1
|
Purice MD, Lago‐Baldaia I, Fernandes VM, Singhvi A. Molecular profiling of invertebrate glia. Glia 2025; 73:632-656. [PMID: 39415317 PMCID: PMC11784859 DOI: 10.1002/glia.24623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/18/2024]
Abstract
Caenorhabditis elegans and Drosophila melanogaster are powerful experimental models for uncovering fundamental tenets of nervous system organization and function. Findings over the last two decades show that molecular and cellular features are broadly conserved between invertebrates and vertebrates, indicating that insights derived from invertebrate models can broadly inform our understanding of glial operating principles across diverse species. In recent years, these model systems have led to exciting discoveries in glial biology and mechanisms of glia-neuron interactions. Here, we summarize studies that have applied current state-of-the-art "-omics" techniques to C. elegans and D. melanogaster glia. Coupled with the remarkable acceleration in the pace of mechanistic studies of glia biology in recent years, these indicate that invertebrate glia also exhibit striking molecular complexity, specificity, and heterogeneity. We provide an overview of these studies and discuss their implications as well as emerging questions where C. elegans and D. melanogaster are well-poised to fill critical knowledge gaps in our understanding of glial biology.
Collapse
Affiliation(s)
- Maria D. Purice
- Division of Basic SciencesFred Hutchinson Cancer CenterSeattleWashingtonUSA
- Department of Biological StructureSchool of Medicine, University of WashingtonSeattleWashingtonUSA
| | - Inês Lago‐Baldaia
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | | | - Aakanksha Singhvi
- Division of Basic SciencesFred Hutchinson Cancer CenterSeattleWashingtonUSA
- Department of Biological StructureSchool of Medicine, University of WashingtonSeattleWashingtonUSA
| |
Collapse
|
2
|
Parisot N, Ribeiro Lopes M, Peignier S, Baa-Puyoulet P, Charles H, Calevro F, Callaerts P. Annotation of transcription factors, chromatin-associated factors, and basal transcription machinery in the pea aphid, Acyrthosiphon pisum, and development of the ATFdb database, a resource for studies of transcriptional regulation. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 177:104217. [PMID: 39579797 DOI: 10.1016/j.ibmb.2024.104217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/15/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
The pea aphid, Acyrthosiphon pisum, is an emerging model system in functional and comparative genomics, in part due to the availability of new genomic approaches and the different sequencing and annotation efforts that the community has dedicated to this important crop pest insect. The pea aphid is also used as a model to study fascinating biological traits of aphids, such as their extensive polyphenisms, their bacteriocyte-confined nutritional symbiosis, or their adaptation to the highly unbalanced diet represented by phloem sap. To get insights into the molecular basis of all these processes, it is important to have an appropriate annotation of transcription factors (TFs), which would enable the reconstruction/inference of gene regulatory networks in aphids. Using the latest version of the A. pisum genome assembly and annotation, which represents the first chromosome-level pea aphid genome, we annotated the complete repertoire of A. pisum TFs and complemented this information by annotating genes encoding chromatin-associated and basal transcription machinery proteins. These annotations were done combining information from the model Drosophila melanogaster, for which we also provide a revisited list of these proteins, and de novo prediction. The comparison between the two model systems allowed the identification of major losses or expansions in each genome, while a deeper analysis was made of ZNF TFs (with certain families expanded in the pea aphid), and the Hox gene cluster (showing reorganization in gene position in the pea aphid compared to D. melanogaster). All annotations are available to the community through the Aphid Transcription Factors database (ATFdb), consolidating the various annotations we generated. ATFdb serves as a valuable resource for gene regulation studies in aphids.
