1
|
Segura RC, Gallaud E, Sythoff AVB, Aavula K, Taylor JA, Vahdat D, Otte F, Pielage J, Cabernard C. Asymmetry of centrosomes in Drosophila neural stem cells requires protein phosphatase 4. Mol Biol Cell 2025; 36:ar58. [PMID: 40072519 PMCID: PMC12086568 DOI: 10.1091/mbc.e25-01-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Asymmetric cell division is used by stem cells to create diverse cell types while self-renewing the stem cell population. Biased segregation of molecularly distinct centrosomes could provide a mechanism to maintain stem cell fate, induce cell differentiation or both. However, the molecular mechanisms generating molecular and functional asymmetric centrosomes remain incompletely understood. Here, we show that in asymmetrically dividing fly neural stem cells, protein phosphatase 4 (Pp4) is necessary for correct centrosome asymmetry establishment during mitosis, and microtubule organizing center (MTOC) maintenance in interphase. Using in vivo live-cell imaging, we show that while wild-type neural stem cells always maintain one active MTOC, Pp4 mutant neuroblasts contain two inactive centrioles in interphase. Furthermore, centrosomes of Pp4 mutant neural stem cells mature in mitosis but fail to correctly transfer the centriolar protein Centrobin (Cnb) from the mother to the daughter centriole. Using superresolution imaging, we find that phosphomimetic Centrobin fails to accurately relocalize in mitosis. We propose that Pp4 regulates the timely relocalization of Cnb in mitosis to establish two molecularly distinct centrosomes. In addition, Pp4 is also necessary to maintain MTOC activity in interphase, ensuring biased centrosome segregation. Mechanistically, Pp4 could regulate centrosome asymmetry by dephosphorylating both Cnb and gamma-Tubulin.
Collapse
Affiliation(s)
- Roberto Carlos Segura
- Department of Biology, University of Washington, Life Sciences Building, Seattle, WA 98105
| | - Emmanuel Gallaud
- Department of Biology, University of Washington, Life Sciences Building, Seattle, WA 98105
| | | | - Kumar Aavula
- Department of Neurobiology, RPTU University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Jennifer A. Taylor
- Department of Biology, University of Washington, Life Sciences Building, Seattle, WA 98105
| | - Danielle Vahdat
- Department of Biology, University of Washington, Life Sciences Building, Seattle, WA 98105
| | - Fabian Otte
- Department of Biology, University of Washington, Life Sciences Building, Seattle, WA 98105
| | - Jan Pielage
- Department of Neurobiology, RPTU University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Clemens Cabernard
- Department of Biology, University of Washington, Life Sciences Building, Seattle, WA 98105
| |
Collapse
|
2
|
Segura RC, Gallaud E, von Barnau Sythoff A, Aavula K, Taylor JA, Vahdat D, Pielage J, Cabernard C. Protein phosphatase 4 is required for centrosome asymmetry in fly neural stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633270. [PMID: 39868139 PMCID: PMC11761633 DOI: 10.1101/2025.01.15.633270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Asymmetric cell division is used by stem cells to create diverse cell types while self-renewing the stem cell population. Biased segregation of molecularly distinct centrosomes could provide a mechanism to maintain stem cell fate, induce cell differentiation or both. However, the molecular mechanisms generating molecular and functional asymmetric centrosomes remain incompletely understood. Here, we show that in asymmetrically dividing fly neural stem cells, Protein phosphatase 4 (Pp4) is necessary for correct centrosome asymmetry establishment during mitosis, and microtubule organizing center (MTOC) maintenance in interphase. Using in-vivo live cell imaging we show that while wild type neural stem cells always maintain one active MTOC, Pp4 mutant neuroblasts contain two inactive centrioles in interphase. Furthermore, centrosomes of Pp4 mutant neural stem cells mature in mitosis but fail to correctly transfer the centriolar protein Centrobin (Cnb) from the mother to the daughter centriole. Using superresolution imaging, we find that phosphomimetic Centrobin fails to accurately relocalize in mitosis. We propose that Pp4 regulates the timely relocalization of Cnb in mitosis to establish two molecularly distinct centrosomes. In addition, Pp4 is also necessary to maintain MTOC activity in interphase, ensuring biased centrosome segregation. Mechanistically, Pp4 could regulate centrosome asymmetry by dephosphorylating both Cnb and gamma-Tubulin. SIGNIFICANCE STATEMENT Asymmetric centrosome segregation occurs in stem cells and has been linked with cell fate decisions. Protein phosphatase 4 (Pp4), a conserved Serine/Threonine phosphatase, regulates centrosome asymmetry in Drosophila neural stem cells by acting upon gamma tubulin and Centrobin. Pp4 regulates centrosome asymmetry establishment in mitosis and interphase, necessary for biased centrosome segregation.
Collapse
|
3
|
Connell M, Xie Y, Deng X, Chen R, Zhu S. Kin17 regulates proper cortical localization of Miranda in Drosophila neuroblasts by regulating Flfl expression. Cell Rep 2024; 43:113823. [PMID: 38386552 PMCID: PMC10980573 DOI: 10.1016/j.celrep.2024.113823] [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: 12/07/2021] [Revised: 10/16/2022] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
During asymmetric division of Drosophila larval neuroblasts, the fate determinant Prospero (Pros) and its adaptor Miranda (Mira) are segregated to the basal cortex through atypical protein kinase C (aPKC) phosphorylation of Mira and displacement from the apical cortex, but Mira localization after aPKC phosphorylation is not well understood. We identify Kin17, a DNA replication and repair protein, as a regulator of Mira localization during asymmetric cell division. Loss of Kin17 leads to aberrant localization of Mira and Pros to the centrosome, cytoplasm, and nucleus. We provide evidence to show that the mislocalization of Mira and Pros is likely due to reduced expression of Falafel (Flfl), a component of protein phosphatase 4 (PP4), and defects in dephosphorylation of serine-96 of Mira. Our work reveals that Mira is likely dephosphorylated by PP4 at the centrosome to ensure proper basal localization of Mira after aPKC phosphorylation and that Kin17 regulates PP4 activity by regulating Flfl expression.
Collapse
Affiliation(s)
- Marisa Connell
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Yonggang Xie
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Xiaobing Deng
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Rui Chen
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Sijun Zhu
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
4
|
Penkert RR, LaFoya B, Moholt-Siebert L, Vargas E, Welch SE, Prehoda KE. The Drosophila neuroblast polarity cycle at a glance. J Cell Sci 2024; 137:jcs261789. [PMID: 38465513 PMCID: PMC10984279 DOI: 10.1242/jcs.261789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Drosophila neural stem cells, or neuroblasts, rapidly proliferate during embryonic and larval development to populate the central nervous system. Neuroblasts divide asymmetrically to create cellular diversity, with each division producing one sibling cell that retains the neuroblast fate and another that differentiates into glia or neurons. This asymmetric outcome is mediated by the transient polarization of numerous factors to the cell cortex during mitosis. The powerful genetics and outstanding imaging tractability of the neuroblast make it an excellent model system for studying the mechanisms of cell polarity. This Cell Science at a Glance article and the accompanying poster explore the phases of the neuroblast polarity cycle and the regulatory circuits that control them. We discuss the key features of the cycle - the targeted recruitment of proteins to specific regions of the plasma membrane and multiple phases of highly dynamic actomyosin-dependent cortical flows that pattern both protein distribution and membrane structure.
Collapse
|
5
|
Réthi-Nagy Z, Ábrahám E, Sinka R, Juhász S, Lipinszki Z. Protein Phosphatase 4 Is Required for Centrobin Function in DNA Damage Repair. Cells 2023; 12:2219. [PMID: 37759442 PMCID: PMC10526779 DOI: 10.3390/cells12182219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Genome stability in human cells relies on the efficient repair of double-stranded DNA breaks, which is mainly achieved by homologous recombination (HR). Among the regulators of various cellular functions, Protein phosphatase 4 (PP4) plays a pivotal role in coordinating cellular response to DNA damage. Meanwhile, Centrobin (CNTRB), initially recognized for its association with centrosomal function and microtubule dynamics, has sparked interest due to its potential contribution to DNA repair processes. In this study, we investigate the involvement of PP4 and its interaction with CNTRB in HR-mediated DNA repair in human cells. Employing a range of experimental strategies, we investigate the physical interaction between PP4 and CNTRB and shed light on the importance of two specific motifs in CNTRB, the PP4-binding FRVP and the ATR kinase recognition SQ sequences, in the DNA repair process. Moreover, we examine cells depleted of PP4 or CNTRB and cells harboring FRVP and SQ mutations in CNTRB, which result in similar abnormal chromosome morphologies. This phenomenon likely results from the impaired resolution of Holliday junctions, which serve as crucial intermediates in HR. Taken together, our results provide new insights into the intricate mechanisms of PP4 and CNTRB-regulated HR repair and their interrelation.
Collapse
Affiliation(s)
- Zsuzsánna Réthi-Nagy
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, H-6726 Szeged, Hungary
| | - Edit Ábrahám
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- National Laboratory for Biotechnology, Institute of Genetics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| | - Rita Sinka
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary;
| | - Szilvia Juhász
- Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| | - Zoltán Lipinszki
- MTA SZBK Lendület Laboratory of Cell Cycle Regulation, Institute of Biochemistry, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary; (Z.R.-N.); (E.Á.)
