify mei-P26 mRNA as a direct and major target of Nos/Pum/CCR4 translational repression in the GSCs. mei-P26 encodes a protein of the Trim-NHL tumor suppressor family that has conserved functions in stem cell lineages. We show that fine-tuning Mei-P26 expression by CCR4 plays a key role in GSC self-renewal. These results identify the molecular mechanism of Nos/Pum function in GSC self-renewal and reveal the role of CCR4-NOT-mediated deadenylation in regulating the balance between GSC self-renewal and differentiation. The CCR4 deadenylase is required for female germline stem cell self-renewal Nos/Pum recruit CCR4-NOT for translational repression in germline stem cells mei-P26 mRNA is a major target of translational repression by Nos/Pum/CCR4 Fine-tuning of mei-P26 by CCR4 is required for germline stem cell self-renewal
Collapse
Affiliation(s)
- Willy Joly
- mRNA Regulation and Development, Institute of Human Genetics, CNRS UPR1142, 141 Rue de la Cardonille, 34396 Montpellier Cedex 5, France
| | | | | | | | | |
Collapse
|
45
|
Mathieu J, Cauvin C, Moch C, Radford SJ, Sampaio P, Perdigoto CN, Schweisguth F, Bardin AJ, Sunkel CE, McKim K, Echard A, Huynh JR. Aurora B and cyclin B have opposite effects on the timing of cytokinesis abscission in Drosophila germ cells and in vertebrate somatic cells. Dev Cell 2013; 26:250-65. [PMID: 23948252 DOI: 10.1016/j.devcel.2013.07.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 04/10/2013] [Accepted: 07/09/2013] [Indexed: 01/28/2023]
Abstract
Abscission is the last step of cytokinesis that physically separates the cytoplasm of sister cells. As the final stage of cell division, abscission is poorly characterized during animal development. Here, we show that Aurora B and Survivin regulate the number of germ cells in each Drosophila egg chamber by inhibiting abscission during differentiation. This inhibition is mediated by an Aurora B-dependent phosphorylation of Cyclin B, as a phosphomimic form of Cyclin B rescues premature abscission caused by a loss of function of Aurora B. We show that Cyclin B localizes at the cytokinesis bridge, where it promotes abscission. We propose that mutual inhibitions between Aurora B and Cyclin B regulate the duration of abscission and thereby the number of sister cells in each cyst. Finally, we show that inhibitions of Aurora B and Cyclin-dependent kinase 1 activity in vertebrate cells also have opposite effects on the timing of abscission, suggesting a possible conservation of these mechanisms.
Collapse
Affiliation(s)
- Juliette Mathieu
- Department of Genetics and Developmental Biology, Institut Curie, F-75248 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Xin T, Xuan T, Tan J, Li M, Zhao G, Li M. The Drosophila putative histone acetyltransferase Enok maintains female germline stem cells through regulating Bruno and the niche. Dev Biol 2013; 384:1-12. [PMID: 24120347 DOI: 10.1016/j.ydbio.2013.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/29/2013] [Accepted: 10/02/2013] [Indexed: 12/16/2022]
Abstract
Maintenance of adult stem cells is largely dependent on the balance between their self-renewal and differentiation. The Drosophila ovarian germline stem cells (GSCs) provide a powerful in vivo system for studying stem cell fate regulation. It has been shown that maintaining the GSC population involves both genetic and epigenetic mechanisms. Although the role of epigenetic regulation in this process is evident, the underlying mechanisms remain to be further explored. In this study, we find that Enoki mushroom (Enok), a Drosophila putative MYST family histone acetyltransferase controls GSC maintenance in the ovary at multiple levels. Removal or knockdown of Enok in the germline causes a GSC maintenance defect. Further studies show that the cell-autonomous role of Enok in maintaining GSCs is not dependent on the BMP/Bam pathway. Interestingly, molecular studies reveal an ectopic expression of Bruno, an RNA binding protein, in the GSCs and their differentiating daughter cells elicited by the germline Enok deficiency. Misexpression of Bruno in GSCs and their immediate descendants results in a GSC loss that can be exacerbated by incorporating one copy of enok mutant allele. These data suggest a role for Bruno in Enok-controlled GSC maintenance. In addition, we observe that Enok is required for maintaining GSCs non-autonomously. Compromised expression of enok in the niche cells impairs the niche maintenance and BMP signal output, thereby causing defective GSC maintenance. This is the first demonstration that the niche size control requires an epigenetic mechanism. Taken together, studies in this paper provide new insights into the GSC fate regulation.