Collapse
Affiliation(s)
- Nicolas Parisot
- INSA Lyon, INRAE, BF2I, UMR0203, F-69621, Villeurbanne, France.
| | | | - Sergio Peignier
- INSA Lyon, INRAE, BF2I, UMR0203, F-69621, Villeurbanne, France
| | | | - Hubert Charles
- INSA Lyon, INRAE, BF2I, UMR0203, F-69621, Villeurbanne, France
| | | | - Patrick Callaerts
- KU Leuven, University of Leuven, Department of Human Genetics, Laboratory of Behavioral and Developmental Genetics, B-3000, Leuven, Belgium.
| |
Collapse
|
3
|
Sheloukhova L, Watanabe H. Evolution of glial cells: a non-bilaterian perspective. Neural Dev 2024; 19:10. [PMID: 38907299 PMCID: PMC11193209 DOI: 10.1186/s13064-024-00184-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 06/06/2024] [Indexed: 06/23/2024] Open
Abstract
Nervous systems of bilaterian animals generally consist of two cell types: neurons and glial cells. Despite accumulating data about the many important functions glial cells serve in bilaterian nervous systems, the evolutionary origin of this abundant cell type remains unclear. Current hypotheses regarding glial evolution are mostly based on data from model bilaterians. Non-bilaterian animals have been largely overlooked in glial studies and have been subjected only to morphological analysis. Here, we provide a comprehensive overview of conservation of the bilateral gliogenic genetic repertoire of non-bilaterian phyla (Cnidaria, Placozoa, Ctenophora, and Porifera). We overview molecular and functional features of bilaterian glial cell types and discuss their possible evolutionary history. We then examine which glial features are present in non-bilaterians. Of these, cnidarians show the highest degree of gliogenic program conservation and may therefore be crucial to answer questions about glial evolution.
Collapse
Affiliation(s)
- Larisa Sheloukhova
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Hiroshi Watanabe
- Evolutionary Neurobiology Unit, Okinawa Institute of Science and Technology, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa, 904-0412, Japan.
| |
Collapse
|
4
|
Agi E, Reifenstein ET, Wit C, Schneider T, Kauer M, Kehribar M, Kulkarni A, von Kleist M, Hiesinger PR. Axonal self-sorting without target guidance in Drosophila visual map formation. Science 2024; 383:1084-1092. [PMID: 38452066 DOI: 10.1126/science.adk3043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/01/2024] [Indexed: 03/09/2024]
Abstract
The idea of guidance toward a target is central to axon pathfinding and brain wiring in general. In this work, we show how several thousand axonal growth cones self-pattern without target-dependent guidance during neural superposition wiring in Drosophila. Ablation of all target lamina neurons or loss of target adhesion prevents the stabilization but not the development of the pattern. Intravital imaging at the spatiotemporal resolution of growth cone dynamics in intact pupae and data-driven dynamics simulations reveal a mechanism by which >30,000 filopodia do not explore potential targets, but instead simultaneously generate and navigate a dynamic filopodial meshwork that steers growth directions. Hence, a guidance mechanism can emerge from the interactions of the axons being guided, suggesting self-organization as a more general feature of brain wiring.
Collapse
Affiliation(s)
- Egemen Agi
- Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany
| | - Eric T Reifenstein
- Department of Mathematics, Free University of Berlin, 14195 Berlin, Germany
| | - Charlotte Wit
- Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany
| | - Teresa Schneider
- Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany
| | - Monika Kauer
- Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany
| | - Melinda Kehribar
- Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany
| | - Abhishek Kulkarni
- Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany
| | - Max von Kleist
- Department of Mathematics, Free University of Berlin, 14195 Berlin, Germany
| | - P Robin Hiesinger
- Division of Neurobiology, Free University of Berlin, 14195 Berlin, Germany
| |
Collapse
|
5
|
Miao H, Wei Y, Lee SG, Wu Z, Kaur J, Kim WJ. Glia-specific expression of neuropeptide receptor Lgr4 regulates development and adult physiology in Drosophila. J Neurosci Res 2024; 102:e25271. [PMID: 38284837 DOI: 10.1002/jnr.25271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 10/10/2023] [Accepted: 10/28/2023] [Indexed: 01/30/2024]
Abstract
Similar to the human brain, Drosophila glia may well be divided into several subtypes that each carries out specific functions. Glial GPCRs play key roles in crosstalk between neurons and glia. Drosophila Lgr4 (dLgr4) is a human relaxin receptor homolog involved in angiogenesis, cardiovascular regulation, collagen remodeling, and wound healing. A recent study suggests that ilp7 might be the ligand for Lgr4 and regulates escape behavior of Drosophila larvae. Here we demonstrate that Drosophila Lgr4 expression in glial cells, not neurons, is necessary for early development, adult behavior, and lifespan. Reducing the Lgr4 level in glial cells disrupts Drosophila development, while knocking down other LGR family members in glia has no impact. Adult-specific knockdown of Lgr4 in glia but not neurons reduce locomotion, male reproductive success, and animal longevity. The investigation of how glial expression of Lgr4 contributes to this behavioral alteration will increase our understanding of how insulin signaling via glia selectively modulates neuronal activity and behavior.