- National Laboratory for Biotechnology, Institute of Genetics, HUN-REN Biological Research Centre, H-6726 Szeged, Hungary
| |
Collapse
|
6
|
Chen D, Gao S, Gao F, Liu A, Li J, Li J, Liu Q. SMEK1 promotes lung adenocarcinoma proliferation and invasion by activating Wnt/β-catenin signaling pathway. Clin Transl Oncol 2023; 25:976-986. [PMID: 36463369 DOI: 10.1007/s12094-022-03001-8] [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: 07/07/2022] [Accepted: 10/31/2022] [Indexed: 12/07/2022]
Abstract
PURPOSE SMEK1, also known as PP4R3α, the regulatory subunit 3α of serine and threonine phosphatase PP4, participates in diversely critical biological processes such as the integration of centromere, deacetylation of histones, asymmetric divisions of neuroblast, and other crucial cellular activities. SMEK1 was formerly reported to play a part in carcinogenesis. This study aims to reveal the role of SMEK1 in lung adenocarcinoma and the underlying molecular mechanism. METHODS Using immunohistochemical (IHC) staining, the protein level of SMEK1 in lung adenocarcinoma and adjacent non-tumor tissue was detected. The functional role of SMEK1 in cell proliferation and invasion was explored using cell counting kit-8 and Transwell assay, respectively. Xenograft tumor experiment was used to investigate the effect of SMEK1 on tumor growth in vivo. The alteration of Wnt/β-catenin signaling pathway was detected by Western blotting, quantitative PCR, and dual-luciferase reporter assays. RESULTS SMEK1 was highly expressed at the protein level in lung adenocarcinoma compared to the adjacent non-tumor tissue. In vitro, suppression of SMEK1 significantly decreased the proliferation, migration, and invasion of lung adenocarcinoma cell lines, while overexpression of SMEK1 enhanced above abilities. The xenograft model demonstrated that down-regulation of SMEK1 significantly inhibited tumor growth in vivo. In addition, we found that SMEK1 could positively regulate Wnt/β-catenin signaling in lung adenocarcinoma cell lines. CONCLUSIONS SMEK1 exerts a cancer-promoting effect in lung adenocarcinoma by activating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Dandan Chen
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Shang Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Fei Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Ai Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Jiangxia Li
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 West Wenhua Road, Jinan, 250012, Shandong, China
| | - Jisheng Li
- Department of Medical Oncology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| | - Qiji Liu
- Key Laboratory for Experimental Teratology of the Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, No. 44 West Wenhua Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
7
|
Identification of replication fork-associated proteins in Drosophila embryos and cultured cells using iPOND coupled to quantitative mass spectrometry. Sci Rep 2022; 12:6903. [PMID: 35484306 PMCID: PMC9050644 DOI: 10.1038/s41598-022-10821-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Replication of the eukaryotic genome requires the formation of thousands of replication forks that must work in concert to accurately replicate the genetic and epigenetic information. Defining replication fork-associated proteins is a key step in understanding how genomes are replicated and repaired in the context of chromatin to maintain genome stability. To identify replication fork-associated proteins, we performed iPOND (Isolation of Proteins on Nascent DNA) coupled to quantitative mass spectrometry in Drosophila embryos and cultured cells. We identified 76 and 278 fork-associated proteins in post-MZT embryos and Drosophila cultured S2 cells, respectively. By performing a targeted screen of a subset of these proteins, we demonstrate that BRWD3, a targeting specificity factor for the DDB1/Cul4 ubiquitin ligase complex (CRL4), functions at or in close proximity to replication forks to promote fork progression and maintain genome stability. Altogether, our work provides a valuable resource for those interested in DNA replication, repair and chromatin assembly during development.
Collapse
|
8
|
Salem Wehbe L, Barakat D, Acker A, El Khoury R, Reichhart JM, Matt N, El Chamy L. Protein Phosphatase 4 Negatively Regulates the Immune Deficiency-NF-κB Pathway during the Drosophila Immune Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:1616-1626. [PMID: 34452932 PMCID: PMC7616922 DOI: 10.4049/jimmunol.1901497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 07/07/2021] [Indexed: 12/31/2022]
Abstract
The evolutionarily conserved immune deficiency (IMD) signaling pathway shields Drosophila against bacterial infections. It regulates the expression of antimicrobial peptides encoding genes through the activation of the NF-κB transcription factor Relish. Tight regulation of the signaling cascade ensures a balanced immune response, which is otherwise highly harmful. Several phosphorylation events mediate intracellular progression of the IMD pathway. However, signal termination by dephosphorylation remains largely elusive. Here, we identify the highly conserved protein phosphatase 4 (PP4) complex as a bona fide negative regulator of the IMD pathway. RNA interference-mediated gene silencing of PP4-19c, PP4R2, and Falafel, which encode the catalytic and regulatory subunits of the phosphatase complex, respectively, caused a marked upregulation of bacterial-induced antimicrobial peptide gene expression in both Drosophila melanogaster S2 cells and adult flies. Deregulated IMD signaling is associated with reduced lifespan of PP4-deficient flies in the absence of any infection. In contrast, flies overexpressing this phosphatase are highly sensitive to bacterial infections. Altogether, our results highlight an evolutionarily conserved function of PP4c in the regulation of NF-κB signaling from Drosophila to mammals.
Collapse
Affiliation(s)
- Layale Salem Wehbe
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | - Dana Barakat
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | - Adrian Acker
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
| | - Rita El Khoury
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| | | | - Nicolas Matt
- Université de Strasbourg, CNRS, M3I UPR 9022, Strasbourg, France; and
| | - Laure El Chamy
- Unité de Recherche Environnement, Génomique et Protéomique, Faculté des Sciences, Université Saint-Joseph de Beyrouth-Liban, Mar Roukos, Mkalles, Beirut, Lebanon
| |
Collapse
|
9
|
Park J, Lee DH. Functional roles of protein phosphatase 4 in multiple aspects of cellular physiology: a friend and a foe. BMB Rep 2021. [PMID: 32192570 PMCID: PMC7196183 DOI: 10.5483/bmbrep.2020.53.4.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protein phosphatase 4 (PP4), one of serine/threonine phosphatases, is involved in many critical cellular pathways, including DNA damage response (DNA repair, cell cycle regulation, and apoptosis), tumorigenesis, cell migration, immune response, stem cell development, glucose metabolism, and diabetes. PP4 has been steadily studied over the past decade about wide spectrum of physiological activities in cells. Given the many vital functions in cells, PP4 has great potential to develop into the finding of key working mechanisms and effective treatments for related diseases such as cancer and diabetes. In this review, we provide an overview of the cellular and molecular mechanisms by which PP4 impacts and also discuss the functional significance of it in cell health.
Collapse
Affiliation(s)
- Jaehong Park
- School of Biological Sciences and Biotechnology Graduate School, Chonnam National University, Gwangju 61186, Korea
| | - Dong-Hyun Lee
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju 61186; Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, Korea
| |
Collapse
|
10
|
Karman Z, Rethi-Nagy Z, Abraham E, Fabri-Ordogh L, Csonka A, Vilmos P, Debski J, Dadlez M, Glover DM, Lipinszki Z. Novel perspectives of target-binding by the evolutionarily conserved PP4 phosphatase. Open Biol 2020; 10:200343. [PMID: 33352067 PMCID: PMC7776573 DOI: 10.1098/rsob.200343] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Abstract
Protein phosphatase 4 (PP4) is an evolutionarily conserved and essential Ser/Thr phosphatase that regulates cell division, development and DNA repair in eukaryotes. The major form of PP4, present from yeast to human, is the PP4c-R2-R3 heterotrimeric complex. The R3 subunit is responsible for substrate-recognition via its EVH1 domain. In typical EVH1 domains, conserved phenylalanine, tyrosine and tryptophan residues form the specific recognition site for their target's proline-rich sequences. Here, we identify novel binding partners of the EVH1 domain of the Drosophila R3 subunit, Falafel, and demonstrate that instead of binding to proline-rich sequences this EVH1 variant specifically recognizes atypical ligands, namely the FxxP and MxPP short linear consensus motifs. This interaction is dependent on an exclusively conserved leucine that replaces the phenylalanine invariant of all canonical EVH1 domains. We propose that the EVH1 domain of PP4 represents a new class of the EVH1 family that can accommodate low proline content sequences, such as the FxxP motif. Finally, our data implicate the conserved Smk-1 domain of Falafel in target-binding. These findings greatly enhance our understanding of the substrate-recognition mechanisms and function of PP4.
Collapse
Affiliation(s)
- Zoltan Karman
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, H‐6725, Hungary
| | - Zsuzsanna Rethi-Nagy
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, H‐6725, Hungary
| | - Edit Abraham
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
| | - Lilla Fabri-Ordogh
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
| | - Akos Csonka
- Department of Traumatology, University of Szeged, Szeged, H‐6725, Hungary
| | - Peter Vilmos
- Biological Research Centre, Institute of Genetics, Szeged, H‐6726, Hungary
| | - Janusz Debski
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Michal Dadlez
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - David M. Glover
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- California Institute of Technology, Pasadena, CA 91125, USA
| | - Zoltan Lipinszki
- Biological Research Centre, Institute of Biochemistry, MTA Lendület Laboratory of Cell Cycle Regulation, Szeged, H‐6726, Hungary
| |
Collapse
|
11
|
Abstract
Asymmetric cell division (ACD) is an evolutionarily conserved mechanism used by prokaryotes and eukaryotes alike to control cell fate and generate cell diversity. A detailed mechanistic understanding of ACD is therefore necessary to understand cell fate decisions in health and disease. ACD can be manifested in the biased segregation of macromolecules, the differential partitioning of cell organelles, or differences in sibling cell size or shape. These events are usually preceded by and influenced by symmetry breaking events and cell polarization. In this Review, we focus predominantly on cell intrinsic mechanisms and their contribution to cell polarization, ACD and binary cell fate decisions. We discuss examples of polarized systems and detail how polarization is established and, whenever possible, how it contributes to ACD. Established and emerging model organisms will be considered alike, illuminating both well-documented and underexplored forms of polarization and ACD.