Collapse
Affiliation(s)
- Tianchi Xin
- MoE Key Laboratory of Developmental Genetics and Neuropsychiatric Diseases, Bio-X Institutes, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 200240 Shanghai, PR China
| | | | | | | | | | | |
Collapse
|
47
|
Gancz D, Gilboa L. Insulin and Target of rapamycin signaling orchestrate the development of ovarian niche-stem cell units in Drosophila. Development 2013; 140:4145-54. [PMID: 24026119 DOI: 10.1242/dev.093773] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue-specific stem cells and their niches are organized into functional units that respond to external cues in order to maintain organ homeostasis. Insulin and Target of rapamycin (Tor) signaling mediate external cues that control adult niches and stem cells. Whether these pathways play a role in the establishment of niche-stem cell units during organogenesis has been little explored. We show that during larval development both Insulin-like receptor (InR) and Tor participate in the establishment of ovarian niches and germline stem cells (GSCs) in Drosophila melanogaster. Tor and InR are required cell-autonomously for the proliferation of precursors for both somatic niches and GSCs. These pathways also promote the formation of terminal filaments (part of the somatic niche). Significantly, InR, but not Tor, signaling non-autonomously promotes primordial germ cell (PGC) differentiation. Somatic attenuation of the pathway retards PGC differentiation, whereas its activation results in their precocious differentiation. We also show that InR-mediated PGC differentiation is independent of somatic ecdysone signaling, but that further differentiation into cysts requires an ecdysone input. These results demonstrate that Tor and InR signaling actively participate in the formation of ovarian niches and stem cells by affecting both cell numbers and differentiation. The dual influence of Tor and InR on both somatic cells and PGCs ensures that these two cell populations develop coordinately. Our work further identifies a novel step in the regulation of germ cell differentiation by demonstrating that following bag of marbles expression, cyst formation requires an additional hormonal input.
Collapse
Affiliation(s)
- Dana Gancz
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
48
|
Barton LJ, Pinto BS, Wallrath LL, Geyer PK. The Drosophila nuclear lamina protein otefin is required for germline stem cell survival. Dev Cell 2013; 25:645-54. [PMID: 23806619 DOI: 10.1016/j.devcel.2013.05.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Revised: 01/11/2013] [Accepted: 05/23/2013] [Indexed: 01/27/2023]
Abstract
LEM domain (LEM-D) proteins are components of an extensive protein network that assembles beneath the inner nuclear envelope. Defects in LEM-D proteins cause tissue-restricted human diseases associated with altered stem cell homeostasis. Otefin (Ote) is a Drosophila LEM-D protein that is intrinsically required for female germline stem cell (GSC) maintenance. Previous studies linked Ote loss with transcriptional activation of the key differentiation gene bag-of-marbles (bam), leading to the model in which Ote tethers the bam gene to the nuclear periphery for gene silencing. Using genetic and phenotypic analyses of multiple ote(-/-) backgrounds, we obtained evidence that is inconsistent with this model. We show that bam repression is maintained in ote(-/-) GSCs and that germ cell loss persists in ote(-/-), bam(-/-) mutants, together demonstrating that GSC loss is independent of bam transcription. We show that the primary defect in ote(-/-) GSCs is a block of differentiation, which ultimately leads to germ cell death.