Collapse
Affiliation(s)
- Hongyu Miao
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
| | - Yanan Wei
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
| | - Seung Gee Lee
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Zekun Wu
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
| | - Jasdeep Kaur
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Woo Jae Kim
- The HIT Center for Life Sciences, Harbin Institute of Technology, Harbin, China
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
6
|
Ferreira AAG, Desplan C. An Atlas of the Developing Drosophila Visual System Glia and Subcellular mRNA Localization of Transcripts in Single Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.06.552169. [PMID: 37609218 PMCID: PMC10441313 DOI: 10.1101/2023.08.06.552169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Glial cells are essential for proper nervous system development and function. To understand glial development and function, we comprehensively annotated glial cells in a single-cell mRNA-sequencing (scRNAseq) atlas of the developing Drosophila visual system. This allowed us to study their developmental trajectories, from larval to adult stages, and to understand how specific types of glia diversify during development. For example, neuropil glia that are initially transcriptionally similar in larvae, split into ensheathing and astrocyte-like glia during pupal stages. Other glial types, such as chiasm glia change gradually during development without splitting into two cell types. The analysis of scRNA-seq allowed us to discover that the transcriptome of glial cell bodies can be distinguished from that of their broken processes. The processes contain distinct enriched mRNAs that were validated in vivo. Therefore, we have identified most glial types in the developing optic lobe and devised a computational approach to identify mRNA species that are localized to cell bodies or cellular processes.
Collapse
Affiliation(s)
| | - Claude Desplan
- Department of Biology, New York University, New York, NY, USA
| |
Collapse
|
7
|
Bostock MP, Prasad AR, Donoghue A, Fernandes VM. Photoreceptors generate neuronal diversity in their target field through a Hedgehog morphogen gradient in Drosophila. eLife 2022; 11:78093. [PMID: 36004721 PMCID: PMC9507128 DOI: 10.7554/elife.78093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Defining the origin of neuronal diversity is a major challenge in developmental neurobiology. The Drosophila visual system is an excellent paradigm to study how cellular diversity is generated. Photoreceptors from the eye disc grow their axons into the optic lobe and secrete Hedgehog (Hh) to induce the lamina, such that for every unit eye there is a corresponding lamina unit made up of post-mitotic precursors stacked into columns. Each differentiated column contains five lamina neuron types (L1-L5), making it the simplest neuropil in the optic lobe, yet how this diversity is generated was unknown. Here, we found that Hh pathway activity is graded along the distal-proximal axis of lamina columns and further determined that this gradient in pathway activity arises from a gradient of Hh ligand. We manipulated Hh pathway activity cell-autonomously in lamina precursors and non-cell autonomously by inactivating the Hh ligand, and by knocking it down in photoreceptors. These manipulations showed that different thresholds of activity specify unique cell identities, with more proximal cell types specified in response to progressively lower Hh levels. Thus, our data establish that Hh acts as a morphogen to pattern the lamina. Although, this is the first such report during Drosophila nervous system development, our work uncovers a remarkable similarity with the vertebrate neural tube, which is patterned by Sonic Hedgehog. Altogether, we show that differentiating neurons can regulate the neuronal diversity of their distant target fields through morphogen gradients.