Collapse
Affiliation(s)
- Bharath Sunchu
- Department of Biology, University of Washington, Life Science Building, Seattle, WA 98195, USA
| | - Clemens Cabernard
- Department of Biology, University of Washington, Life Science Building, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Keegan SE, Hughes SC. Role of nuclear-cytoplasmic protein localization during Drosophila neuroblast development. Genome 2020; 64:75-85. [PMID: 32526151 DOI: 10.1139/gen-2020-0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nuclear-cytoplasmic localization is an efficient way to regulate transcription factors and chromatin remodelers. Altering the location of existing protein pools also facilitates a more rapid response to changes in cell activity or extracellular signals. There are several examples of proteins that are regulated by nucleo-cytoplasmic shuttling, which are required for Drosophila neuroblast development. Disruption of the localization of homologs of these proteins has also been linked to several neurodegenerative disorders in humans. Drosophila has been used extensively to model the neurodegenerative disorders caused by aberrant nucleo-cytoplasmic localization. Here, we focus on the role of alternative nucleo-cytoplasmic protein localization in regulating proliferation and cell fate decisions in the Drosophila neuroblast and in neurodegenerative disorders. We also explore the analogous role of RNA binding proteins and mRNA localization in the context of regulation of nucleo-cytoplasmic localization during neural development and a role in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sophie E Keegan
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Sarah C Hughes
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
13
|
DAF-16 and SMK-1 Contribute to Innate Immunity During Adulthood in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2020; 10:1521-1539. [PMID: 32161087 PMCID: PMC7202018 DOI: 10.1534/g3.120.401166] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Aging is accompanied by a progressive decline in immune function termed "immunosenescence". Deficient surveillance coupled with the impaired function of immune cells compromises host defense in older animals. The dynamic activity of regulatory modules that control immunity appears to underlie age-dependent modifications to the immune system. In the roundworm Caenorhabditis elegans levels of PMK-1 p38 MAP kinase diminish over time, reducing the expression of immune effectors that clear bacterial pathogens. Along with the PMK-1 pathway, innate immunity in C. elegans is regulated by the insulin signaling pathway. Here we asked whether DAF-16, a Forkhead box (FOXO) transcription factor whose activity is inhibited by insulin signaling, plays a role in host defense later in life. While in younger C. elegans DAF-16 is inactive unless stimulated by environmental insults, we found that even in the absence of acute stress the transcriptional activity of DAF-16 increases in an age-dependent manner. Beginning in the reproductive phase of adulthood, DAF-16 upregulates a subset of its transcriptional targets, including genes required to kill ingested microbes. Accordingly, DAF-16 has little to no role in larval immunity, but functions specifically during adulthood to confer resistance to bacterial pathogens. We found that DAF-16-mediated immunity in adults requires SMK-1, a regulatory subunit of the PP4 protein phosphatase complex. Our data suggest that as the function of one branch of the innate immune system of C. elegans (PMK-1) declines over time, DAF-16-mediated immunity ramps up to become the predominant means of protecting adults from infection, thus reconfiguring immunity later in life.
Collapse
|
14
|
Yang Q, Wu J, Zhao J, Xu T, Han P, Song X. The Expression Profiles of lncRNAs and Their Regulatory Network During Smek1/2 Knockout Mouse Neural Stem Cells Differentiation. Curr Bioinform 2020. [DOI: 10.2174/1574893614666190308160507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background:
Previous studies indicated that the cell fate of neural stem cells (NSCs)
after differentiation is determined by Smek1, one isoform of suppressor of Mek null (Smek). Smek
deficiency prevents NSCs from differentiation, thus affects the development of nervous system. In
recent years, lncRNAs have been found to participate in numerous developmental and biological
pathways. However, the effects of knocking out Smek on the expression profiles of lncRNAs
during the differentiation remain unknown.
Objective:
This study is to explore the expression profiles of lncRNAs and their possible function
during the differentiation from Smek1/2 knockout NSCs.
Methods:
We obtained NSCs from the C57BL/6J mouse fetal cerebral cortex. One group of NSCs
was from wildtype mouse (WT group), while another group was from knocked out Smek1/2 (KO
group).
Results:
By analyzing the RNA-Seq data, we found that after knocking out Smek1/2, the
expression profiles of mRNAs and lncRNAs revealed significant changes. Analyses indicated that
these affected mRNAs have connections with the pathway network for the differentiation and
proliferation of NSCs. Furthermore, we performed a co-expression network analysis on the
differentially expressed mRNAs and lncRNAs, which helped reveal the possible regulatory rules
of lncRNAs during the differentiation after knocking out Smek1/2.
Conclusion:
By comparing group WT with KO, we found 366 differentially expressed mRNAs
and 12 lncRNAs. GO and KEGG enrichment analysis on these mRNAs suggested their
relationships with differentiation and proliferation of NSCs. Some of these mRNAs and lncRNAs
have been verified to play regulatory roles in nervous system. Analyses on the co-expression
network also indicated the possible functions of affected mRNAs and lncRNAs during NSCs
differentiation after knocking out Smek1/2.
Collapse
Affiliation(s)
- Qichang Yang
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Jing Wu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Jian Zhao
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Tianyi Xu
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| | - Ping Han
- The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, 210019, China
| | - Xiaofeng Song
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, Jiangsu, 211106, China
| |
Collapse
|
15
|
Loyer N, Januschke J. Where does asymmetry come from? Illustrating principles of polarity and asymmetry establishment in Drosophila neuroblasts. Curr Opin Cell Biol 2020; 62:70-77. [DOI: 10.1016/j.ceb.2019.07.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022]
|
16
|
Crews ST. Drosophila Embryonic CNS Development: Neurogenesis, Gliogenesis, Cell Fate, and Differentiation. Genetics 2019; 213:1111-1144. [PMID: 31796551 PMCID: PMC6893389 DOI: 10.1534/genetics.119.300974] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/26/2019] [Indexed: 01/04/2023] Open
Abstract
The Drosophila embryonic central nervous system (CNS) is a complex organ consisting of ∼15,000 neurons and glia that is generated in ∼1 day of development. For the past 40 years, Drosophila developmental neuroscientists have described each step of CNS development in precise molecular genetic detail. This has led to an understanding of how an intricate nervous system emerges from a single cell. These studies have also provided important, new concepts in developmental biology, and provided an essential model for understanding similar processes in other organisms. In this article, the key genes that guide Drosophila CNS development and how they function is reviewed. Features of CNS development covered in this review are neurogenesis, gliogenesis, cell fate specification, and differentiation.
Collapse
Affiliation(s)
- Stephen T Crews
- Department of Biochemistry and Biophysics, Integrative Program for Biological and Genome Sciences, School of Medicine, The University of North Carolina at Chapel Hill, North Carolina 27599
| |
Collapse
|
17
|
Lukaszewicz AI, Nguyen C, Melendez E, Lin DP, Teo JL, Lai KKY, Huttner WB, Shi SH, Kahn M. The Mode of Stem Cell Division Is Dependent on the Differential Interaction of β-Catenin with the Kat3 Coactivators CBP or p300. Cancers (Basel) 2019; 11:cancers11070962. [PMID: 31324005 PMCID: PMC6678591 DOI: 10.3390/cancers11070962] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 01/03/2023] Open
Abstract
Normal long-term repopulating somatic stem cells (SSCs) preferentially divide asymmetrically, with one daughter cell remaining in the niche and the other going on to be a transient amplifying cell required for generating new tissue in homeostatic maintenance and repair processes, whereas cancer stem cells (CSCs) favor symmetric divisions. We have previously proposed that differential β-catenin modulation of transcriptional activity via selective interaction with either the Kat3 coactivator CBP or its closely related paralog p300, regulates symmetric versus asymmetric division in SSCs and CSCs. We have previously demonstrated that SSCs that divide asymmetrically per force retain one of the dividing daughter cells in the stem cell niche, even when treated with specific CBP/β-catenin antagonists, whereas CSCs can be removed from their niche via forced stochastic symmetric differentiative divisions. We now demonstrate that loss of p73 in early corticogenesis biases β-catenin Kat3 coactivator usage and enhances β-catenin/CBP transcription at the expense of β-catenin/p300 transcription. Biased β-catenin coactivator usage has dramatic consequences on the mode of division of neural stem cells (NSCs), but not neurogenic progenitors. The observed increase in symmetric divisions due to enhanced β-catenin/CBP interaction and transcription leads to an immediate increase in NSC symmetric differentiative divisions. Moreover, we demonstrate for the first time that the complex phenotype caused by the loss of p73 can be rescued in utero by treatment with the small-molecule-specific CBP/β-catenin antagonist ICG-001. Taken together, our results demonstrate the causal relationship between the choice of β-catenin Kat3 coactivator and the mode of stem cell division.
Collapse
Affiliation(s)
- Agnes I Lukaszewicz
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cu Nguyen
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Elizabeth Melendez
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - David P Lin
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Jia-Ling Teo
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Keane K Y Lai
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden 01307, Germany
| | - Song-Hai Shi
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael Kahn
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90033, USA.
- Center for Molecular Pathways and Drug Discovery, University of Southern California, Los Angeles, CA 90033, USA.
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA.
- Department of Molecular Pharmacology and Toxicology, University of Southern California, Los Angeles, CA 90033, USA.
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
18
|
Hannaford M, Loyer N, Tonelli F, Zoltner M, Januschke J. A chemical-genetics approach to study the role of atypical Protein Kinase C in Drosophila. Development 2019; 146:dev170589. [PMID: 30635282 PMCID: PMC6361133 DOI: 10.1242/dev.170589] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022]
Abstract
Studying the function of proteins using genetics in cycling cells is complicated by the fact that there is often a delay between gene inactivation and the time point of phenotypic analysis. This is particularly true when studying kinases that have pleiotropic functions and multiple substrates. Drosophila neuroblasts (NBs) are rapidly dividing stem cells and an important model system for the study of cell polarity. Mutations in multiple kinases cause NB polarity defects, but their precise functions at particular time points in the cell cycle are unknown. Here, we use chemical genetics and report the generation of an analogue-sensitive allele of Drosophila atypical Protein Kinase C (aPKC). We demonstrate that the resulting mutant aPKC kinase can be specifically inhibited in vitro and in vivo Acute inhibition of aPKC during NB polarity establishment abolishes asymmetric localization of Miranda, whereas its inhibition during NB polarity maintenance does not in the time frame of normal mitosis. However, aPKC helps to sharpen the pattern of Miranda, by keeping it off the apical and lateral cortex after nuclear envelope breakdown.
