1
|
Raz AA, Yamashita YM. Rewinding the clock: mechanisms of dedifferentiation. Curr Opin Genet Dev 2025; 93:102353. [PMID: 40311173 DOI: 10.1016/j.gde.2025.102353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 05/03/2025]
Abstract
Adult stem cells maintain tissue homeostasis through the production of differentiating cells. Considerable recent work has identified that stem cells themselves are replaceable through the process of dedifferentiation. The capacity and mechanisms of dedifferentiation vary widely among species and organ contexts. However, some core features are commonly present. In this review, we summarize 'hallmarks' of dedifferentiation, including mechanisms for maintenance of potency, sensation of loss, and migration, and review the current understanding of dedifferentiation as a true replacement mechanism.
Collapse
Affiliation(s)
- Amelie A Raz
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
| | - Yukiko M Yamashita
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Howard Hughes Medical Institute, Cambridge, MA, USA; Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
2
|
Powell AM, Williams AE, Ables ET. Fusome morphogenesis is sufficient to promote female germline stem cell self-renewal in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642432. [PMID: 40161740 PMCID: PMC11952372 DOI: 10.1101/2025.03.10.642432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Many tissue-resident stem cells are retained through asymmetric cell division, a process that ensures stem cell self-renewal through each mitotic cell cycle. Asymmetric organelle distribution has been proposed as a mechanism by which stem cells are marked for long-term retention; however, it is not clear whether biased organelle localization is a cause or an effect of asymmetric division. In Drosophila females, an endoplasmic reticulum-like organelle called the fusome is continually regenerated in germline stem cells (GSCs) and associated with GSC division. Here, we report that the β-importin Tnpo-SR is essential for fusome regeneration. Depletion of Tnpo-SR disrupts cytoskeletal organization during interphase and nuclear membrane remodeling during mitosis. Tnpo-SR does not localize to microtubules, centrosomes, or the fusome, suggesting that its role in maintaining these processes is indirect. Despite this, we find that restoring fusome morphogenesis in Tnpo-SR-depleted GSCs is sufficient to rescue GSC maintenance and cell cycle progression. We conclude that Tnpo-SR functionally fusome regeneration to cell cycle progression, supporting the model that asymmetric rebuilding of fusome promotes maintenance of GSC identity and niche retention.
Collapse
Affiliation(s)
- Amanda M. Powell
- Department of Biology, East Carolina University, Greenville, NC, 27858
| | - Anna E. Williams
- Department of Biology, East Carolina University, Greenville, NC, 27858
- Current address: Biochemistry, Cell and Developmental Biology Graduate Program, Emory University, Atlanta, GA, 30322
| | | |
Collapse
|
3
|
Kawasaki T, Nishimura T, Tani N, Ramos C, Karaulanov E, Shinya M, Saito K, Taylor E, Ketting RF, Ishiguro KI, Tanaka M, Siegfried KR, Sakai N. Meioc-Piwil1 complexes regulate rRNA transcription for differentiation of spermatogonial stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.17.623901. [PMID: 39605693 PMCID: PMC11601514 DOI: 10.1101/2024.11.17.623901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Ribosome biogenesis is vital for sustaining stem cell properties, yet its regulatory mechanisms are obscure. Herein, we show unique properties of zebrafish meioc mutants in which spermatogonial stem cells (SSCs) do not differentiate or upregulate rRNAs. Meioc colocalized with Piwil1 in perinuclear germ granules, but Meioc depletion resulted in Piwil1 accumulation in nucleoli. Nucleolar Piwil1 interacted with 45S pre-rRNA. piwil1 +/- spermatogonia with reduced Piwil1 upregulated rRNAs, and piwil1 +/- ;meioc -/- spermatogonia recovered differentiation later than those in meioc -/- . Further, Piwil1 interacted with Setdb1 and HP1α, and meioc -/- spermatogonia exhibited high levels of H3K9me3 and methylated CpG in the 45S-rDNA region. These results indicate that zebrafish SSCs maintain low levels of rRNA transcription with repressive marks similar to Drosophila piRNA targets of RNA polymerase II, and that Meioc has a unique function on preventing localization of Piwil1 in nucleoli to upregulate rRNA transcripts and to promote SSC differentiation.
Collapse
Affiliation(s)
- Toshihiro Kawasaki
- Department of Gene Function and Phenomics, National Institute of Genetics
- Department of Genetics, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Mishima 411-8540, Japan
| | - Toshiya Nishimura
- Division of Biological Science, Nagoya University, Nagoya 464-8601, Japan
| | - Naoki Tani
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Carina Ramos
- Biology Department, University of Massachusetts Boston, Boston, MA 02125
| | | | - Minori Shinya
- Department of Gene Function and Phenomics, National Institute of Genetics
- Department of Genetics, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Mishima 411-8540, Japan
| | - Kenji Saito
- Department of Gene Function and Phenomics, National Institute of Genetics
| | - Emily Taylor
- Biology Department, University of Massachusetts Boston, Boston, MA 02125
| | | | - Kei-ichiro Ishiguro
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Minoru Tanaka
- Division of Biological Science, Nagoya University, Nagoya 464-8601, Japan
| | | | - Noriyoshi Sakai
- Department of Gene Function and Phenomics, National Institute of Genetics
- Department of Genetics, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Mishima 411-8540, Japan
| |
Collapse
|
4
|
Jones SD, Miller JEB, Amos MM, Hernández JM, Piaszynski KM, Geyer PK. Emerin preserves stem cell survival through maintenance of centrosome and nuclear lamina structure. Development 2024; 151:dev204219. [PMID: 39465887 PMCID: PMC11586520 DOI: 10.1242/dev.204219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024]
Abstract
Drosophila female germline stem cells (GSCs) complete asymmetric mitosis in the presence of an intact, but permeable, nuclear envelope and nuclear lamina (NL). This asymmetric division requires a modified centrosome cycle, wherein mitotic centrosomes with mature pericentriolar material (PCM) embed in the NL and interphase centrosomes with reduced PCM leave the NL. This centrosome cycle requires Emerin, an NL protein required for GSC survival and germ cell differentiation. In emerin mutants, interphase GSC centrosomes retain excess PCM, remain embedded in the NL and nucleate microtubule asters at positions of NL distortion. Here, we investigate the contributions of abnormal interphase centrosomes to GSC loss. Remarkably, reducing interphase PCM in emerin mutants rescues GSC survival and partially restores germ cell differentiation. Direct tests of the effects of abnormal centrosomes were achieved by expression of constitutively active Polo kinase to drive enlargement of interphase centrosomes in wild-type GSCs. Notably, these conditions failed to alter NL structure or decrease GSC survival. However, coupling enlarged interphase centrosomes with nuclear distortion promoted GSC loss. These studies establish that Emerin maintains centrosome structure to preserve stem cell survival.
Collapse
Affiliation(s)
- Samuel D. Jones
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Jack E. B. Miller
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Madilynn M. Amos
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Julianna M. Hernández
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Katherine M. Piaszynski
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Pamela K. Geyer
- Department of Biochemistry and Molecular Biology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
5
|
Vallés AM, Rubin T, Macaisne N, Dal Toe L, Molla-Herman A, Antoniewski C, Huynh JR. Transcriptomic analysis of meiotic genes during the mitosis-to-meiosis transition in Drosophila females. Genetics 2024; 228:iyae130. [PMID: 39225982 DOI: 10.1093/genetics/iyae130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Germline cells produce gametes, which are specialized cells essential for sexual reproduction. Germline cells first amplify through several rounds of mitosis before switching to the meiotic program, which requires specific sets of proteins for DNA recombination, chromosome pairing, and segregation. Surprisingly, we previously found that some proteins of the synaptonemal complex, a prophase I meiotic structure, are already expressed and required in the mitotic region of Drosophila females. Here, to assess if additional meiotic genes were expressed earlier than expected, we isolated mitotic and meiotic cell populations to compare their RNA content. Our transcriptomic analysis reveals that all known meiosis I genes are already expressed in the mitotic region; however, only some of them are translated. As a case study, we focused on mei-W68, the Drosophila homolog of Spo11, to assess its expression at both the mRNA and protein levels and used different mutant alleles to assay for a premeiotic function. We could not detect any functional role for Mei-W68 during homologous chromosome pairing in dividing germ cells. Our study paves the way for further functional analysis of meiotic genes expressed in the mitotic region.