Collapse
Affiliation(s)
- Matthew P Bostock
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Anadika R Prasad
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Alicia Donoghue
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Vilaiwan M Fernandes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
8
|
Konstantinides N, Holguera I, Rossi AM, Escobar A, Dudragne L, Chen YC, Tran TN, Martínez Jaimes AM, Özel MN, Simon F, Shao Z, Tsankova NM, Fullard JF, Walldorf U, Roussos P, Desplan C. A complete temporal transcription factor series in the fly visual system. Nature 2022; 604:316-322. [PMID: 35388222 PMCID: PMC9074256 DOI: 10.1038/s41586-022-04564-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 02/18/2022] [Indexed: 01/17/2023]
Abstract
The brain consists of thousands of neuronal types that are generated by stem cells producing different neuronal types as they age. In Drosophila, this temporal patterning is driven by the successive expression of temporal transcription factors (tTFs)1-6. Here we used single-cell mRNA sequencing to identify the complete series of tTFs that specify most Drosophila optic lobe neurons. We verify that tTFs regulate the progression of the series by activating the next tTF(s) and repressing the previous one(s), and also identify more complex mechanisms of regulation. Moreover, we establish the temporal window of origin and birth order of each neuronal type in the medulla and provide evidence that these tTFs are sufficient to explain the generation of all of the neuronal diversity in this brain region. Finally, we describe the first steps of neuronal differentiation and show that these steps are conserved in humans. We find that terminal differentiation genes, such as neurotransmitter-related genes, are present as transcripts, but not as proteins, in immature larval neurons. This comprehensive analysis of a temporal series of tTFs in the optic lobe offers mechanistic insights into how tTF series are regulated, and how they can lead to the generation of a complete set of neurons.
Collapse
Affiliation(s)
- Nikolaos Konstantinides
- Department of Biology, New York University, New York, NY, USA.
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France.
| | - Isabel Holguera
- Department of Biology, New York University, New York, NY, USA
| | - Anthony M Rossi
- Department of Biology, New York University, New York, NY, USA
- Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | | | | | - Yen-Chung Chen
- Department of Biology, New York University, New York, NY, USA
| | - Thinh N Tran
- Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | | | | | - Félix Simon
- Department of Biology, New York University, New York, NY, USA
| | - Zhiping Shao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Institute for Genomics and Multiscale Biology, New York, NY, USA
| | - Nadejda M Tsankova
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Institute for Genomics and Multiscale Biology, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Uwe Walldorf
- Developmental Biology, Saarland University, Homburg, Germany
| | - Panos Roussos
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, Institute for Genomics and Multiscale Biology, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, New York, NY, USA
| | - Claude Desplan
- Department of Biology, New York University, New York, NY, USA.
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
9
|
Zhu H, Zhao SD, Ray A, Zhang Y, Li X. A comprehensive temporal patterning gene network in Drosophila medulla neuroblasts revealed by single-cell RNA sequencing. Nat Commun 2022; 13:1247. [PMID: 35273186 PMCID: PMC8913700 DOI: 10.1038/s41467-022-28915-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 02/12/2022] [Indexed: 12/24/2022] Open
Abstract
During development, neural progenitors are temporally patterned to sequentially generate a variety of neural types. In Drosophila neural progenitors called neuroblasts, temporal patterning is regulated by cascades of Temporal Transcription Factors (TTFs). However, known TTFs were mostly identified through candidate approaches and may not be complete. In addition, many fundamental questions remain concerning the TTF cascade initiation, progression, and termination. In this work, we use single-cell RNA sequencing of Drosophila medulla neuroblasts of all ages to identify a list of previously unknown TTFs, and experimentally characterize their roles in temporal patterning and neuronal specification. Our study reveals a comprehensive temporal gene network that patterns medulla neuroblasts from start to end. Furthermore, the speed of the cascade progression is regulated by Lola transcription factors expressed in all medulla neuroblasts. Our comprehensive study of the medulla neuroblast temporal cascade illustrates mechanisms that may be conserved in the temporal patterning of neural progenitors. During development, neural progenitors generate a variety of neural types sequentially. Here the authors examine gene expression patterns in Drosophila neural progenitors at single-cell level, and identify a gene regulatory network controlling the sequential generation of different neural types.