Collapse
Affiliation(s)
- Lacy J Barton
- Department of Biochemistry, University of Iowa, College of Medicine, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
49
|
Xuan T, Xin T, He J, Tan J, Gao Y, Feng S, He L, Zhao G, Li M. dBre1/dSet1-dependent pathway for histone H3K4 trimethylation has essential roles in controlling germline stem cell maintenance and germ cell differentiation in the Drosophila ovary. Dev Biol 2013; 379:167-81. [PMID: 23624310 DOI: 10.1016/j.ydbio.2013.04.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 03/27/2013] [Accepted: 04/13/2013] [Indexed: 11/28/2022]
Abstract
The Drosophila ovarian germline stem cells (GSCs) constantly experience self-renewal and differentiation, ensuring the female fertility throughout life. The balance between GSC self-renewal and differentiation is exquisitely regulated by the stem cell niche, the stem cells themselves and systemic factors. Increasing evidence has shown that the GSC regulation also involves epigenetic mechanisms including chromatin remodeling and histone modification. Here, we find that dBre1, an E3 ubiquitin ligase, functions in controlling GSC self-renewal and germ cell differentiation via distinct mechanisms. Removal or knock down of dBre1 function in the germline or somatic niche cell lineage leads to a gradual GSC loss and disruption of H3K4 trimethylation in the Drosophila ovary. Further studies suggest that the defective GSC maintenance is attributable to compromised BMP signaling emitted from the stem cell niche and impaired adhesion of GSCs to their niche. On the other hand, dBre1-RNAi expression in escort cells causes a loss of H3K4 trimethylation and accumulation of spectrosome-containing single germ cells in the germarium. Reducing dpp or dally levels suppresses the germ cell differentiation defects, indicating that dBre1 limits BMP signaling activities for the differentiation control. Strikingly, all phenotypes observed in dBre1 mutant ovaries can be mimicked by RNAi-based reduced expression of dSet1, a Drosophila H3K4 trimethylase. Moreover, genetic studies favor that dBre1 interacts with dSet1 in controlling GSC maintenance and germ cell differentiation. Taken together, we identify a dBre1/dSet1-dependent pathway for the H3K4 methylation involved in the cell fate regulation in the Drosophila ovary.
Collapse
Affiliation(s)
- Tao Xuan
- MoE Key Laboratory of Developmental Genetics and Neuropsychiatric Diseases, Bio-X Institutes, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 200240 Shanghai, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Matsuoka S, Hiromi Y, Asaoka M. Egfr signaling controls the size of the stem cell precursor pool in the Drosophila ovary. Mech Dev 2013; 130:241-53. [PMID: 23376160 DOI: 10.1016/j.mod.2013.01.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/11/2013] [Accepted: 01/15/2013] [Indexed: 12/29/2022]
Abstract
In many animals, germline progenitors are kept undifferentiated to give rise to germline stem cells (GSCs), enabling continuous production of gametes throughout animal life. In the Drosophila ovary, GSCs arise from a subset of primordial germ cells (PGCs) that stay undifferentiated even after gametogenesis has started. How a certain population of PGCs is protected against differentiation, and the significance of its regulatory mechanisms on GSC establishment remain elusive. Here we show that epidermal growth factor receptor (Egfr) signaling in somatic stromal intermingled cells (ICs), activated by its ligand produced in germ cells, controls the size of the PGC pool at the onset of gametogenesis. Egfr signaling in ICs limits the number of cells that express the heparan sulfate proteoglycan Dally, which is required for the movement and stability of the locally-produced stromal morphogen, Decapentaplegic (Dpp, a BMP2/4 homologue). Dpp is received by PGCs and maintains them in an undifferentiated state. Altering Egfr signaling levels changes the size of the PGC pool and affects the number of GSCs established during development. While excess GSC formation is compensated by the adult stage, insufficient GSC formation can lead to adult ovarioles that completely lack GSCs, suggesting that ensuring an absolute size of the PGC pool is crucial for the GSC system.
Collapse
Affiliation(s)
- Shinya Matsuoka
- Department of Developmental Genetics, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | | | | |
Collapse
|