Collapse
Affiliation(s)
- Ana Maria Vallés
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Thomas Rubin
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Nicolas Macaisne
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Laurine Dal Toe
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Anahi Molla-Herman
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| | - Christophe Antoniewski
- ARTbio Bioinformatics Analysis Facility, IBPS, CNRS, Sorbonne Université, Institut Français de Bioinformatique, 75005 Paris, France
| | - Jean-René Huynh
- Center for Interdisciplinary Research in Biology, Collège de France, Université PSL, CNRS, INSERM, 75005 Paris, France
| |
Collapse
|
6
|
Samanta P, Ghosh R, Pakhira S, Mondal M, Biswas S, Sarkar R, Bhowmik A, Saha P, Hajra S. Ribosome biogenesis and ribosomal proteins in cancer stem cells: a new therapeutic prospect. Mol Biol Rep 2024; 51:1016. [PMID: 39325314 DOI: 10.1007/s11033-024-09963-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Ribosome has been considered as the fundamental macromolecular machine involved in protein synthesis in both prokaryotic and eukaryotic cells. This protein synthesis machinery consists of several rRNAs and numerous proteins. All of these factors are synthesized, translocated and assembled in a tightly regulated process known as ribosome biogenesis. Any impairment in this process causes development of several diseases like cancer. According to growing evidences, cancer cells display alteration of several ribosomal proteins. Besides, most of them are considered as key molecules involved in ribosome biogenesis, suggesting a correlation between those proteins and formation of ribosomes. Albeit, defective ribosome biogenesis in several cancers has gained prime importance, regulation of this process in cancer stem cells (CSCs) are still unrecognized. In this article, we aim to summarize the alteration of ribosome biogenesis and ribosomal proteins in CSCs. Moreover, we want to highlight the relation of ribosome biogenesis with hypoxia and drug resistance in CSCs based on the existing evidences. Lastly, this review wants to pay attention about the promising anti-cancer drugs which have potential to inhibit ribosome biogenesis in cancer cells as well as CSCs.
Collapse
Affiliation(s)
- Priya Samanta
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Rituparna Ghosh
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Shampa Pakhira
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Mrinmoyee Mondal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Souradeep Biswas
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Rupali Sarkar
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Subhadip Hajra
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
7
|
Wang H, Santuari L, Wijsman T, Wachsman G, Haase H, Nodine M, Scheres B, Heidstra R. Arabidopsis ribosomal RNA processing meerling mutants exhibit suspensor-derived polyembryony due to direct reprogramming of the suspensor. THE PLANT CELL 2024; 36:2550-2569. [PMID: 38513608 PMCID: PMC11218825 DOI: 10.1093/plcell/koae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Embryo development in Arabidopsis (Arabidopsis thaliana) starts off with an asymmetric division of the zygote to generate the precursors of the embryo proper and the supporting extraembryonic suspensor. The suspensor degenerates as the development of the embryo proper proceeds beyond the heart stage. Until the globular stage, the suspensor maintains embryonic potential and can form embryos in the absence of the developing embryo proper. We report a mutant called meerling-1 (mrl-1), which shows a high penetrance of suspensor-derived polyembryony due to delayed development of the embryo proper. Eventually, embryos from both apical and suspensor lineages successfully develop into normal plants and complete their life cycle. We identified the causal mutation as a genomic rearrangement altering the promoter of the Arabidopsis U3 SMALL NUCLEOLAR RNA-ASSOCIATED PROTEIN 18 (UTP18) homolog that encodes a nucleolar-localized WD40-repeat protein involved in processing 18S preribosomal RNA. Accordingly, root-specific knockout of UTP18 caused growth arrest and accumulation of unprocessed 18S pre-rRNA. We generated the mrl-2 loss-of-function mutant and observed asynchronous megagametophyte development causing embryo sac abortion. Together, our results indicate that promoter rearrangement decreased UTP18 protein abundance during early stage embryo proper development, triggering suspensor-derived embryogenesis. Our data support the existence of noncell autonomous signaling from the embryo proper to prevent direct reprogramming of the suspensor toward embryonic fate.
Collapse
Affiliation(s)
- Honglei Wang
- Cluster of Plant Developmental Biology, Laboratory of Cell and Developmental Biology, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Luca Santuari
- Cluster of Plant Developmental Biology, Laboratory of Molecular Biology, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Tristan Wijsman
- Cluster of Plant Developmental Biology, Laboratory of Cell and Developmental Biology, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Guy Wachsman
- Molecular Genetics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Hannah Haase
- Cluster of Plant Developmental Biology, Laboratory of Molecular Biology, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Michael Nodine
- Cluster of Plant Developmental Biology, Laboratory of Molecular Biology, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Ben Scheres
- Cluster of Plant Developmental Biology, Laboratory of Molecular Biology, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Renze Heidstra
- Cluster of Plant Developmental Biology, Laboratory of Cell and Developmental Biology, Wageningen University & Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
8
|
Abstract
Although differential transcription drives the development of multicellular organisms, the ultimate readout of a protein-coding gene is ribosome-dependent mRNA translation. Ribosomes were once thought of as uniform molecular machines, but emerging evidence indicates that the complexity and diversity of ribosome biogenesis and function should be given a fresh look in the context of development. This Review begins with a discussion of different developmental disorders that have been linked with perturbations in ribosome production and function. We then highlight recent studies that reveal how different cells and tissues exhibit variable levels of ribosome production and protein synthesis, and how changes in protein synthesis capacity can influence specific cell fate decisions. We finish by touching upon ribosome heterogeneity in stress responses and development. These discussions highlight the importance of considering both ribosome levels and functional specialization in the context of development and disease.
Collapse
Affiliation(s)
- Chunyang Ni
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
9
|
Ni C, Buszczak M. The homeostatic regulation of ribosome biogenesis. Semin Cell Dev Biol 2023; 136:13-26. [PMID: 35440410 PMCID: PMC9569395 DOI: 10.1016/j.semcdb.2022.03.043] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/22/2022]
Abstract
The continued integrity of biological systems depends on a balance between interdependent elements at the molecular, cellular, and organismal levels. This is particularly true for the generation of ribosomes, which influence almost every aspect of cell and organismal biology. Ribosome biogenesis (RiBi) is an energetically demanding process that involves all three RNA polymerases, numerous RNA processing factors, chaperones, and the coordinated expression of 79-80 ribosomal proteins (r-proteins). Work over the last several decades has revealed that the dynamic regulation of ribosome production represents a major mechanism by which cells maintain homeostasis in response to changing environmental conditions and acute stress. More recent studies suggest that cells and tissues within multicellular organisms exhibit dramatically different levels of ribosome production and protein synthesis, marked by the differential expression of RiBi factors. Thus, distinct bottlenecks in the RiBi process, downstream of rRNA transcription, may exist within different cell populations of multicellular organisms during development and in adulthood. This review will focus on our current understanding of the mechanisms that link the complex molecular process of ribosome biogenesis with cellular and organismal physiology. We will discuss diverse topics including how different steps in the RiBi process are coordinated with one another, how MYC and mTOR impact RiBi, and how RiBi levels change between stem cells and their differentiated progeny. In turn, we will also review how regulated changes in ribosome production itself can feedback to influence cell fate and function.
Collapse
Affiliation(s)
- Chunyang Ni
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA.
| |
Collapse
|
10
|
Breznak SM, Kotb NM, Rangan P. Dynamic regulation of ribosome levels and translation during development. Semin Cell Dev Biol 2023; 136:27-37. [PMID: 35725716 DOI: 10.1016/j.semcdb.2022.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/20/2022] [Accepted: 06/12/2022] [Indexed: 01/11/2023]
Abstract
The ability of ribosomes to translate mRNAs into proteins is the basis of all life. While ribosomes are essential for cell viability, reduction in levels of ribosomes can affect cell fate and developmental transitions in a tissue specific manner and can cause a plethora of related diseases called ribosomopathies. How dysregulated ribosomes homeostasis influences cell fate and developmental transitions is not fully understood. Model systems such as Drosophila and C. elegans oogenesis have been used to address these questions since defects in conserved steps in ribosome biogenesis result in stem cell differentiation and developmental defects. In this review, we first explore how ribosome levels affect stem cell differentiation. Second, we describe how ribosomal modifications and incorporation of ribosomal protein paralogs contribute to development. Third, we summarize how cells with perturbed ribosome biogenesis are sensed and eliminated during organismal growth.
Collapse
Affiliation(s)
- Shane M Breznak
- Department of Biological Sciences/RNA Institute, University at Albany SUNY, Albany, NY, 12222, USA
| | - Noor M Kotb
- Department of Biomedical Sciences, The School of Public Health, University at Albany SUNY, 11 Albany, NY 12222, USA
| | - Prashanth Rangan
- Department of Cell, Developmental, and Regenerative Biology, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
11
|
Marchetti M, Zhang C, Edgar BA. An improved organ explant culture method reveals stem cell lineage dynamics in the adult Drosophila intestine. eLife 2022; 11:e76010. [PMID: 36005292 PMCID: PMC9578704 DOI: 10.7554/elife.76010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
In recent years, live-imaging techniques have been developed for the adult midgut of Drosophila melanogaster that allow temporal characterization of key processes involved in stem cell and tissue homeostasis. However, these organ culture techniques have been limited to imaging sessions of <16 hours, an interval too short to track dynamic processes such as damage responses and regeneration, which can unfold over several days. Therefore, we developed an organ explant culture protocol capable of sustaining midguts ex vivo for up to 3 days. This was made possible by the formulation of a culture medium specifically designed for adult Drosophila tissues with an increased Na+/K+ ratio and trehalose concentration, and by placing midguts at an air-liquid interface for enhanced oxygenation. We show that midgut progenitor cells can respond to gut epithelial damage ex vivo, proliferating and differentiating to replace lost cells, but are quiescent in healthy intestines. Using ex vivo gene induction to promote stem cell proliferation using RasG12V or string and Cyclin E overexpression, we demonstrate that progenitor cell lineages can be traced through multiple cell divisions using live imaging. We show that the same culture set-up is useful for imaging adult renal tubules and ovaries for up to 3 days and hearts for up to 10 days. By enabling both long-term imaging and real-time ex vivo gene manipulation, our simple culture protocol provides a powerful tool for studies of epithelial biology and cell lineage behavior.