Collapse
Affiliation(s)
- Hailun Zhu
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sihai Dave Zhao
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Alokananda Ray
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yu Zhang
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xin Li
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
10
|
Rapti G. Open Frontiers in Neural Cell Type Investigations; Lessons From Caenorhabditis elegans and Beyond, Toward a Multimodal Integration. Front Neurosci 2022; 15:787753. [PMID: 35321480 PMCID: PMC8934944 DOI: 10.3389/fnins.2021.787753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Nervous system cells, the building blocks of circuits, have been studied with ever-progressing resolution, yet neural circuits appear still resistant to schemes of reductionist classification. Due to their sheer numbers, complexity and diversity, their systematic study requires concrete classifications that can serve reduced dimensionality, reproducibility, and information integration. Conventional hierarchical schemes transformed through the history of neuroscience by prioritizing criteria of morphology, (electro)physiological activity, molecular content, and circuit function, influenced by prevailing methodologies of the time. Since the molecular biology revolution and the recent advents in transcriptomics, molecular profiling gains ground toward the classification of neurons and glial cell types. Yet, transcriptomics entails technical challenges and more importantly uncovers unforeseen spatiotemporal heterogeneity, in complex and simpler nervous systems. Cells change states dynamically in space and time, in response to stimuli or throughout their developmental trajectory. Mapping cell type and state heterogeneity uncovers uncharted terrains in neurons and especially in glial cell biology, that remains understudied in many aspects. Examining neurons and glial cells from the perspectives of molecular neuroscience, physiology, development and evolution highlights the advantage of multifaceted classification schemes. Among the amalgam of models contributing to neuroscience research, Caenorhabditis elegans combines nervous system anatomy, lineage, connectivity and molecular content, all mapped at single-cell resolution, and can provide valuable insights for the workflow and challenges of the multimodal integration of cell type features. This review reflects on concepts and practices of neuron and glial cells classification and how research, in C. elegans and beyond, guides nervous system experimentation through integrated multidimensional schemes. It highlights underlying principles, emerging themes, and open frontiers in the study of nervous system development, regulatory logic and evolution. It proposes unified platforms to allow integrated annotation of large-scale datasets, gene-function studies, published or unpublished findings and community feedback. Neuroscience is moving fast toward interdisciplinary, high-throughput approaches for combined mapping of the morphology, physiology, connectivity, molecular function, and the integration of information in multifaceted schemes. A closer look in mapped neural circuits and understudied terrains offers insights for the best implementation of these approaches.
Collapse
|
11
|
McLaughlin CN, Brbić M, Xie Q, Li T, Horns F, Kolluru SS, Kebschull JM, Vacek D, Xie A, Li J, Jones RC, Leskovec J, Quake SR, Luo L, Li H. Single-cell transcriptomes of developing and adult olfactory receptor neurons in Drosophila. eLife 2021; 10:e63856. [PMID: 33555999 PMCID: PMC7870146 DOI: 10.7554/elife.63856] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Recognition of environmental cues is essential for the survival of all organisms. Transcriptional changes occur to enable the generation and function of the neural circuits underlying sensory perception. To gain insight into these changes, we generated single-cell transcriptomes of Drosophila olfactory- (ORNs), thermo-, and hygro-sensory neurons at an early developmental and adult stage using single-cell and single-nucleus RNA sequencing. We discovered that ORNs maintain expression of the same olfactory receptors across development. Using receptor expression and computational approaches, we matched transcriptomic clusters corresponding to anatomically and physiologically defined neuron types across multiple developmental stages. We found that cell-type-specific transcriptomes partly reflected axon trajectory choices in development and sensory modality in adults. We uncovered stage-specific genes that could regulate the wiring and sensory responses of distinct ORN types. Collectively, our data reveal transcriptomic features of sensory neuron biology and provide a resource for future studies of their development and physiology.