Collapse
Affiliation(s)
- Matthew Hannaford
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD5 1EH, UK
| | - Nicolas Loyer
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD5 1EH, UK
| | - Francesca Tonelli
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee DD5 1EH, UK
| | - Martin Zoltner
- Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dow Street, Dundee DD5 1EH, UK
| | - Jens Januschke
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD5 1EH, UK
| |
Collapse
|
19
|
Hannaford MR, Ramat A, Loyer N, Januschke J. aPKC-mediated displacement and actomyosin-mediated retention polarize Miranda in Drosophila neuroblasts. eLife 2018; 7:29939. [PMID: 29364113 PMCID: PMC5783611 DOI: 10.7554/elife.29939] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/14/2018] [Indexed: 02/07/2023] Open
Abstract
Cell fate assignment in the nervous system of vertebrates and invertebrates often hinges on the unequal distribution of molecules during progenitor cell division. We address asymmetric fate determinant localization in the developing Drosophila nervous system, specifically the control of the polarized distribution of the cell fate adapter protein Miranda. We reveal a step-wise polarization of Miranda in larval neuroblasts and find that Miranda’s dynamics and cortical association are differently regulated between interphase and mitosis. In interphase, Miranda binds to the plasma membrane. Then, before nuclear envelope breakdown, Miranda is phosphorylated by aPKC and displaced into the cytoplasm. This clearance is necessary for the subsequent establishment of asymmetric Miranda localization. After nuclear envelope breakdown, actomyosin activity is required to maintain Miranda asymmetry. Therefore, phosphorylation by aPKC and differential binding to the actomyosin network are required at distinct phases of the cell cycle to polarize fate determinant localization in neuroblasts.
Collapse
Affiliation(s)
- Matthew Robert Hannaford
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Anne Ramat
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Nicolas Loyer
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Jens Januschke
- Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
20
|
Chang WH, Choi SH, Moon BS, Cai M, Lyu J, Bai J, Gao F, Hajjali I, Zhao Z, Campbell DB, Weiner LP, Lu W. Smek1/2 is a nuclear chaperone and cofactor for cleaved Wnt receptor Ryk, regulating cortical neurogenesis. Proc Natl Acad Sci U S A 2017; 114:E10717-E10725. [PMID: 29180410 PMCID: PMC5740651 DOI: 10.1073/pnas.1715772114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The receptor-like tyrosine kinase (Ryk), a Wnt receptor, is important for cell fate determination during corticogenesis. During neuronal differentiation, the Ryk intracellular domain (ICD) is cleaved. Cleavage of Ryk and nuclear translocation of Ryk-ICD are required for neuronal differentiation. However, the mechanism of translocation and how it regulates neuronal differentiation remain unclear. Here, we identified Smek1 and Smek2 as Ryk-ICD partners that regulate its nuclear localization and function together with Ryk-ICD in the nucleus through chromatin recruitment and gene transcription regulation. Smek1/2 double knockout mice displayed pronounced defects in the production of cortical neurons, especially interneurons, while the neural stem cell population increased. In addition, both Smek and Ryk-ICD bound to the Dlx1/2 intergenic regulator element and were involved in its transcriptional regulation. These findings demonstrate a mechanism of the Ryk signaling pathway in which Smek1/2 and Ryk-ICD work together to mediate neural cell fate during corticogenesis.
Collapse
Affiliation(s)
- Wen-Hsuan Chang
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033
- The Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089
| | - Si Ho Choi
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033;
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, South Korea
| | - Byoung-San Moon
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033
| | - Mingyang Cai
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033
| | - Jungmook Lyu
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033
- Myung-Gok Eye Research Institute, Department of Medical Science, Konyang University, Daejeon 320-832, South Korea
| | - Jinlun Bai
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033
| | - Fan Gao
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033
| | - Ibrahim Hajjali
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033
| | - Zhongfang Zhao
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Daniel B Campbell
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Leslie P Weiner
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA 90033
| | - Wange Lu
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA 90033;
| |
Collapse
|
21
|
Yablonovitch AL, Fu J, Li K, Mahato S, Kang L, Rashkovetsky E, Korol AB, Tang H, Michalak P, Zelhof AC, Nevo E, Li JB. Regulation of gene expression and RNA editing in Drosophila adapting to divergent microclimates. Nat Commun 2017; 8:1570. [PMID: 29146998 PMCID: PMC5691062 DOI: 10.1038/s41467-017-01658-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022] Open
Abstract
Determining the mechanisms by which a species adapts to its environment is a key endeavor in the study of evolution. In particular, relatively little is known about how transcriptional processes are fine-tuned to adjust to different environmental conditions. Here we study Drosophila melanogaster from 'Evolution Canyon' in Israel, which consists of two opposing slopes with divergent microclimates. We identify several hundred differentially expressed genes and dozens of differentially edited sites between flies from each slope, correlate these changes with genetic differences, and use CRISPR mutagenesis to validate that an intronic SNP in prominin regulates its editing levels. We also demonstrate that while temperature affects editing levels at more sites than genetic differences, genetically regulated sites tend to be less affected by temperature. This work shows the extent to which gene expression and RNA editing differ between flies from different microclimates, and provides insights into the regulation responsible for these differences.
Collapse
Affiliation(s)
- Arielle L Yablonovitch
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Biophysics Program, Stanford University, Stanford, CA, 94305, USA
| | - Jeremy Fu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kexin Li
- Institute of Evolution, University of Haifa, Haifa, 3498838, Israel.,Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, 100093, Beijing, China
| | - Simpla Mahato
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Lin Kang
- Edward Via College of Osteopathic Medicine, Blacksburg, VA, 24060, USA
| | | | - Abraham B Korol
- Institute of Evolution, University of Haifa, Haifa, 3498838, Israel
| | - Hua Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Pawel Michalak
- Edward Via College of Osteopathic Medicine, Blacksburg, VA, 24060, USA.,Biocomplexity Institute, Virginia Tech, Blacksburg, VA, 24061, USA.,Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24060, USA
| | - Andrew C Zelhof
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Eviatar Nevo
- Institute of Evolution, University of Haifa, Haifa, 3498838, Israel.
| | - Jin Billy Li
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA. .,Biophysics Program, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
22
|
Hall ET, Pradhan-Sundd T, Samnani F, Verheyen EM. The protein phosphatase 4 complex promotes the Notch pathway and wingless transcription. Biol Open 2017; 6:1165-1173. [PMID: 28652317 PMCID: PMC5576076 DOI: 10.1242/bio.025221] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The Wnt/Wingless (Wg) pathway controls cell fate specification, tissue differentiation and organ development across organisms. Using an in vivo RNAi screen to identify novel kinase and phosphatase regulators of the Wg pathway, we identified subunits of the serine threonine phosphatase Protein Phosphatase 4 (PP4). Knockdown of the catalytic and regulatory subunits of PP4 cause reductions in the Wg pathway targets Senseless and Distal-less. We find that PP4 regulates the Wg pathway by controlling Notch-driven wg transcription. Genetic interaction experiments identified that PP4 likely promotes Notch signaling within the nucleus of the Notch-receiving cell. Although the PP4 complex is implicated in various cellular processes, its role in the regulation of Wg and Notch pathways was previously uncharacterized. Our study identifies a novel role of PP4 in regulating Notch pathway, resulting in aberrations in Notch-mediated transcriptional regulation of the Wingless ligand. Furthermore, we show that PP4 regulates proliferation independent of its interaction with Notch. Summary: The protein phosphatase 4 complex promotes Notch signaling and target gene expression during Drosophila wing development.
Collapse
Affiliation(s)
- Eric T Hall
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, British Columbia V5A 1S6, Canada
| | - Tirthadipa Pradhan-Sundd
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, British Columbia V5A 1S6, Canada
| | - Faaria Samnani
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, British Columbia V5A 1S6, Canada
| | - Esther M Verheyen
- Department of Molecular Biology and Biochemistry, Centre for Cell Biology, Development and Disease, Simon Fraser University, British Columbia V5A 1S6, Canada
| |
Collapse
|
23
|
Moon BS, Yun HM, Chang WH, Steele BH, Cai M, Choi SH, Lu W. Smek promotes corticogenesis through regulating Mbd3's stability and Mbd3/NuRD complex recruitment to genes associated with neurogenesis. PLoS Biol 2017; 15:e2001220. [PMID: 28467410 PMCID: PMC5414985 DOI: 10.1371/journal.pbio.2001220] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/06/2017] [Indexed: 11/18/2022] Open
Abstract
The fate of neural progenitor cells (NPCs) during corticogenesis is determined by a complex interplay of genetic or epigenetic components, but the underlying mechanism is incompletely understood. Here, we demonstrate that Suppressor of Mek null (Smek) interact with methyl-CpG-binding domain 3 (Mbd3) and the complex plays a critical role in self-renewal and neuronal differentiation of NPCs. We found that Smek promotes Mbd3 polyubiquitylation and degradation, blocking recruitment of the repressive Mbd3/nucleosome remodeling and deacetylase (NuRD) complex at the neurogenesis-associated gene loci, and, as a consequence, increasing acetyl histone H3 activity and cortical neurogenesis. Furthermore, overexpression of Mbd3 significantly blocked neuronal differentiation of NPCs, and Mbd3 depletion rescued neurogenesis defects seen in Smek1/2 knockout mice. These results reveal a novel molecular mechanism underlying Smek/Mbd3/NuRD axis-mediated control of NPCs' self-renewal and neuronal differentiation during mammalian corticogenesis.
Collapse
Affiliation(s)
- Byoung-San Moon
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Hyung-Mun Yun
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Wen-Hsuan Chang
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Bradford H. Steele
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Mingyang Cai
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Si Ho Choi
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Wange Lu
- Broad Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
24
|
Winfree LM, Speese SD, Logan MA. Protein phosphatase 4 coordinates glial membrane recruitment and phagocytic clearance of degenerating axons in Drosophila. Cell Death Dis 2017; 8:e2623. [PMID: 28230857 PMCID: PMC5386485 DOI: 10.1038/cddis.2017.40] [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: 07/15/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/15/2022]
Abstract
Neuronal damage induced by injury, stroke, or neurodegenerative disease elicits swift immune responses from glial cells, including altered gene expression, directed migration to injury sites, and glial clearance of damaged neurons through phagocytic engulfment. Collectively, these responses hinder further cellular damage, but the mechanisms that underlie these important protective glial reactions are still unclear. Here, we show that the evolutionarily conserved trimeric protein phosphatase 4 (PP4) serine/threonine phosphatase complex is a novel set of factors required for proper glial responses to nerve injury in the adult Drosophila brain. Glial-specific knockdown of PP4 results in reduced recruitment of glia to severed axons and delayed glial clearance of degenerating axonal debris. We show that PP4 functions downstream of the the glial engulfment receptor Draper to drive glial morphogenesis through the guanine nucleotide exchange factor SOS and the Rho GTPase Rac1, revealing that PP4 molecularly couples Draper to Rac1-mediated cytoskeletal remodeling to ensure glial infiltration of injury sites and timely removal of damaged neurons from the CNS.