Collapse
Affiliation(s)
- Marco Marchetti
- Department of Oncological Sciences, Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Chenge Zhang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Bruce A Edgar
- Department of Oncological Sciences, Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| |
Collapse
|
12
|
Upregulation of ribosome biogenesis via canonical E-boxes is required for Myc-driven proliferation. Dev Cell 2022; 57:1024-1036.e5. [PMID: 35472319 DOI: 10.1016/j.devcel.2022.03.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 12/29/2021] [Accepted: 03/25/2022] [Indexed: 11/23/2022]
Abstract
The transcription factor Myc drives cell growth across animal phyla and is activated in most forms of human cancer. However, it is unclear which Myc target genes need to be regulated to induce growth and whether multiple targets act additively or if induction of each target is individually necessary. Here, we identified Myc target genes whose regulation is conserved between humans and flies and deleted Myc-binding sites (E-boxes) in the promoters of fourteen of these genes in Drosophila. E-box mutants of essential genes were homozygous viable, indicating that the E-boxes are not required for basal expression. Eight E-box mutations led to Myc-like phenotypes; the strongest mutant, ppanEbox-/-, also made the flies resistant to Myc-induced cell growth without affecting Myc-induced apoptosis. The ppanEbox-/- flies are healthy and display only a minor developmental delay, suggesting that it may be possible to treat or prevent tumorigenesis by targeting individual downstream targets of Myc.
Collapse
|
13
|
Mercer M, Jang S, Ni C, Buszczak M. The Dynamic Regulation of mRNA Translation and Ribosome Biogenesis During Germ Cell Development and Reproductive Aging. Front Cell Dev Biol 2021; 9:710186. [PMID: 34805139 PMCID: PMC8595405 DOI: 10.3389/fcell.2021.710186] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/07/2021] [Indexed: 01/21/2023] Open
Abstract
The regulation of mRNA translation, both globally and at the level of individual transcripts, plays a central role in the development and function of germ cells across species. Genetic studies using flies, worms, zebrafish and mice have highlighted the importance of specific RNA binding proteins in driving various aspects of germ cell formation and function. Many of these mRNA binding proteins, including Pumilio, Nanos, Vasa and Dazl have been conserved through evolution, specifically mark germ cells, and carry out similar functions across species. These proteins typically influence mRNA translation by binding to specific elements within the 3′ untranslated region (UTR) of target messages. Emerging evidence indicates that the global regulation of mRNA translation also plays an important role in germ cell development. For example, ribosome biogenesis is often regulated in a stage specific manner during gametogenesis. Moreover, oocytes need to produce and store a sufficient number of ribosomes to support the development of the early embryo until the initiation of zygotic transcription. Accumulating evidence indicates that disruption of mRNA translation regulatory mechanisms likely contributes to infertility and reproductive aging in humans. These findings highlight the importance of gaining further insights into the mechanisms that control mRNA translation within germ cells. Future work in this area will likely have important impacts beyond germ cell biology.
Collapse
Affiliation(s)
- Marianne Mercer
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Seoyeon Jang
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Chunyang Ni
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michael Buszczak
- Department of Molecular Biology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
14
|
Jang S, Lee J, Mathews J, Ruess H, Williford AO, Rangan P, Betrán E, Buszczak M. The Drosophila ribosome protein S5 paralog RpS5b promotes germ cell and follicle cell differentiation during oogenesis. Development 2021; 148:272089. [PMID: 34495316 DOI: 10.1242/dev.199511] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/01/2021] [Indexed: 01/15/2023]
Abstract
Emerging evidence suggests that ribosome heterogeneity may have important functional consequences in the translation of specific mRNAs within different cell types and under various conditions. Ribosome heterogeneity comes in many forms, including post-translational modification of ribosome proteins (RPs), absence of specific RPs and inclusion of different RP paralogs. The Drosophila genome encodes two RpS5 paralogs: RpS5a and RpS5b. While RpS5a is ubiquitously expressed, RpS5b exhibits enriched expression in the reproductive system. Deletion of RpS5b results in female sterility marked by developmental arrest of egg chambers at stages 7-8, disruption of vitellogenesis and posterior follicle cell (PFC) hyperplasia. While transgenic rescue experiments suggest functional redundancy between RpS5a and RpS5b, molecular, biochemical and ribo-seq experiments indicate that RpS5b mutants display increased rRNA transcription and RP production, accompanied by increased protein synthesis. Loss of RpS5b results in microtubule-based defects and in mislocalization of Delta and Mindbomb1, leading to failure of Notch pathway activation in PFCs. Together, our results indicate that germ cell-specific expression of RpS5b promotes proper egg chamber development by ensuring the homeostasis of functional ribosomes.
Collapse
Affiliation(s)
- Seoyeon Jang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeon Lee
- Lydia Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeremy Mathews
- Lydia Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Holly Ruess
- Lydia Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anna O Williford
- Department of Biology, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Prashanth Rangan
- RNA Institute, Department of Biological Sciences, University at Albany, SUNY, Albany, NY 12222, USA
| | - Esther Betrán
- Department of Biology, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Drosophila female germline stem cells undergo mitosis without nuclear breakdown. Curr Biol 2021; 31:1450-1462.e3. [PMID: 33548191 DOI: 10.1016/j.cub.2021.01.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/18/2020] [Accepted: 01/11/2021] [Indexed: 02/02/2023]
Abstract
Stem cell homeostasis requires nuclear lamina (NL) integrity. In Drosophila germ cells, compromised NL integrity activates the ataxia telangiectasia and Rad3-related (ATR) and checkpoint kinase 2 (Chk2) checkpoint kinases, blocking germ cell differentiation and causing germline stem cell (GSC) loss. Checkpoint activation occurs upon loss of either the NL protein emerin or its partner barrier-to-autointegration factor, two proteins required for nuclear reassembly at the end of mitosis. Here, we examined how mitosis contributes to NL structural defects linked to checkpoint activation. These analyses led to the unexpected discovery that wild-type female GSCs utilize a non-canonical mode of mitosis, one that retains a permeable but intact nuclear envelope and NL. We show that the interphase NL is remodeled during mitosis for insertion of centrosomes that nucleate the mitotic spindle within the confines of the nucleus. We show that depletion or loss of NL components causes mitotic defects, including compromised chromosome segregation associated with altered centrosome positioning and structure. Further, in emerin mutant GSCs, centrosomes remain embedded in the interphase NL. Notably, these embedded centrosomes carry large amounts of pericentriolar material and nucleate astral microtubules, revealing a role for emerin in the regulation of centrosome structure. Epistasis studies demonstrate that defects in centrosome structure are upstream of checkpoint activation, suggesting that these centrosome defects might trigger checkpoint activation and GSC loss. Connections between NL proteins and centrosome function have implications for mechanisms associated with NL dysfunction in other stem cell populations, including NL-associated diseases, such as laminopathies.
Collapse
|
16
|
Hinnant TD, Merkle JA, Ables ET. Coordinating Proliferation, Polarity, and Cell Fate in the Drosophila Female Germline. Front Cell Dev Biol 2020; 8:19. [PMID: 32117961 PMCID: PMC7010594 DOI: 10.3389/fcell.2020.00019] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/10/2020] [Indexed: 01/05/2023] Open
Abstract
Gametes are highly specialized cell types produced by a complex differentiation process. Production of viable oocytes requires a series of precise and coordinated molecular events. Early in their development, germ cells are an interconnected group of mitotically dividing cells. Key regulatory events lead to the specification of mature oocytes and initiate a switch to the meiotic cell cycle program. Though the chromosomal events of meiosis have been extensively studied, it is unclear how other aspects of oocyte specification are temporally coordinated. The fruit fly, Drosophila melanogaster, has long been at the forefront as a model system for genetics and cell biology research. The adult Drosophila ovary continuously produces germ cells throughout the organism’s lifetime, and many of the cellular processes that occur to establish oocyte fate are conserved with mammalian gamete development. Here, we review recent discoveries from Drosophila that advance our understanding of how early germ cells balance mitotic exit with meiotic initiation. We discuss cell cycle control and establishment of cell polarity as major themes in oocyte specification. We also highlight a germline-specific organelle, the fusome, as integral to the coordination of cell division, cell polarity, and cell fate in ovarian germ cells. Finally, we discuss how the molecular controls of the cell cycle might be integrated with cell polarity and cell fate to maintain oocyte production.
Collapse
Affiliation(s)
- Taylor D Hinnant
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Julie A Merkle
- Department of Biology, University of Evansville, Evansville, IN, United States
| | - Elizabeth T Ables
- Department of Biology, East Carolina University, Greenville, NC, United States
| |
Collapse
|
17
|
Duan T, Green N, Tootle TL, Geyer PK. Nuclear architecture as an intrinsic regulator of Drosophila female germline stem cell maintenance. CURRENT OPINION IN INSECT SCIENCE 2020; 37:30-38. [PMID: 32087561 PMCID: PMC7089816 DOI: 10.1016/j.cois.2019.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/08/2019] [Accepted: 11/13/2019] [Indexed: 05/08/2023]
Abstract
Homeostasis of Drosophila germline stem cells (GSC) depends upon the integration of intrinsic and extrinsic signals. This review highlights emerging data that support nuclear architecture as an intrinsic regulator of GSC maintenance and germ cell differentiation. Here, we focus on the nuclear lamina (NL) and the nucleolus, two compartments that undergo alterations in composition upon germ cell differentiation. Loss of NL or nucleolar components leads to GSC loss, resulting from activation of GSC quality control checkpoint pathways. We suggest that the NL and nucleolus integrate signals needed for the switch between GSC maintenance and germ cell differentiation, and propose regulation of nuclear actin pools as one mechanism that connects these compartments.
Collapse
Affiliation(s)
- Tingting Duan
- Departments of Biochemistry, University of Iowa, College of Medicine, Iowa City, IA 52242, USA
| | - Nicole Green
- Anatomy and Cell Biology, University of Iowa, College of Medicine, Iowa City, IA 52242, USA
| | - Tina L Tootle
- Anatomy and Cell Biology, University of Iowa, College of Medicine, Iowa City, IA 52242, USA
| | - Pamela K Geyer
- Departments of Biochemistry, University of Iowa, College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
18
|
Koyama LAJ, Aranda-Díaz A, Su YH, Balachandra S, Martin JL, Ludington WB, Huang KC, O'Brien LE. Bellymount enables longitudinal, intravital imaging of abdominal organs and the gut microbiota in adult Drosophila. PLoS Biol 2020; 18:e3000567. [PMID: 31986129 PMCID: PMC7004386 DOI: 10.1371/journal.pbio.3000567] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/06/2020] [Accepted: 01/13/2020] [Indexed: 12/30/2022] Open
Abstract
Cell- and tissue-level processes often occur across days or weeks, but few imaging methods can capture such long timescales. Here, we describe Bellymount, a simple, noninvasive method for longitudinal imaging of the Drosophila abdomen at subcellular resolution. Bellymounted animals remain live and intact, so the same individual can be imaged serially to yield vivid time series of multiday processes. This feature opens the door to longitudinal studies of Drosophila internal organs in their native context. Exploiting Bellymount's capabilities, we track intestinal stem cell lineages and gut microbial colonization in single animals, revealing spatiotemporal dynamics undetectable by previously available methods.
Collapse
Affiliation(s)
- Leslie Ann Jaramillo Koyama
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, United States of America.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Andrés Aranda-Díaz
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - Yu-Han Su
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Shruthi Balachandra
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Judy Lisette Martin
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, United States of America
| | - William B Ludington
- Department of Embryology, Carnegie Institution of Washington, Baltimore, Maryland, United States of America
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, California, United States of America.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America.,Chan Zuckerberg Biohub, San Francisco, California, United States of America
| | - Lucy Erin O'Brien
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
19
|
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: 1.8] [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
|
20
|
Drummond-Barbosa D. Local and Physiological Control of Germline Stem Cell Lineages in Drosophila melanogaster. Genetics 2019; 213:9-26. [PMID: 31488592 PMCID: PMC6727809 DOI: 10.1534/genetics.119.300234] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
The long-term survival of any multicellular species depends on the success of its germline in producing high-quality gametes and maximizing survival of the offspring. Studies in Drosophila melanogaster have led our growing understanding of how germline stem cell (GSC) lineages maintain their function and adjust their behavior according to varying environmental and/or physiological conditions. This review compares and contrasts the local regulation of GSCs by their specialized microenvironments, or niches; discusses how diet and diet-dependent factors, mating, and microorganisms modulate GSCs and their developing progeny; and briefly describes the tie between physiology and development during the larval phase of the germline cycle. Finally, it concludes with broad comparisons with other organisms and some future directions for further investigation.
Collapse
Affiliation(s)
- Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
21
|
Teixeira FK, Lehmann R. Translational Control during Developmental Transitions. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a032987. [PMID: 30082467 DOI: 10.1101/cshperspect.a032987] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The many steps of gene expression, from the transcription of a gene to the production of its protein product, are well understood. Yet, transcriptional regulation has been the focal point for the study of gene expression during development. However, quantitative studies reveal that messenger RNA (mRNA) levels are not necessarily good predictors of the respective proteins' levels in a cell. This discrepancy is, at least in part, the result of developmentally regulated, translational mechanisms that control the spatiotemporal regulation of gene expression. In this review, we focus on translational regulatory mechanisms mediating global transitions in gene expression: the shift from the maternal to the embryonic developmental program in the early embryo and the switch from the self-renewal of stem cells to differentiation in the adult.
Collapse
Affiliation(s)
| | - 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, New York 10016
| |
Collapse
|
22
|
Reilein A, Cimetta E, Tandon NM, Kalderon D, Vunjak-Novakovic G. Live imaging of stem cells in the germarium of the Drosophila ovary using a reusable gas-permeable imaging chamber. Nat Protoc 2019; 13:2601-2614. [PMID: 30349048 DOI: 10.1038/s41596-018-0054-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Live imaging of stem cells and their support cells can be used to visualize cellular dynamics and fluctuations of intracellular signals, proteins, and organelles in order to better understand stem cell behavior in the niche. We describe a simple protocol for imaging stem cells in the Drosophila ovary that improves on alternative protocols in that flies of any age can be used, dissection is simplified because the epithelial sheath that surrounds each ovariole need not be removed, and ovarioles are imaged in a closed chamber with a large volume of medium that buffers oxygen, pH, and temperature. We also describe how to construct the imaging chamber, which can be easily modified and used to image other tissues and non-adherent cells. Imaging is limited by follicle cells moving out of the germarium in culture around the time of egg chamber budding; however, the epithelial sheath delays this abnormal cell migration. This protocol requires an hour to prepare the ovarioles, followed by half an hour on the confocal microscope to locate germaria and set z limits. Successful imaging time depends on germarial morphology at the time of dissection, but we suggest 10-11 h to encompass all specimens.
Collapse
Affiliation(s)
- Amy Reilein
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Elisa Cimetta
- Department of Industrial Engineering (DII), Padova University, Padua, Italy. .,Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Corso Stati Uniti, Padua, Italy.
| | - Nina M Tandon
- EpiBone, Inc., Brooklyn, NY, USA.,Department of Electrical Engineering, The Cooper Union for the Advancement of Science and Art, New York, NY, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | | |
Collapse
|
23
|
Jarzebowski L, Le Bouteiller M, Coqueran S, Raveux A, Vandormael-Pournin S, David A, Cumano A, Cohen-Tannoudji M. Mouse adult hematopoietic stem cells actively synthesize ribosomal RNA. RNA (NEW YORK, N.Y.) 2018; 24:1803-1812. [PMID: 30242063 PMCID: PMC6239186 DOI: 10.1261/rna.067843.118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/14/2018] [Indexed: 06/08/2023]
Abstract
The contribution of basal cellular processes to the regulation of tissue homeostasis has just started to be appreciated. However, our knowledge of the modulation of ribosome biogenesis activity in situ within specific lineages remains very limited. This is largely due to the lack of assays that enable quantitation of ribosome biogenesis in small numbers of cells in vivo. We used a technique, named Flow-FISH, combining cell surface antibody staining and flow cytometry with intracellular ribosomal RNA (rRNA) FISH, to measure the levels of pre-rRNAs of hematopoietic cells in vivo. Here, we show that Flow-FISH reports and quantifies ribosome biogenesis activity in hematopoietic cell populations, thereby providing original data on this fundamental process notably in rare populations such as hematopoietic stem and progenitor cells. We unravel variations in pre-rRNA levels between different hematopoietic progenitor compartments and during erythroid differentiation. In particular, our data indicate that, contrary to what may be anticipated from their quiescent state, hematopoietic stem cells have significant ribosome biogenesis activity. Moreover, variations in pre-rRNA levels do not correlate with proliferation rates, suggesting that cell type-specific mechanisms might regulate ribosome biogenesis in hematopoietic stem cells and progenitors. Our study contributes to a better understanding of the cellular physiology of the hematopoietic system in vivo in unperturbed situations.