Collapse
Affiliation(s)
- Lilly M Winfree
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Sean D Speese
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Mary A Logan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
25
|
Drosophila melanogaster Neuroblasts: A Model for Asymmetric Stem Cell Divisions. Results Probl Cell Differ 2017; 61:183-210. [PMID: 28409305 DOI: 10.1007/978-3-319-53150-2_8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Asymmetric cell division (ACD) is a fundamental mechanism to generate cell diversity, giving rise to daughter cells with different developmental potentials. ACD is manifested in the asymmetric segregation of proteins or mRNAs, when the two daughter cells differ in size or are endowed with different potentials to differentiate into a particular cell type (Horvitz and Herskowitz, Cell 68:237-255, 1992). Drosophila neuroblasts, the neural stem cells of the developing fly brain, are an ideal system to study ACD since this system encompasses all of these characteristics. Neuroblasts are intrinsically polarized cells, utilizing polarity cues to orient the mitotic spindle, segregate cell fate determinants asymmetrically, and regulate spindle geometry and physical asymmetry. The neuroblast system has contributed significantly to the elucidation of the basic molecular mechanisms underlying ACD. Recent findings also highlight its usefulness to study basic aspects of stem cell biology and tumor formation. In this review, we will focus on what has been learned about the basic mechanisms underlying ACD in fly neuroblasts.
Collapse
|
26
|
Loss of flfl Triggers JNK-Dependent Cell Death in Drosophila. BIOMED RESEARCH INTERNATIONAL 2015; 2015:623573. [PMID: 26583122 PMCID: PMC4637051 DOI: 10.1155/2015/623573] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/05/2015] [Indexed: 01/21/2023]
Abstract
falafel (flfl) encodes a Drosophila homolog of human SMEK whose in vivo functions remain elusive. In this study, we performed gain-of-function and loss-of-function analysis in Drosophila and identified flfl as a negative regulator of JNK pathway-mediated cell death. While ectopic expression of flfl suppresses TNF-triggered JNK-dependent cell death, loss of flfl promotes JNK activation and cell death in the developing eye and wing. These data report for the first time an essential physiological function of flfl in maintaining tissue homeostasis and organ development. As the JNK signaling pathway has been evolutionary conserved from fly to human, a similar role of PP4R3 in JNK-mediated physiological process is speculated.
Collapse
|
27
|
Lipinszki Z, Lefevre S, Savoian MS, Singleton MR, Glover DM, Przewloka MR. Centromeric binding and activity of Protein Phosphatase 4. Nat Commun 2015; 6:5894. [PMID: 25562660 PMCID: PMC4354016 DOI: 10.1038/ncomms6894] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 11/18/2014] [Indexed: 02/02/2023] Open
Abstract
The cell division cycle requires tight coupling between protein phosphorylation and dephosphorylation. However, understanding the cell cycle roles of multimeric protein phosphatases has been limited by the lack of knowledge of how their diverse regulatory subunits target highly conserved catalytic subunits to their sites of action. Phosphoprotein phosphatase 4 (PP4) has been recently shown to participate in the regulation of cell cycle progression. We now find that the EVH1 domain of the regulatory subunit 3 of Drosophila PP4, Falafel (Flfl), directly interacts with the centromeric protein C (CENP-C). Unlike other EVH1 domains that interact with proline-rich ligands, the crystal structure of the Flfl amino-terminal EVH1 domain bound to a CENP-C peptide reveals a new target-recognition mode for the phosphatase subunit. We also show that binding of Flfl to CENP-C is required to bring PP4 activity to centromeres to maintain CENP-C and attached core kinetochore proteins at chromosomes during mitosis.
Collapse
Affiliation(s)
- Zoltan Lipinszki
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Stephane Lefevre
- Macromolecular Structure and Function Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, UK
| | - Matthew S. Savoian
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Martin R. Singleton
- Macromolecular Structure and Function Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, UK
| | - David M. Glover
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - Marcin R. Przewloka
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| |
Collapse
|
28
|
Zhang F, Huang ZX, Bao H, Cong F, Wang H, Chai PC, Xi Y, Ge W, Somers WG, Yang Y, Cai Y, Yang X. Phosphotyrosyl phosphatase activator facilitates Miranda localization through dephosphorylation in dividing neuroblasts. Development 2015. [DOI: 10.1242/dev.127233] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
The mechanism for the basal targeting of the Miranda (Mira) complex during the asymmetric division of Drosophila neuroblasts (NBs) is yet to be fully understood. We have identified conserved Phosphotyrosyl Phosphatase Activator (PTPA) as a novel mediator for the basal localization of the Mira complex in larval brain NBs. In ptpa NBs, Mira remains cytoplasmic during early mitosis where its basal localization is delayed until anaphase. Detailed analyses indicate that PTPA acts independently of, and prior to, aPKC activity to localize Mira. Mechanistically, our data show that the phosphorylation status of the Thr591 (T591) residue determines the subcellular localization of Mira and that PTPA facilitates the dephosphorylation of T591. Furthermore, PTPA associates with the Protein Phosphatase 4 complex to mediate Mira localization. Based on these results, a two-step process for Mira basal localization during NB division is revealed where PTPA/PP4-mediated cortical association followed by apical aPKC-mediated basal restriction.
Collapse
Affiliation(s)
- Fan Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Zhen-Xing Huang
- Institute of Molecular and Cell Biology, ASTAR, Singapore
- Temasek Life Sciences Laboratory, Singapore
| | - Hongcun Bao
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Fei Cong
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | | | | | - Yongmei Xi
- Institute of Genetics, School of Medicine, Zhejiang University, Zhejiang, China
| | - Wanzhong Ge
- Institute of Genetics, School of Medicine, Zhejiang University, Zhejiang, China
| | - W. Gregory Somers
- Department of Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Australia
| | - Ying Yang
- Temasek Life Sciences Laboratory, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Yu Cai
- Temasek Life Sciences Laboratory, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Xiaohang Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Institute of Genetics, School of Medicine, Zhejiang University, Zhejiang, China
| |
Collapse
|
29
|
Drosophila neuroblasts as a new model for the study of stem cell self-renewal and tumour formation. Biosci Rep 2014; 34:BSR20140008. [PMID: 24965943 PMCID: PMC4114065 DOI: 10.1042/bsr20140008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Drosophila larval brain stem cells (neuroblasts) have emerged as an important model for the study of stem cell asymmetric division and the mechanisms underlying the transformation of neural stem cells into tumour-forming cancer stem cells. Each Drosophila neuroblast divides asymmetrically to produce a larger daughter cell that retains neuroblast identity, and a smaller daughter cell that is committed to undergo differentiation. Neuroblast self-renewal and differentiation are tightly controlled by a set of intrinsic factors that regulate ACD (asymmetric cell division). Any disruption of these two processes may deleteriously affect the delicate balance between neuroblast self-renewal and progenitor cell fate specification and differentiation, causing neuroblast overgrowth and ultimately lead to tumour formation in the fly. In this review, we discuss the mechanisms underlying Drosophila neural stem cell self-renewal and differentiation. Furthermore, we highlight emerging evidence in support of the notion that defects in ACD in mammalian systems, which may play significant roles in the series of pathogenic events leading to the development of brain cancers.
Collapse
|
30
|
Distinct phosphatases antagonize the p53 response in different phases of the cell cycle. Proc Natl Acad Sci U S A 2014; 111:7313-8. [PMID: 24711418 DOI: 10.1073/pnas.1322021111] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The basic machinery that detects DNA damage is the same throughout the cell cycle. Here, we show, in contrast, that reversal of DNA damage responses (DDRs) and recovery are fundamentally different in G1 and G2 phases of the cell cycle. We find that distinct phosphatases are required to counteract the checkpoint response in G1 vs. G2. Whereas WT p53-induced phosphatase 1 (Wip1) promotes recovery in G2-arrested cells by antagonizing p53, it is dispensable for recovery from a G1 arrest. Instead, we identify phosphoprotein phosphatase 4 catalytic subunit (PP4) to be specifically required for cell cycle restart after DNA damage in G1. PP4 dephosphorylates Krüppel-associated box domain-associated protein 1-S473 to repress p53-dependent transcriptional activation of p21 when the DDR is silenced. Taken together, our results show that PP4 and Wip1 are differentially required to counteract the p53-dependent cell cycle arrest in G1 and G2, by antagonizing early or late p53-mediated responses, respectively.
Collapse
|
31
|
Williams BC, Filter JJ, Blake-Hodek KA, Wadzinski BE, Fuda NJ, Shalloway D, Goldberg ML. Greatwall-phosphorylated Endosulfine is both an inhibitor and a substrate of PP2A-B55 heterotrimers. eLife 2014; 3:e01695. [PMID: 24618897 PMCID: PMC3949306 DOI: 10.7554/elife.01695] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 01/30/2014] [Indexed: 11/13/2022] Open
Abstract
During M phase, Endosulfine (Endos) family proteins are phosphorylated by Greatwall kinase (Gwl), and the resultant pEndos inhibits the phosphatase PP2A-B55, which would otherwise prematurely reverse many CDK-driven phosphorylations. We show here that PP2A-B55 is the enzyme responsible for dephosphorylating pEndos during M phase exit. The kinetic parameters for PP2A-B55's action on pEndos are orders of magnitude lower than those for CDK-phosphorylated substrates, suggesting a simple model for PP2A-B55 regulation that we call inhibition by unfair competition. As the name suggests, during M phase PP2A-B55's attention is diverted to pEndos, which binds much more avidly and is dephosphorylated more slowly than other substrates. When Gwl is inactivated during the M phase-to-interphase transition, the dynamic balance changes: pEndos dephosphorylated by PP2A-B55 cannot be replaced, so the phosphatase can refocus its attention on CDK-phosphorylated substrates. This mechanism explains simultaneously how PP2A-B55 and Gwl together regulate pEndos, and how pEndos controls PP2A-B55. DOI: http://dx.doi.org/10.7554/eLife.01695.001.