Collapse
Affiliation(s)
- Léonard Jarzebowski
- Early Mammalian Development and Stem Cell Biology, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Marie Le Bouteiller
- Early Mammalian Development and Stem Cell Biology, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Sabrina Coqueran
- Early Mammalian Development and Stem Cell Biology, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Aurélien Raveux
- Early Mammalian Development and Stem Cell Biology, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Sandrine Vandormael-Pournin
- Early Mammalian Development and Stem Cell Biology, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| | - Alexandre David
- Team "Signaling and Cancer," Institut de Génomique Fonctionnelle, Montpellier 34094, France
| | - Ana Cumano
- Lymphocyte Development Unit, Institut Pasteur, Paris 75015, France
| | - Michel Cohen-Tannoudji
- Early Mammalian Development and Stem Cell Biology, Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris 75015, France
- CNRS UMR 3738, Institut Pasteur, Paris 75015, France
| |
Collapse
|
24
|
Barui A, Datta P. Biophysical factors in the regulation of asymmetric division of stem cells. Biol Rev Camb Philos Soc 2018; 94:810-827. [PMID: 30467934 DOI: 10.1111/brv.12479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/14/2018] [Accepted: 10/18/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Ananya Barui
- Centre for Healthcare Science and TechnologyIndian Institute of Engineering Science and Technology, Shibpur Howrah West Bengal 711103 India
| | - Pallab Datta
- Centre for Healthcare Science and TechnologyIndian Institute of Engineering Science and Technology, Shibpur Howrah West Bengal 711103 India
| |
Collapse
|
25
|
Martin JL, Sanders EN, Moreno-Roman P, Jaramillo Koyama LA, Balachandra S, Du X, O'Brien LE. Long-term live imaging of the Drosophila adult midgut reveals real-time dynamics of division, differentiation and loss. eLife 2018; 7:36248. [PMID: 30427308 PMCID: PMC6277200 DOI: 10.7554/elife.36248] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/12/2018] [Indexed: 12/18/2022] Open
Abstract
Organ renewal is governed by the dynamics of cell division, differentiation and loss. To study these dynamics in real time, we present a platform for extended live imaging of the adult Drosophila midgut, a premier genetic model for stem-cell-based organs. A window cut into a living animal allows the midgut to be imaged while intact and physiologically functioning. This approach prolongs imaging sessions to 12–16 hr and yields movies that document cell and tissue dynamics at vivid spatiotemporal resolution. By applying a pipeline for movie processing and analysis, we uncover new and intriguing cell behaviors: that mitotic stem cells dynamically re-orient, that daughter cells use slow kinetics of Notch activation to reach a fate-specifying threshold, and that enterocytes extrude via ratcheted constriction of a junctional ring. By enabling real-time study of midgut phenomena that were previously inaccessible, our platform opens a new realm for dynamic understanding of adult organ renewal.
Collapse
Affiliation(s)
- Judy Lisette Martin
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Erin Nicole Sanders
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States
| | - Paola Moreno-Roman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Department of Biology, Stanford University, Stanford, United States
| | - Leslie Ann Jaramillo Koyama
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States.,Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States
| | - Shruthi Balachandra
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - XinXin Du
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| | - Lucy Erin O'Brien
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
26
|
Nguyen PD, Currie PD. In vivo imaging: shining a light on stem cells in the living animal. Development 2018; 145:145/7/dev150441. [DOI: 10.1242/dev.150441] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Stem cells are undifferentiated cells that play crucial roles during development, growth and regeneration. Traditionally, these cells have been primarily characterised by histology, cell sorting, cell culture and ex vivo methods. However, as stem cells interact in a complex environment within specific tissue niches, there has been increasing interest in examining their in vivo behaviours, particularly in response to injury. Advances in imaging technologies and genetic tools have converged to enable unprecedented access to the endogenous stem cell niche. In this Spotlight article, we highlight how in vivo imaging can probe a range of biological processes that relate to stem cell activity, behaviour and control.
Collapse
Affiliation(s)
- Phong Dang Nguyen
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Cancer Genomics Netherlands, 3584 CT Utrecht, The Netherlands
| | - Peter David Currie
- Australian Regenerative Medicine Institute, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
- EMBL Australia, Level 1, 15 Innovation Walk, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| |
Collapse
|
27
|
Stypulkowski E, Asangani IA, Witze ES. The depalmitoylase APT1 directs the asymmetric partitioning of Notch and Wnt signaling during cell division. Sci Signal 2018; 11:eaam8705. [PMID: 29295957 PMCID: PMC5914505 DOI: 10.1126/scisignal.aam8705] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Asymmetric cell division results in two distinctly fated daughter cells. A molecular hallmark of asymmetric division is the unequal partitioning of cell fate determinants. We have previously established that growth factor signaling promotes protein depalmitoylation to foster polarized protein localization, which, in turn, drives migration and metastasis. We report protein palmitoylation as a key mechanism for the asymmetric partitioning of the cell fate determinants Numb and β-catenin through the activity of the depalmitoylating enzyme APT1. Using point mutations, we showed that specific palmitoylated residues on Numb were required for its asymmetric localization. By live-cell imaging, we showed that reciprocal interactions between APT1 and the Rho family GTPase CDC42 promoted the asymmetric localization of Numb and β-catenin to the plasma membrane. This, in turn, restricted Notch- or Wnt-responsive transcriptional activity to one daughter cell. Moreover, we showed that altering APT1 abundance changed the transcriptional signatures of MDA-MB-231 triple receptor-negative breast cancer cells, similar to changes in Notch and β-catenin-mediated Wnt signaling. We also showed that loss of APT1 depleted a specific subpopulation of tumorigenic cells in colony formation assays. Together, our findings suggest that APT1-mediated depalmitoylation is a major mechanism of asymmetric cell division that maintains Notch- and Wnt-associated protein dynamics, gene expression, and cellular functions.
Collapse
Affiliation(s)
- Ewa Stypulkowski
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Irfan A Asangani
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric S Witze
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Noormohammadi A, Calculli G, Gutierrez-Garcia R, Khodakarami A, Koyuncu S, Vilchez D. Mechanisms of protein homeostasis (proteostasis) maintain stem cell identity in mammalian pluripotent stem cells. Cell Mol Life Sci 2018; 75:275-290. [PMID: 28748323 PMCID: PMC11105389 DOI: 10.1007/s00018-017-2602-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 01/10/2023]
Abstract
Protein homeostasis, or proteostasis, is essential for cell function, development, and organismal viability. The composition of the proteome is adjusted to the specific requirements of a particular cell type and status. Moreover, multiple metabolic and environmental conditions challenge the integrity of the proteome. To maintain the quality of the proteome, the proteostasis network monitors proteins from their synthesis through their degradation. Whereas somatic stem cells lose their ability to maintain proteostasis with age, immortal pluripotent stem cells exhibit a stringent proteostasis network associated with their biological function and intrinsic characteristics. Moreover, growing evidence indicates that enhanced proteostasis mechanisms play a central role in immortality and cell fate decisions of pluripotent stem cells. Here, we will review new insights into the melding fields of proteostasis and pluripotency and their implications for the understanding of organismal development and survival.
Collapse
Affiliation(s)
- Alireza Noormohammadi
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Giuseppe Calculli
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Ricardo Gutierrez-Garcia
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Amirabbas Khodakarami
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - Seda Koyuncu
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany
| | - David Vilchez
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, 50931, Cologne, Germany.
| |
Collapse
|
29
|
Wu YC, Lee KS, Song Y, Gehrke S, Lu B. The bantam microRNA acts through Numb to exert cell growth control and feedback regulation of Notch in tumor-forming stem cells in the Drosophila brain. PLoS Genet 2017; 13:e1006785. [PMID: 28520736 PMCID: PMC5453605 DOI: 10.1371/journal.pgen.1006785] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 06/01/2017] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Notch (N) signaling is central to the self-renewal of neural stem cells (NSCs) and other tissue stem cells. Its deregulation compromises tissue homeostasis and contributes to tumorigenesis and other diseases. How N regulates stem cell behavior in health and disease is not well understood. Here we show that N regulates bantam (ban) microRNA to impact cell growth, a process key to NSC maintenance and particularly relied upon by tumor-forming cancer stem cells. Notch signaling directly regulates ban expression at the transcriptional level, and ban in turn feedback regulates N activity through negative regulation of the Notch inhibitor Numb. This feedback regulatory mechanism helps maintain the robustness of N signaling activity and NSC fate. Moreover, we show that a Numb-Myc axis mediates the effects of ban on nucleolar and cellular growth independently or downstream of N. Our results highlight intricate transcriptional as well as translational control mechanisms and feedback regulation in the N signaling network, with important implications for NSC biology and cancer biology.
Collapse
Affiliation(s)
- Yen-Chi Wu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Kyu-Sun Lee
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States of America
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Yan Song
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States of America
- School of Life Sciences and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Stephan Gehrke
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States of America
| |
Collapse
|
30
|
Abstract
The Drosophila ovary represents a key in vivo model used to study germline stem cell (GSC) maintenance and stem cell daughter differentiation because these cells and their somatic cell neighbors can be identified at single-cell resolution within their native environment. Here we describe a fluorescent-based technique for the acquisition of 4D datasets of the Drosophila ovariole for periods that can exceed 12 consecutive hours. Live-cell imaging facilitates the investigation of molecular and cellular dynamics that were not previously possible using still images.
Collapse
Affiliation(s)
- Nevine A Shalaby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Institute for Biology, Freie Universität Berlin, 14195, Berlin, Germany.
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Pillitteri LJ, Guo X, Dong J. Asymmetric cell division in plants: mechanisms of symmetry breaking and cell fate determination. Cell Mol Life Sci 2016; 73:4213-4229. [PMID: 27286799 PMCID: PMC5522748 DOI: 10.1007/s00018-016-2290-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/02/2016] [Accepted: 06/02/2016] [Indexed: 02/07/2023]
Abstract
Asymmetric cell division is a fundamental mechanism that generates cell diversity while maintaining self-renewing stem cell populations in multicellular organisms. Both intrinsic and extrinsic mechanisms underpin symmetry breaking and differential daughter cell fate determination in animals and plants. The emerging picture suggests that plants deal with the problem of symmetry breaking using unique cell polarity proteins, mobile transcription factors, and cell wall components to influence asymmetric divisions and cell fate. There is a clear role for altered auxin distribution and signaling in distinguishing two daughter cells and an emerging role for epigenetic modifications through chromatin remodelers and DNA methylation in plant cell differentiation. The importance of asymmetric cell division in determining final plant form provides the impetus for its study in the areas of both basic and applied science.
Collapse
Affiliation(s)
- Lynn Jo Pillitteri
- Department of Biology, Western Washington University, Bellingham, WA, 98225, USA
| | - Xiaoyu Guo
- Waksman Institute of Microbiology, Rutgers the State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Juan Dong
- Waksman Institute of Microbiology, Rutgers the State University of New Jersey, Piscataway, NJ, 08854, USA.
- Department of Plant Biology and Pathology, Rutgers the State University of New Jersey, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
32
|
Wissel S, Kieser A, Yasugi T, Duchek P, Roitinger E, Gokcezade J, Steinmann V, Gaul U, Mechtler K, Förstemann K, Knoblich JA, Neumüller RA. A Combination of CRISPR/Cas9 and Standardized RNAi as a Versatile Platform for the Characterization of Gene Function. G3 (BETHESDA, MD.) 2016; 6:2467-78. [PMID: 27280787 PMCID: PMC4978900 DOI: 10.1534/g3.116.028571] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/31/2016] [Indexed: 12/21/2022]
Abstract
Traditional loss-of-function studies in Drosophila suffer from a number of shortcomings, including off-target effects in the case of RNA interference (RNAi) or the stochastic nature of mosaic clonal analysis. Here, we describe minimal in vivo GFP interference (miGFPi) as a versatile strategy to characterize gene function and to conduct highly stringent, cell type-specific loss-of-function experiments in Drosophila miGFPi combines CRISPR/Cas9-mediated tagging of genes at their endogenous locus with an immunotag and an exogenous 21 nucleotide RNAi effector sequence with the use of a single reagent, highly validated RNAi line targeting this sequence. We demonstrate the utility and time effectiveness of this method by characterizing the function of the Polymerase I (Pol I)-associated transcription factor Tif-1a, and the previously uncharacterized gene MESR4, in the Drosophila female germline stem cell lineage. In addition, we show that miGFPi serves as a powerful technique to functionally characterize individual isoforms of a gene. We exemplify this aspect of miGFPi by studying isoform-specific loss-of-function phenotypes of the longitudinals lacking (lola) gene in neural stem cells. Altogether, the miGFPi strategy constitutes a generalized loss-of-function approach that is amenable to the study of the function of all genes in the genome in a stringent and highly time effective manner.
Collapse
Affiliation(s)
- Sebastian Wissel
- Institute of Molecular Biotechnology Austria, 1030 Vienna, Austria
| | - Anja Kieser
- Gene Center, Ludwig-Maximilians-University Munich, 81377, Germany
| | - Tetsuo Yasugi
- Institute of Molecular Biotechnology Austria, 1030 Vienna, Austria
| | - Peter Duchek
- Institute of Molecular Biotechnology Austria, 1030 Vienna, Austria
| | - Elisabeth Roitinger
- Institute of Molecular Biotechnology Austria, 1030 Vienna, Austria Institute of Molecular Pathology, 1030 Vienna, Austria
| | - Joseph Gokcezade
- Institute of Molecular Biotechnology Austria, 1030 Vienna, Austria
| | | | - Ulrike Gaul
- Gene Center, Ludwig-Maximilians-University Munich, 81377, Germany
| | - Karl Mechtler
- Institute of Molecular Biotechnology Austria, 1030 Vienna, Austria Institute of Molecular Pathology, 1030 Vienna, Austria
| | - Klaus Förstemann
- Gene Center, Ludwig-Maximilians-University Munich, 81377, Germany
| | | | - Ralph A Neumüller
- Institute of Molecular Biotechnology Austria, 1030 Vienna, Austria Gene Center, Ludwig-Maximilians-University Munich, 81377, Germany
| |
Collapse
|
33
|
Minakhina S, Naryshkina T, Changela N, Tan W, Steward R. Zfrp8/PDCD2 Interacts with RpS2 Connecting Ribosome Maturation and Gene-Specific Translation. PLoS One 2016; 11:e0147631. [PMID: 26807849 PMCID: PMC4726551 DOI: 10.1371/journal.pone.0147631] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 11/25/2015] [Indexed: 11/28/2022] Open
Abstract
Zfrp8/PDCD2 is a highly conserved protein essential for stem cell maintenance in both flies and mammals. It is also required in fast proliferating cells such as cancer cells. Our previous studies suggested that Zfrp8 functions in the formation of mRNP (mRNA ribonucleoprotein) complexes and also controls RNA of select Transposable Elements (TEs). Here we show that in Zfrp8/PDCD2 knock down (KD) ovaries, specific mRNAs and TE transcripts show increased nuclear accumulation. We also show that Zfrp8/PDCD2 interacts with the (40S) small ribosomal subunit through direct interaction with RpS2 (uS5). By studying the distribution of endogenous and transgenic fluorescently tagged ribosomal proteins we demonstrate that Zfrp8/PDCD2 regulates the cytoplasmic levels of components of the small (40S) ribosomal subunit, but does not control nuclear/nucleolar localization of ribosomal proteins. Our results suggest that Zfrp8/PDCD2 functions at late stages of ribosome assembly and may regulate the binding of specific mRNA-RNPs to the small ribosomal subunit ultimately controlling their cytoplasmic localization and translation.
Collapse
Affiliation(s)
- Svetlana Minakhina
- Waksman Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- * E-mail: (SM); (RS)
| | - Tatyana Naryshkina
- Waksman Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Neha Changela
- Waksman Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - William Tan
- Waksman Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Ruth Steward
- Waksman Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- * E-mail: (SM); (RS)
| |
Collapse
|
34
|
Abstract
RNA-binding proteins (RBPs) play integral roles in gene regulation, yet only a small fraction of RBPs has been studied in the context of stem cells. Here we applied an RNAi screen for RBPs in mouse embryonic stem cells (ESCs) and identified 16 RBPs involved in pluripotency maintenance. Interestingly, six identified RBPs, including Krr1 and Ddx47, are part of a complex called small subunit processome (SSUP) that mediates 18S rRNA biogenesis. The SSUP components are preferentially expressed in stem cells and enhance the global translational rate, which is critical to sustain the protein levels of labile pluripotency factors such as Nanog and Esrrb. Furthermore, the SSUP proteins are required for efficient reprogramming of induced pluripotent stem cells. Our study uncovers the role of the SSUP and the importance of translational control in stem cell fate decision.