Collapse
Affiliation(s)
- Byron C Williams
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Joshua J Filter
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | | | - Brian E Wadzinski
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, United States
| | - Nicholas J Fuda
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - David Shalloway
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Michael L Goldberg
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| |
Collapse
|
32
|
Jusiak B, Karandikar UC, Kwak SJ, Wang F, Wang H, Chen R, Mardon G. Regulation of Drosophila eye development by the transcription factor Sine oculis. PLoS One 2014; 9:e89695. [PMID: 24586968 PMCID: PMC3934907 DOI: 10.1371/journal.pone.0089695] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/21/2014] [Indexed: 11/18/2022] Open
Abstract
Homeodomain transcription factors of the Sine oculis (SIX) family direct multiple regulatory processes throughout the metazoans. Sine oculis (So) was first characterized in the fruit fly Drosophila melanogaster, where it is both necessary and sufficient for eye development, regulating cell survival, proliferation, and differentiation. Despite its key role in development, only a few direct targets of So have been described previously. In the current study, we aim to expand our knowledge of So-mediated transcriptional regulation in the developing Drosophila eye using ChIP-seq to map So binding regions throughout the genome. We find 7,566 So enriched regions (peaks), estimated to map to 5,952 genes. Using overlap between the So ChIP-seq peak set and genes that are differentially regulated in response to loss or gain of so, we identify putative direct targets of So. We find So binding enrichment in genes not previously known to be regulated by So, including genes that encode cell junction proteins and signaling pathway components. In addition, we analyze a subset of So-bound novel genes in the eye, and find eight genes that have previously uncharacterized eye phenotypes and may be novel direct targets of So. Our study presents a greatly expanded list of candidate So targets and serves as basis for future studies of So-mediated gene regulation in the eye.
Collapse
Affiliation(s)
- Barbara Jusiak
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Umesh C. Karandikar
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Su-Jin Kwak
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Feng Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Hui Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rui Chen
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Graeme Mardon
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Cell and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
33
|
Lyu J, Kim HR, Yamamoto V, Choi SH, Wei Z, Joo CK, Lu W. Protein phosphatase 4 and Smek complex negatively regulate Par3 and promote neuronal differentiation of neural stem/progenitor cells. Cell Rep 2013; 5:593-600. [PMID: 24209749 PMCID: PMC3855259 DOI: 10.1016/j.celrep.2013.09.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 08/22/2013] [Accepted: 09/23/2013] [Indexed: 01/24/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent cells that can self-renew and differentiate into neurons and glial cells. However, mechanisms that control their fate decisions are poorly understood. Here, we show that Smek1, a regulatory subunit of the serine/threonine protein phosphatase PP4, promotes neuronal differentiation and suppresses the proliferative capacity of NPCs. We identify the cell polarity protein Par3, a negative regulator of neuronal differentiation, as a Smek1 substrate and demonstrate that Smek1 suppresses its activity. We also show that Smek1, which is predominantly nuclear in NPCs, is excluded from the nucleus during mitosis, allowing it to interact with cortical/cytoplasmic Par3 and mediate its dephosphorylation by the catalytic subunit PP4c. These results identify the PP4/Smek1 complex as a key regulator of neurogenesis.
Collapse
Affiliation(s)
- Jungmook Lyu
- Catholic Institute for Visual Science, Department of Ophthalmology and Visual Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, California 90042, USA
| | - Hee-Ryang Kim
- Catholic Institute for Visual Science, Department of Ophthalmology and Visual Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Vicky Yamamoto
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, California 90042, USA
| | - Si Ho Choi
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, California 90042, USA
| | - Zong Wei
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, California 90042, USA
| | - Choun-Ki Joo
- Catholic Institute for Visual Science, Department of Ophthalmology and Visual Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Wange Lu
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, California 90042, USA
| |
Collapse
|
34
|
Williams SE, Fuchs E. Oriented divisions, fate decisions. Curr Opin Cell Biol 2013; 25:749-58. [PMID: 24021274 DOI: 10.1016/j.ceb.2013.08.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 08/13/2013] [Accepted: 08/15/2013] [Indexed: 12/11/2022]
Abstract
During development, the establishment of proper tissue architecture depends upon the coordinated control of cell divisions not only in space and time, but also direction. Execution of an oriented cell division requires establishment of an axis of polarity and alignment of the mitotic spindle along this axis. Frequently, the cleavage plane also segregates fate determinants, either unequally or equally between daughter cells, the outcome of which is either an asymmetric or symmetric division, respectively. The last few years have witnessed tremendous growth in understanding both the extrinsic and intrinsic cues that position the mitotic spindle, the varied mechanisms in which the spindle orientation machinery is controlled in diverse organisms and organ systems, and the manner in which the division axis influences the signaling pathways that direct cell fate choices.
Collapse
Affiliation(s)
- Scott E Williams
- Department of Pathology & Laboratory Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
35
|
Abstract
In this issue of Neuron, Xie et al. (2013) identified protein phosphatase 4c (PP4c) as a new component in the regulation of spindle orientation during mammalian neurogenesis. Importantly, their findings uncovered a novel and critical temporal aspect of the regulation of spindle orientation during neurogenesis.
Collapse
Affiliation(s)
- Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
36
|
Goh LH, Zhou X, Lee MC, Lin S, Wang H, Luo Y, Yang X. Clueless regulates aPKC activity and promotes self-renewal cell fate in Drosophila lgl mutant larval brains. Dev Biol 2013; 381:353-64. [PMID: 23835532 DOI: 10.1016/j.ydbio.2013.06.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 06/13/2013] [Accepted: 06/30/2013] [Indexed: 11/16/2022]
Abstract
Asymmetric cell division of Drosophila neural stem cells or neuroblasts is an important process which gives rise to two different daughter cells, one of which is the stem cell itself and the other, a committed or differentiated daughter cell. During neuroblast asymmetric division, atypical Protein Kinase C (aPKC) activity is tightly regulated; aberrant levels of activity could result in tumorigenesis in third instar larval brain. We identified clueless (clu), a genetic interactor of parkin (park), as a novel regulator of aPKC activity. It preferentially binds to the aPKC/Bazooka/Partition Defective 6 complex and stabilizes aPKC levels. In clu mutants, Miranda (Mira) and Numb are mislocalized in small percentages of dividing neuroblasts. Adult mutants are short-lived with severe locomotion defects. Clu promotes tumorigenesis caused by loss of function of lethal(2) giant larvae (lgl) in the larval brain. Removal of clu in lgl mutants rescues Mira and Numb mislocalization and restores the enlarged brain size. Western blot analyses indicate that the rescue is due to the down-regulation of aPKC levels in the lgl clu double mutant. Interestingly, the phenotype of the park mutant, which causes Parkinson's Disease-like symptoms in adult flies, is reminiscent of that of clu in neuroblast asymmetric division. Our study provides the first clue for the potential missing pathological link between temporally separated neurogenesis and neurodegeneration events; the minor defects during early neurogenesis could be a susceptible factor contributing to neurodegenerative diseases at later stages of life.
Collapse
Affiliation(s)
- Li Hui Goh
- Institute of Molecular and Cell Biology, Singapore
| | | | | | | | | | | | | |
Collapse
|
37
|
Xie Y, Jüschke C, Esk C, Hirotsune S, Knoblich JA. The phosphatase PP4c controls spindle orientation to maintain proliferative symmetric divisions in the developing neocortex. Neuron 2013; 79:254-65. [PMID: 23830831 PMCID: PMC3725415 DOI: 10.1016/j.neuron.2013.05.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2013] [Indexed: 12/28/2022]
Abstract
In the developing neocortex, progenitor cells expand through symmetric division before they generate cortical neurons through multiple rounds of asymmetric cell division. Here, we show that the orientation of the mitotic spindle plays a crucial role in regulating the transition between those two division modes. We demonstrate that the protein phosphatase PP4c regulates spindle orientation in early cortical progenitor cells. Upon removing PP4c, mitotic spindles fail to orient in parallel to the neuroepithelial surface and progenitors divide with random orientation. As a result, their divisions become asymmetric and neurogenesis starts prematurely. Biochemical and genetic experiments show that PP4c acts by dephosphorylating the microtubule binding protein Ndel1, thereby enabling complex formation with Lis1 to form a functional spindle orientation complex. Our results identify a key regulator of cortical development and demonstrate that changes in the orientation of progenitor division are responsible for the transition between symmetric and asymmetric cell division. PP4c is required for spindle orientation in cortical progenitors Loss of PP4c leads to premature neuronal differentiation Spindle misorientation causes layering defects during a critical time window PP4c acts on Ndel1 and Lis1
Collapse
Affiliation(s)
- Yunli Xie
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Dr. Bohr-Gasse 3-5, 1030 Vienna, Austria
| | | | | | | | | |
Collapse
|
38
|
Theobald B, Bonness K, Musiyenko A, Andrews JF, Urban G, Huang X, Dean NM, Honkanen RE. Suppression of Ser/Thr phosphatase 4 (PP4C/PPP4C) mimics a novel post-mitotic action of fostriecin, producing mitotic slippage followed by tetraploid cell death. Mol Cancer Res 2013; 11:845-55. [PMID: 23671329 DOI: 10.1158/1541-7786.mcr-13-0032] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Fostriecin is a natural product purified from Sterptomyces extracts with antitumor activity sufficient to warrant human clinical trials. Unfortunately, difficulties associated with supply and stable drug formulation stalled further development. At a molecular level, fostriecin is known to act as a catalytic inhibitor of four PPP-family phosphatases, and reports describing the design of molecules in this class suggest derivatives targeting enzymes within the fostriecin-sensitive subfamily can be successful. However, it is not clear if the tumor-selective cytotoxicity of fostriecin results from the inhibition of a specific phosphatase, multiple phosphatases, or a limited subset of fostriecin sensitive phosphatases. How the inhibition of sensitive phosphatases contributes to tumor-selective cytotoxicity is also not clear. Here, high-content time-lapse imaging of live cells revealed novel insight into the cellular actions of fostriecin, showing that fostriecin-induced apoptosis is not simply induced following a sustained mitotic arrest. Rather, apoptosis occurred in an apparent second interphase produced when tetraploid cells undergo mitotic slippage. Comparison of the actions of fostriecin and antisense-oligonucleotides specifically targeting human fostriecin-sensitive phosphatases revealed that the suppression PP4C alone is sufficient to mimic many actions of fostriecin. Importantly, targeted suppression of PP4C induced apoptosis, with death occurring in tetraploid cells following mitotic slippage. This effect was not observed following the suppression of PP1C, PP2AC, or PP5C. These data clarify PP4C as a fostriecin-sensitive phosphatase and demonstrate that the suppression of PP4C triggers mitotic slippage/apoptosis. IMPLICATIONS Future development of fostriecin class inhibitors should consider PP4C as a potentially important target. Mol Cancer Res; 11(8); 845-55. ©2013 AACR.