Collapse
Affiliation(s)
- Kwon Tae You
- Center for RNA Research, Institute for Basic Science, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Joha Park
- Center for RNA Research, Institute for Basic Science, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - V Narry Kim
- Center for RNA Research, Institute for Basic Science, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| |
Collapse
|
35
|
In Vitro Culturing and Live Imaging of Drosophila Egg Chambers: A History and Adaptable Method. Methods Mol Biol 2016; 1457:35-68. [PMID: 27557572 DOI: 10.1007/978-1-4939-3795-0_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of the Drosophila egg chamber encompasses a myriad of diverse germline and somatic events, and as such, the egg chamber has become a widely used and influential developmental model. Advantages of this system include physical accessibility, genetic tractability, and amenability to microscopy and live culturing, the last of which is the focus of this chapter. To provide adequate context, we summarize the structure of the Drosophila ovary and egg chamber, the morphogenetic events of oogenesis, the history of egg-chamber live culturing, and many of the important discoveries that this culturing has afforded. Subsequently, we discuss various culturing methods that have facilitated analyses of different stages of egg-chamber development and different types of cells within the egg chamber, and we present an optimized protocol for live culturing Drosophila egg chambers.We designed this protocol for culturing late-stage Drosophila egg chambers and live imaging epithelial tube morphogenesis, but with appropriate modifications, it can be used to culture egg chambers of any stage. The protocol employs a liquid-permeable, weighted "blanket" to gently hold egg chambers against the coverslip in a glass-bottomed culture dish so the egg chambers can be imaged on an inverted microscope. This setup provides a more buffered, stable, culturing environment than previously published methods by using a larger volume of culture media, but the setup is also compatible with small volumes. This chapter should aid researchers in their efforts to culture and live-image Drosophila egg chambers, further augmenting the impressive power of this model system.
Collapse
|
36
|
Abstract
Drosophila oogenesis is a powerful model for studying a wide spectrum of cellular and developmental processes in vivo. Oogenesis starts in a specialized structure called the germarium, which harbors the stem cells for both germ and somatic cells. The germarium produces egg chambers, each of which will develop into an egg. Active areas of research in Drosophila germaria include stem cell self-renewal, division, and maintenance, cell cycle control and differentiation, oocyte specification, intercellular communication, and signaling, among others. The solid knowledge base, the genetic tractability of the Drosophila model, as well as the availability and fast development of tools and imaging techniques for oogenesis research ensure that studies in this model will keep being instrumental for novel discoveries within cell and developmental biology also in the future. This chapter focuses on antibody staining in Drosophila germaria and provides a protocol for immunostaining as well as an overview of commonly used antibodies for visualization of different cell types and cellular structures. The protocol is well-suited for subsequent confocal microscopy analyses, and in addition we present key adaptations of the protocol that are useful when performing structured illumination microscopy (SIM) super-resolution imaging.
Collapse
Affiliation(s)
- Anette Lie-Jensen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379, Oslo, Norway
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, N-0379, Oslo, Norway
| | - Kaisa Haglund
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379, Oslo, Norway.
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, N-0379, Oslo, Norway.
| |
Collapse
|
37
|
Molla-Herman A, Vallés AM, Ganem-Elbaz C, Antoniewski C, Huynh JR. tRNA processing defects induce replication stress and Chk2-dependent disruption of piRNA transcription. EMBO J 2015; 34:3009-27. [PMID: 26471728 PMCID: PMC4687792 DOI: 10.15252/embj.201591006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 09/01/2015] [Accepted: 09/04/2015] [Indexed: 02/01/2023] Open
Abstract
RNase P is a conserved endonuclease that processes the 5' trailer of tRNA precursors. We have isolated mutations in Rpp30, a subunit of RNase P, and find that these induce complete sterility in Drosophila females. Here, we show that sterility is not due to a shortage of mature tRNAs, but that atrophied ovaries result from the activation of several DNA damage checkpoint proteins, including p53, Claspin, and Chk2. Indeed, we find that tRNA processing defects lead to increased replication stress and de-repression of transposable elements in mutant ovaries. We also report that transcription of major piRNA sources collapse in mutant germ cells and that this correlates with a decrease in heterochromatic H3K9me3 marks on the corresponding piRNA-producing loci. Our data thus link tRNA processing, DNA replication, and genome defense by small RNAs. This unexpected connection reveals constraints that could shape genome organization during evolution.
Collapse
Affiliation(s)
- Anahi Molla-Herman
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France CNRS UMR3215, Inserm U934, Paris, France
| | - Ana Maria Vallés
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France CNRS UMR3215, Inserm U934, Paris, France
| | - Carine Ganem-Elbaz
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France CNRS UMR3215, Inserm U934, Paris, France
| | - Christophe Antoniewski
- GED, UPMC, CNRS UMR 7622, IBPS, Developmental Biology Laboratory (IBPS-LBD), Paris, France
| | - Jean-René Huynh
- Department of Genetics and Developmental Biology, Institut Curie, Paris, France CNRS UMR3215, Inserm U934, Paris, France
| |
Collapse
|
38
|
Takada H, Kurisaki A. Emerging roles of nucleolar and ribosomal proteins in cancer, development, and aging. Cell Mol Life Sci 2015; 72:4015-25. [PMID: 26206377 PMCID: PMC11113460 DOI: 10.1007/s00018-015-1984-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/17/2015] [Accepted: 07/02/2015] [Indexed: 10/23/2022]
Abstract
Changes in nucleolar morphology and function are tightly associated with cellular activity, such as growth, proliferation, and cell cycle progression. Historically, these relationships have been extensively examined in cancer cells, which frequently exhibit large nucleoli and increased ribosome biogenesis. Recent findings indicate that alteration of nucleolar activity is a key regulator of development and aging. In this review, we have provided evidences that the nucleolus is not just a housekeeping factor but is actively involved in the regulation of cell proliferation, differentiation, and senescence both in vitro and in vivo. In addition, we have discussed how alteration of nucleolar function and nucleolar proteins induces specific physiological effects rather than widespread effects.
Collapse
Affiliation(s)
- Hitomi Takada
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-1, Higashi, Tsukuba, Ibaraki, 305-8562, Japan
| | - Akira Kurisaki
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Central 4, 1-1-1, Higashi, Tsukuba, Ibaraki, 305-8562, Japan.
| |
Collapse
|
39
|
Christophorou N, Rubin T, Bonnet I, Piolot T, Arnaud M, Huynh JR. Microtubule-driven nuclear rotations promote meiotic chromosome dynamics. Nat Cell Biol 2015; 17:1388-400. [PMID: 26458247 DOI: 10.1038/ncb3249] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 09/03/2015] [Indexed: 11/09/2022]
Abstract
At the onset of meiosis, each chromosome needs to find its homologue and pair to ensure proper segregation. In Drosophila, pairing occurs during the mitotic cycles preceding meiosis. Here we show that germ cell nuclei undergo marked movements during this developmental window. We demonstrate that microtubules and Dynein are driving nuclear rotations and are required for centromere pairing and clustering. We further found that Klaroid (SUN) and Klarsicht (KASH) co-localize with centromeres at the nuclear envelope and are required for proper chromosome motions and pairing. We identified Mud (NuMA in vertebrates) as co-localizing with centromeres, Klarsicht and Klaroid. Mud is also required to maintain the integrity of the nuclear envelope and for the correct assembly of the synaptonemal complex. Our findings reveal a mechanism for chromosome pairing in Drosophila, and indicate that microtubules, centrosomes and associated proteins play a crucial role in the dynamic organization of chromosomes inside the nucleus.
Collapse
Affiliation(s)
- Nicolas Christophorou
- Department of Genetics and Developmental Biology, Institut Curie, F-75248 Paris, France.,CNRS UMR3215, Inserm, U934 F-75248 Paris, France
| | - Thomas Rubin
- Department of Genetics and Developmental Biology, Institut Curie, F-75248 Paris, France.,CNRS UMR3215, Inserm, U934 F-75248 Paris, France
| | - Isabelle Bonnet
- Laboratoire Physico-Chimie, Institut Curie, F-75248 Paris, France.,CNRS UMR 168, UPMC, F-75248 Paris, France
| | - Tristan Piolot
- Department of Genetics and Developmental Biology, Institut Curie, F-75248 Paris, France.,CNRS UMR3215, Inserm, U934 F-75248 Paris, France
| | - Marion Arnaud
- Department of Genetics and Developmental Biology, Institut Curie, F-75248 Paris, France.,CNRS UMR3215, Inserm, U934 F-75248 Paris, France
| | - Jean-René Huynh
- Department of Genetics and Developmental Biology, Institut Curie, F-75248 Paris, France.,CNRS UMR3215, Inserm, U934 F-75248 Paris, France
| |
Collapse
|
40
|
Abstract
Stem cells are necessary for the maintenance of many adult tissues. Signals within the stem cell microenvironment, or niche, regulate the self-renewal and differentiation capability of these cells. Misregulation of these signals through mutation or damage can lead to overgrowth or depletion of different stem cell pools. In this review, we focus on the Drosophila testis and ovary, both of which contain well-defined niches, as well as the mouse testis, which has become a more approachable stem cell system with recent technical advances. We discuss the signals that regulate gonadal stem cells in their niches, how these signals mediate self-renewal and differentiation under homeostatic conditions, and how stress, whether from mutations or damage, can cause changes in cell fate and drive stem cell competition.