Collapse
Affiliation(s)
- Benjamin Theobald
- Department of Biochemistry and Molecular Biology, MSB 2362, University of South Alabama, Mobile, AL 36688, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Hedgehog signaling acts with the temporal cascade to promote neuroblast cell cycle exit. PLoS Biol 2013; 11:e1001494. [PMID: 23468593 PMCID: PMC3582610 DOI: 10.1371/journal.pbio.1001494] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
During the development of the Drosophila nervous system, the developmentally regulated Hedgehog pathway, together with a series of temporal transcription factors, schedules the end of neurogenesis. In Drosophila postembryonic neuroblasts, transition in gene expression programs of a cascade of transcription factors (also known as the temporal series) acts together with the asymmetric division machinery to generate diverse neurons with distinct identities and regulate the end of neuroblast proliferation. However, the underlying mechanism of how this “temporal series” acts during development remains unclear. Here, we show that Hh signaling in the postembryonic brain is temporally regulated; excess (earlier onset of) Hh signaling causes premature neuroblast cell cycle exit and under-proliferation, whereas loss of Hh signaling causes delayed cell cycle exit and excess proliferation. Moreover, the Hh pathway functions downstream of Castor but upstream of Grainyhead, two components of the temporal series, to schedule neuroblast cell cycle exit. Interestingly, hh is likely a target of Castor. Hence, Hh signaling provides a link between the temporal series and the asymmetric division machinery in scheduling the end of neurogenesis. In almost all metazoans, neurons are produced by a group of neural stem cells/progenitors in a precise temporal manner, which is important for generating a functional nervous system. In Drosophila, this “timing” mechanism is mainly governed by the sequential switching of transcription factors in neural stem cells called neuroblasts, such that neuronal fate is associated with its birth order. These temporal factors also coordinate the termination of neuroblast division towards the end of neurogenesis. In this study, we show that Hedgehog (Hh) signaling also regulates the division rate of neuroblasts during their proliferative phase at larval stage, as well as the cessation of proliferation at early pupal stage. Excessive Hh signaling causes premature neuroblast cell cycle exit and early termination of neurogenesis, while loss of Hh signaling results in prolonged proliferation of neuroblasts beyond its physiological window. We also find that Hh signaling acts in concert with the temporal transcription factors, and is itself regulated by these factors. We hypothesize that this mode of interaction (temporal transcription factors with developmentally regulated signals like Hh) during neurogenesis could be widely conserved in other organisms.
Collapse
|
40
|
Hartenstein V, Wodarz A. Initial neurogenesis in Drosophila. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 2:701-21. [PMID: 24014455 DOI: 10.1002/wdev.111] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Early neurogenesis comprises the phase of nervous system development during which neural progenitor cells are born. In early development, the embryonic ectoderm is subdivided by a conserved signaling mechanism into two main domains, the epidermal ectoderm and the neurectoderm. Subsequently, cells of the neurectoderm are internalized and form a cell layer of proliferating neural progenitors. In vertebrates, the entire neurectoderm folds into the embryo to give rise to the neural tube. In Drosophila and many other invertebrates, a subset of neurectodermal cells, called neuroblasts (NBs), delaminates and forms the neural primordium inside the embryo where they divide in an asymmetric, stem cell-like mode. The remainder of the neurectodermal cells that stay behind at the surface loose their neurogenic potential and later give rise to the ventral part of the epidermis. The genetic and molecular analysis of the mechanisms controlling specification and proliferation of NBs in the Drosophila embryo, which played a significant part in pioneering the field of modern developmental neurobiology, represents the topic of this review.
Collapse
Affiliation(s)
- Volker Hartenstein
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
41
|
Sousa-Nunes R, Somers WG. Mechanisms of asymmetric progenitor divisions in the Drosophila central nervous system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:79-102. [PMID: 23696353 DOI: 10.1007/978-94-007-6621-1_6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Drosophila central nervous system develops from polarised asymmetric divisions of precursor cells, called neuroblasts. Decades of research on neuroblasts have resulted in a substantial understanding of the factors and molecular events responsible for fate decisions of neuroblasts and their progeny. Furthermore, the cell-cycle dependent mechanisms responsible for asymmetric cortical protein localisation, resulting in the unequal partitioning between daughters, are beginning to be exposed. Disruption to the appropriate partitioning of proteins between neuroblasts and differentiation-committed daughters can lead to supernumerary neuroblast-like cells and the formation of tumours. Many of the factors responsible for regulating asymmetric division of Drosophila neuroblasts are evolutionarily conserved and, in many cases, have been shown to play a functionally conserved role in mammalian neurogenesis. Recent genome-wide studies coupled with advancements in live-imaging technologies have opened further avenues of research into neuroblast biology. We review our current understanding of the molecular mechanisms regulating neuroblast divisions, a powerful system to model mammalian neurogenesis and tumourigenesis.
Collapse
Affiliation(s)
- Rita Sousa-Nunes
- MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, London, SE1 1UL, UK.
| | | |
Collapse
|
42
|
Crona F, Dahlberg O, Lundberg LE, Larsson J, Mannervik M. Gene regulation by the lysine demethylase KDM4A in Drosophila. Dev Biol 2012. [PMID: 23195220 DOI: 10.1016/j.ydbio.2012.11.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lysine methylation of histones is associated with both transcriptionally active chromatin and with silent chromatin, depending on what residue is modified. Histone methyltransferases and demethylases ensure that histone methylations are dynamic and can vary depending on cell cycle- or developmental stage. KDM4A demethylates H3K36me3, a modification enriched in the 3' end of active genes. The genomic targets and the role of KDM4 proteins in development remain largely unknown. We therefore generated KDM4A mutant Drosophila, and identified 99 mis-regulated genes in first instar larvae. Around half of these genes were down-regulated and the other half up-regulated in dKDM4A mutants. Although heterochromatin protein 1a (HP1a) can stimulate dKDM4A demethylase activity in vitro, we find that they antagonize each other in control of dKDM4A-regulated genes. Appropriate expression levels for some dKDM4A-regulated genes rely on the demethylase activity of dKDM4A, whereas others do not. Surprisingly, although highly expressed, many demethylase-dependent and independent genes are devoid of H3K36me3 in wild-type as well as in dKDM4A mutant larvae, suggesting that some of the most strongly affected genes in dKDM4A mutant animals are not regulated by H3K36 methylation. By contrast, dKDM4A over-expression results in a global decrease in H3K36me3 levels and male lethality, which might be caused by impaired dosage compensation. Our results show that a modest increase in global H3K36me3 levels is compatible with viability, fertility, and the expression of most genes, whereas decreased H3K36me3 levels are detrimental in males.
Collapse
Affiliation(s)
- Filip Crona
- Stockholm University, Wenner-Gren Institute, Developmental Biology, Arrhenius laboratories E3, SE-106 91 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
43
|
Berger C, Harzer H, Burkard T, Steinmann J, van der Horst S, Laurenson AS, Novatchkova M, Reichert H, Knoblich J. FACS purification and transcriptome analysis of drosophila neural stem cells reveals a role for Klumpfuss in self-renewal. Cell Rep 2012; 2:407-18. [PMID: 22884370 PMCID: PMC3828055 DOI: 10.1016/j.celrep.2012.07.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 06/27/2012] [Accepted: 07/23/2012] [Indexed: 12/05/2022] Open
Abstract
Drosophila neuroblasts (NBs) have emerged as a model for stem cell biology that is ideal for genetic analysis but is limited by the lack of cell-type-specific gene expression data. Here, we describe a method for isolating large numbers of pure NBs and differentiating neurons that retain both cell-cycle and lineage characteristics. We determine transcriptional profiles by mRNA sequencing and identify 28 predicted NB-specific transcription factors that can be arranged in a network containing hubs for Notch signaling, growth control, and chromatin regulation. Overexpression and RNA interference for these factors identify Klumpfuss as a regulator of self-renewal. We show that loss of Klumpfuss function causes premature differentiation and that overexpression results in the formation of transplantable brain tumors. Our data represent a valuable resource for investigating Drosophila developmental neurobiology, and the described method can be applied to other invertebrate stem cell lineages as well.
Collapse
Affiliation(s)
- Christian Berger
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Heike Harzer
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Thomas R. Burkard
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Jonas Steinmann
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Suzanne van der Horst
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | | | - Maria Novatchkova
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
- Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, 1030 Vienna, Austria
| | - Heinrich Reichert
- University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Juergen A. Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Dr. Bohr-Gasse 3, 1030 Vienna, Austria
- Corresponding author
| |
Collapse
|
44
|
Chang KC, Wang C, Wang H. Balancing self-renewal and differentiation by asymmetric division: insights from brain tumor suppressors in Drosophila neural stem cells. Bioessays 2012; 34:301-10. [PMID: 22287225 DOI: 10.1002/bies.201100090] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Balancing self-renewal and differentiation of stem cells is an important issue in stem cell and cancer biology. Recently, the Drosophila neuroblast (NB), neural stem cell has emerged as an excellent model for stem cell self-renewal and tumorigenesis. It is of great interest to understand how defects in the asymmetric division of neural stem cells lead to tumor formation. Here, we review recent advances in asymmetric division and the self-renewal control of Drosophila NBs. We summarize molecular mechanisms of asymmetric cell division and discuss how the defects in asymmetric division lead to tumor formation. Gain-of-function or loss-of-function of various proteins in the asymmetric machinery can drive NB overgrowth and tumor formation. These proteins control either the asymmetric protein localization or mitotic spindle orientation of NBs. We also discuss other mechanisms of brain tumor suppression that are beyond the control of asymmetric division.