Collapse
Affiliation(s)
- Leah Joy Greenspan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; , ,
| | - Margaret de Cuevas
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; , ,
| | - Erika Matunis
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; , ,
| |
Collapse
|
41
|
Brombin A, Joly JS, Jamen F. New tricks for an old dog: ribosome biogenesis contributes to stem cell homeostasis. Curr Opin Genet Dev 2015; 34:61-70. [PMID: 26343009 DOI: 10.1016/j.gde.2015.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/24/2015] [Accepted: 07/29/2015] [Indexed: 01/21/2023]
Abstract
Although considered a 'house-keeping' function, ribosome biogenesis is regulated differently between cells and can be modulated in a cell-type-specific manner. These differences are required to generate specialized ribosomes that contribute to the translational control of gene expression by selecting mRNA subsets to be translated. Thus, differences in ribosome biogenesis between stem and differentiated cells indirectly contribute to determine cell identity. The concept of the existence of stem cell-specific mechanisms of ribosome biogenesis has progressed from an attractive theory to a useful working model with important implications for basic and medical research.
Collapse
Affiliation(s)
- Alessandro Brombin
- CASBAH Group, University Paris-Saclay, University Paris-Sud, UMR CNRS 9197, Neuroscience Paris-Saclay Institute (NeuroPSI), Bât. 32/33, 1 Avenue de la Terrasse, F-91190 Gif-sur-Yvette, France; INRA, USC 1126, F-91190 Gif-sur-Yvette, France
| | - Jean-Stéphane Joly
- CASBAH Group, University Paris-Saclay, University Paris-Sud, UMR CNRS 9197, Neuroscience Paris-Saclay Institute (NeuroPSI), Bât. 32/33, 1 Avenue de la Terrasse, F-91190 Gif-sur-Yvette, France; INRA, USC 1126, F-91190 Gif-sur-Yvette, France
| | - Françoise Jamen
- CASBAH Group, University Paris-Saclay, University Paris-Sud, UMR CNRS 9197, Neuroscience Paris-Saclay Institute (NeuroPSI), Bât. 32/33, 1 Avenue de la Terrasse, F-91190 Gif-sur-Yvette, France; INRA, USC 1126, F-91190 Gif-sur-Yvette, France.
| |
Collapse
|
42
|
Ribosome biogenesis dysfunction leads to p53-mediated apoptosis and goblet cell differentiation of mouse intestinal stem/progenitor cells. Cell Death Differ 2015; 22:1865-76. [PMID: 26068591 DOI: 10.1038/cdd.2015.57] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 03/31/2015] [Accepted: 04/13/2015] [Indexed: 02/07/2023] Open
Abstract
Ribosome biogenesis is an essential cellular process. Its impairment is associated with developmental defects and increased risk of cancer. The in vivo cellular responses to defective ribosome biogenesis and the underlying molecular mechanisms are still incompletely understood. In particular, the consequences of impaired ribosome biogenesis within the intestinal epithelium in mammals have not been investigated so far. Here we adopted a genetic approach to investigate the role of Notchless (NLE), an essential actor of ribosome biogenesis, in the adult mouse intestinal lineage. Nle deficiency led to defects in the synthesis of large ribosomal subunit in crypts cells and resulted in the rapid elimination of intestinal stem cells and progenitors through distinct types of cellular responses, including apoptosis, cell cycle arrest and biased differentiation toward the goblet cell lineage. Similar observations were made using the rRNA transcription inhibitor CX-5461 on intestinal organoids culture. Importantly, we found that p53 activation was responsible for most of the cellular responses observed, including differentiation toward the goblet cell lineage. Moreover, we identify the goblet cell-specific marker Muc2 as a direct transcriptional target of p53. Nle-deficient ISCs and progenitors disappearance persisted in the absence of p53, underlying the existence of p53-independent cellular responses following defective ribosome biogenesis. Our data indicate that NLE is a crucial factor for intestinal homeostasis and provide new insights into how perturbations of ribosome biogenesis impact on cell fate decisions within the intestinal epithelium.
Collapse
|
43
|
Katajisto P, Döhla J, Chaffer CL, Pentinmikko N, Marjanovic N, Iqbal S, Zoncu R, Chen W, Weinberg RA, Sabatini DM. Stem cells. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science 2015; 348:340-3. [PMID: 25837514 DOI: 10.1126/science.1260384] [Citation(s) in RCA: 392] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/12/2015] [Indexed: 12/19/2022]
Abstract
By dividing asymmetrically, stem cells can generate two daughter cells with distinct fates. However, evidence is limited in mammalian systems for the selective apportioning of subcellular contents between daughters. We followed the fates of old and young organelles during the division of human mammary stemlike cells and found that such cells apportion aged mitochondria asymmetrically between daughter cells. Daughter cells that received fewer old mitochondria maintained stem cell traits. Inhibition of mitochondrial fission disrupted both the age-dependent subcellular localization and segregation of mitochondria and caused loss of stem cell properties in the progeny cells. Hence, mechanisms exist for mammalian stemlike cells to asymmetrically sort aged and young mitochondria, and these are important for maintaining stemness properties.
Collapse
Affiliation(s)
- Pekka Katajisto
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA. Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland.
| | - Julia Döhla
- Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland
| | | | - Nalle Pentinmikko
- Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland
| | - Nemanja Marjanovic
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - Sharif Iqbal
- Institute of Biotechnology, University of Helsinki, P.O. Box 00014, Helsinki, Finland
| | - Roberto Zoncu
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA
| | - Walter Chen
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Boston, MA 02142, USA. Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA. Howard Hughes Medical Institute, MIT, Cambridge, MA 02139, USA. Broad Institute, Cambridge, MA 02142, USA. The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
44
|
Buszczak M, Signer RAJ, Morrison SJ. Cellular differences in protein synthesis regulate tissue homeostasis. Cell 2015; 159:242-51. [PMID: 25303523 DOI: 10.1016/j.cell.2014.09.016] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Indexed: 12/12/2022]
Abstract
Although sometimes considered a "house-keeping" function, multiple aspects of protein synthesis are regulated differently among somatic cells, including stem cells, and can be modulated in a cell-type-specific manner. These differences are required to establish and maintain differences in cell identity, cell function, tissue homeostasis, and tumor suppression.
Collapse
Affiliation(s)
- Michael Buszczak
- Department of Molecular Biology, Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Robert A J Signer
- Howard Hughes Medical Institute, Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J Morrison
- Howard Hughes Medical Institute, Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
45
|
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
|
46
|
Manning L, Starz-Gaiano M. Culturing Drosophila Egg Chambers and Investigating Developmental Processes Through Live Imaging. Methods Mol Biol 2015; 1328:73-88. [PMID: 26324430 DOI: 10.1007/978-1-4939-2851-4_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Drosophila oogenesis provides many examples of essential processes in development. A myriad of genetic tools combined with recent advances in culturing egg chambers ex vivo has revealed several surprising mechanisms that govern how this tissue develops, and which could not have been determined in fixed tissues. Here we describe a straightforward protocol for dissecting ovaries, culturing egg chambers, and observing egg development in real time by fluorescent microscopy. This technique is suitable for observation of early- or late-stage egg development, and can be adapted to study a variety of cellular, molecular, or developmental processes. Ongoing analysis of oogenesis in living egg chambers has tremendous potential for discovery of new developmental mechanisms.
Collapse
Affiliation(s)
- Lathiena Manning
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA
| | | |
Collapse
|
47
|
A small subunit processome protein promotes cancer by altering translation. Oncogene 2014; 34:4471-81. [DOI: 10.1038/onc.2014.376] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 08/31/2014] [Accepted: 10/03/2014] [Indexed: 12/27/2022]
|
48
|
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: 1.3] [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
|
49
|
|
50
|
Zhang Q, Shalaby NA, Buszczak M. Changes in rRNA transcription influence proliferation and cell fate within a stem cell lineage. Science 2014; 343:298-301. [PMID: 24436420 DOI: 10.1126/science.1246384] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Ribosome biogenesis drives cell growth and proliferation, but mechanisms that modulate this process within specific lineages remain poorly understood. Here, we identify a Drosophila RNA polymerase I (Pol I) regulatory complex composed of Under-developed (Udd), TAF1B, and a TAF1C-like factor. Disruption of udd or TAF1B results in reduced ovarian germline stem cell (GSC) proliferation. Female GSCs display high levels of ribosomal RNA (rRNA) transcription, and Udd becomes enriched in GSCs relative to their differentiating daughters. Increasing Pol I transcription delays differentiation, whereas reducing rRNA production induces both morphological changes that accompany multicellular cyst formation and specific decreased expression of the bone morphogenetic protein (BMP) pathway component Mad. These findings demonstrate that modulating rRNA synthesis fosters changes in the cell fate, growth, and proliferation of female Drosophila GSCs and their daughters.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9148, USA
| | | | | |
Collapse
|