Collapse
Affiliation(s)
- Kai Chen Chang
- Neuroscience & Behavioral Disorder Program, Duke-NUS Graduate Medical School Singapore, Singapore, Singapore
| | | | | |
Collapse
|
45
|
Neumüller RA, Richter C, Fischer A, Novatchkova M, Neumüller KG, Knoblich JA. Genome-wide analysis of self-renewal in Drosophila neural stem cells by transgenic RNAi. Cell Stem Cell 2011; 8:580-93. [PMID: 21549331 PMCID: PMC3093620 DOI: 10.1016/j.stem.2011.02.022] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 11/01/2010] [Accepted: 02/16/2011] [Indexed: 01/14/2023]
Abstract
The balance between stem cell self-renewal and differentiation is precisely controlled to ensure tissue homeostasis and prevent tumorigenesis. Here we use genome-wide transgenic RNAi to identify 620 genes potentially involved in controlling this balance in Drosophila neuroblasts. We quantify all phenotypes and derive measurements for proliferation, lineage, cell size, and cell shape. We identify a set of transcriptional regulators essential for self-renewal and use hierarchical clustering and integration with interaction data to create functional networks for the control of neuroblast self-renewal and differentiation. Our data identify key roles for the chromatin remodeling Brm complex, the spliceosome, and the TRiC/CCT-complex and show that the alternatively spliced transcription factor Lola and the transcriptional elongation factors Ssrp and Barc control self-renewal in neuroblast lineages. As our data are strongly enriched for genes highly expressed in murine neural stem cells, they are likely to provide valuable insights into mammalian stem cell biology as well.
Collapse
Affiliation(s)
- Ralph A Neumüller
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
46
|
Miskei M, Ádám C, Kovács L, Karányi Z, Dombrádi V. Molecular evolution of phosphoprotein phosphatases in Drosophila. PLoS One 2011; 6:e22218. [PMID: 21789237 PMCID: PMC3137614 DOI: 10.1371/journal.pone.0022218] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 06/20/2011] [Indexed: 12/25/2022] Open
Abstract
Phosphoprotein phosphatases (PPP), these ancient and important regulatory enzymes are present in all eukaryotic organisms. Based on the genome sequences of 12 Drosophila species we traced the evolution of the PPP catalytic subunits and noted a substantial expansion of the gene family. We concluded that the 18–22 PPP genes of Drosophilidae were generated from a core set of 8 indispensable phosphatases that are present in most of the insects. Retropositons followed by tandem gene duplications extended the phosphatase repertoire, and sporadic gene losses contributed to the species specific variations in the PPP complement. During the course of these studies we identified 5, up till now uncharacterized phosphatase retrogenes: PpY+, PpD5+, PpD6+, Pp4+, and Pp6+ which are found only in some ancient Drosophila. We demonstrated that all of these new PPP genes exhibit a distinct male specific expression. In addition to the changes in gene numbers, the intron-exon structure and the chromosomal localization of several PPP genes was also altered during evolution. The G−C content of the coding regions decreased when a gene moved into the heterochromatic region of chromosome Y. Thus the PPP enzymes exemplify the various types of dynamic rearrangements that accompany the molecular evolution of a gene family in Drosophilidae.
Collapse
Affiliation(s)
- Márton Miskei
- Centre for Agricultural and Applied Economic Sciences, Institute of Horticultural Sciences, Department of Plant Biotechnology, University of Debrecen, Debrecen, Hungary
| | - Csaba Ádám
- Department of Medical Chemistry, Research Center for Molecular Medicine, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - László Kovács
- Department of Medical Chemistry, Research Center for Molecular Medicine, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | - Zsolt Karányi
- First Department of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktor Dombrádi
- Department of Medical Chemistry, Research Center for Molecular Medicine, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
- * E-mail:
| |
Collapse
|
47
|
Sousa-Nunes R, Somers WG. Phosphorylation and dephosphorylation events allow for rapid segregation of fate determinants during Drosophila neuroblast asymmetric divisions. Commun Integr Biol 2011; 3:46-9. [PMID: 20539782 DOI: 10.4161/cib.3.1.9635] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 07/24/2009] [Indexed: 01/29/2023] Open
Abstract
Drosophila neuroblasts display remarkable asymmetry throughout mitosis. The most prominent asymmetry is the size difference between daughter cells at cytokinesis. The larger cell retains stem cell identity, i.e., remains a neuroblast while the smaller cell, called a ganglion mother cell (GMC), will generate differentiated neural and glial progeny. Preceding this size difference, several protein complexes localize to opposite sides of the neuroblast cortex (apical and basal in the embryo and, by analogy, referred to as such in larval neuroblasts although their asymmetry no longer correlates with such axis). The plane of division is coordinated with this molecular asymmetry such that apical and basal complexes are unequally partitioned between the two daughter cells: apical complexes are inherited by the self-renewing neuroblast while basal complexes are inherited by the GMC. This unequal segregation has been extensively shown to be functionally significant. Apical complexes contain factors required for cellular selfrenewal and basal complexes contain factors required for the differentiation of the GMC progeny. Curiously, however, some "basal" neuroblast proteins such as the scaffold protein Miranda (Mira) and its associated fate determinant Prospero (Pros), are initially apically localized prior to translocating to the opposite side of the cell cortex by the onset of mitosis. This is because mira mRNA is apically enriched, where it remains throughout the cell cycle, suggesting that Mira protein is translated within the apical environment.1,2 The transition from apical to basal enrichment of Mira and Pros takes place within minutes.2 Here, we summarize the known phosphorylation events and roles during neuroblast asymmetric divisions, as well as very recent work, including our own, identifying the first protein phosphatases implicated in this process. We then discuss models previously proposed, as well as a new model, for apical-to-basal transition of the Mira complex in light of our new results.
Collapse
|
48
|
Lyu J, Jho EH, Lu W. Smek promotes histone deacetylation to suppress transcription of Wnt target gene brachyury in pluripotent embryonic stem cells. Cell Res 2011; 21:911-21. [PMID: 21423269 PMCID: PMC3203701 DOI: 10.1038/cr.2011.47] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 11/22/2010] [Accepted: 11/25/2010] [Indexed: 01/03/2023] Open
Abstract
In embryonic stem cells (ESCs), Wnt-responsive development-related genes are silenced to maintain pluripotency and their expression is activated during differentiation. Acetylation of histones by histone acetyltransferases stimulates transcription, whereas deacetylation of histones by HDACs is correlated with transcriptional repression. Although Wnt-mediated gene transcription has been intimately linked to the acetylation or deacetylation of histones, how Wnt signaling regulates this type of histone modification is poorly understood. Here, we report that Smek, a regulatory subunit of protein phosphatase 4 (PP4) complex, plays an important role in histone deacetylation and silencing of the Wnt-responsive gene, brachyury, in ESCs. Smek mediates recruitment of PP4c and HDAC1 to the Tcf/Lef binding site of the brachyury promoter and inhibits brachyury expression in ESCs. Activation of Wnt signaling during differentiation causes disruption of the Smek/PP4c/HDAC1 complex, resulting in an increase in histones H3 and H4 acetylation at the brachyury gene locus. These results suggest that the Smek-containing PP4 complex represses transcription of Wnt-responsive development-related genes through histone deacetylation, and that this complex is essential for ESC pluripotency maintenance.
Collapse
Affiliation(s)
- Jungmook Lyu
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Eek-hoon Jho
- Department of Life Science, University of Seoul, Seoul 130-743, Korea
| | - Wange Lu
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
- Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
49
|
Chang KC, Garcia-Alvarez G, Somers G, Sousa-Nunes R, Rossi F, Lee YY, Soon SB, Gonzalez C, Chia W, Wang H. Interplay between the transcription factor Zif and aPKC regulates neuroblast polarity and self-renewal. Dev Cell 2010; 19:778-85. [PMID: 21074726 DOI: 10.1016/j.devcel.2010.10.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 08/17/2010] [Accepted: 09/14/2010] [Indexed: 12/14/2022]
Abstract
How a cell decides to self-renew or differentiate is a critical issue in stem cell and cancer biology. Atypical protein kinase C (aPKC) promotes self-renewal of Drosophila larval brain neural stem cells, neuroblasts. However, it is unclear how aPKC cortical polarity and protein levels are regulated. Here, we have identified a zinc-finger protein, Zif, which is required for the expression and asymmetric localization of aPKC. aPKC displays ectopic cortical localization with upregulated protein levels in dividing zif mutant neuroblasts, leading to neuroblast overproliferation. We show that Zif is a transcription factor that directly represses aPKC transcription. We further show that Zif is phosphorylated by aPKC both in vitro and in vivo. Phosphorylation of Zif by aPKC excludes it from the nucleus, leading to Zif inactivation in neuroblasts. Thus, reciprocal repression between Zif and aPKC act as a critical regulatory mechanism for establishing cell polarity and controlling neuroblast self-renewal.
Collapse
Affiliation(s)
- Kai Chen Chang
- Temasek Life Sciences Laboratory, 1 Research Link, Singapore 117604
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Prehoda KE. Polarization of Drosophila neuroblasts during asymmetric division. Cold Spring Harb Perspect Biol 2010; 1:a001388. [PMID: 20066083 DOI: 10.1101/cshperspect.a001388] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During Drosophila development, neuroblasts divide to generate progeny with two different fates. One daughter cell self-renews to maintain the neuroblast pool, whereas the other differentiates to populate the central nervous system. The difference in fate arises from the asymmetric distribution of proteins that specify either self-renewal or differentiation, which is brought about by their polarization into separate apical and basal cortical domains during mitosis. Neuroblast symmetry breaking is regulated by numerous proteins, many of which have only recently been discovered. The atypical protein kinase C (aPKC) is a broad regulator of polarity that localizes to the neuroblast apical cortical region and directs the polarization of the basal domain. Recent work suggests that polarity can be explained in large part by the mechanisms that restrict aPKC activity to the apical domain and those that couple asymmetric aPKC activity to the polarization of downstream factors. Polarized aPKC activity is created by a network of regulatory molecules, including Bazooka/Par-3, Cdc42, and the tumor suppressor Lgl, which represses basal recruitment. Direct phosphorylation by aPKC leads to cortical release of basal domain factors, preventing them from occupying the apical domain. In this framework, neuroblast polarity arises from a complex system that orchestrates robust aPKC polarity, which in turn polarizes substrates by coupling phosphorylation to cortical release.
Collapse
Affiliation(s)
- Kenneth E Prehoda
- Department of Chemistry and Institute of Molecular Biology, University of Oregon, Eugene, Oregon 97403, USA.
| |
Collapse
|