Collapse
Affiliation(s)
- Colleen N McLaughlin
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Maria Brbić
- Department of Computer Science, Stanford UniversityStanfordUnited States
| | - Qijing Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
- Neurosciences Graduate Program, Stanford UniversityStanfordUnited States
| | - Tongchao Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Felix Horns
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Biophysics Graduate Program, Stanford UniversityStanfordUnited States
| | - Sai Saroja Kolluru
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubStanfordUnited States
| | - Justus M Kebschull
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - David Vacek
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Anthony Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Jiefu Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
- Biology Graduate Program, Stanford UniversityStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Jure Leskovec
- Department of Computer Science, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubStanfordUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Hongjie Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
12
|
Lago-Baldaia I, Fernandes VM, Ackerman SD. More Than Mortar: Glia as Architects of Nervous System Development and Disease. Front Cell Dev Biol 2020; 8:611269. [PMID: 33381506 PMCID: PMC7767919 DOI: 10.3389/fcell.2020.611269] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Glial cells are an essential component of the nervous system of vertebrates and invertebrates. In the human brain, glia are as numerous as neurons, yet the importance of glia to nearly every aspect of nervous system development has only been expounded over the last several decades. Glia are now known to regulate neural specification, synaptogenesis, synapse function, and even broad circuit function. Given their ubiquity, it is not surprising that the contribution of glia to neuronal disease pathogenesis is a growing area of research. In this review, we will summarize the accumulated evidence of glial participation in several distinct phases of nervous system development and organization-neural specification, circuit wiring, and circuit function. Finally, we will highlight how these early developmental roles of glia contribute to nervous system dysfunction in neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Inês Lago-Baldaia
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Vilaiwan M. Fernandes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Sarah D. Ackerman
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR, United States
| |
Collapse
|
13
|
Junkunlo K, Söderhäll K, Söderhäll I. A transcription factor glial cell missing (Gcm) in the freshwater crayfish Pacifastacus leniusculus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 113:103782. [PMID: 32679114 DOI: 10.1016/j.dci.2020.103782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 06/11/2023]
Abstract
The transcription factor glial cell missing, Gcm, is known to be an important protein in the determination of glial cell fate as well as embryonic plasmatocyte differentiation in Drosophila melanogaster. So far, no function for Gcm in crustaceans has been reported. In this study, we show the cDNA sequence of a Gcm homologue in the freshwater crayfish Pacifastacus leniusculus. The P. leniusculus Gcm transcript is expressed exclusively in brain and nervous tissue, and by in situ hybridization we show that the expression is restricted to a small number of large cells with morphology similar to neurosecretory cells. Furthermore, we show that the expression of Gcm coincides with the expression of a Repo homologue, that is induced in expression by Gcm in Drosophila. Moreover, the Gcm transcript is increased shortly and transiently after injection of cystamine, a substance that inhibits transglutaminase and also strongly affects the movement behavior of crayfish. This finding of Gcm transcripts in a subpopulation of brain cells in very low numbers may enable more detailed studies about Gcm in adult crustaceans.
Collapse
Affiliation(s)
- Kingkamon Junkunlo
- Department of Comparative Physiology, Uppsala University, Norbyvägen 18 A, SE752 36, Uppsala, Sweden
| | - Kenneth Söderhäll
- Science for Life Laboratory, Department of Comparative Physiology, Uppsala University, Norbyvägen 18A, 752 36, Uppsala, Sweden
| | - Irene Söderhäll
- Science for Life Laboratory, Department of Comparative Physiology, Uppsala University, Norbyvägen 18A, 752 36, Uppsala, Sweden.
| |
Collapse
|
14
|
Kato K, Orihara-Ono M, Awasaki T. Multiple lineages enable robust development of the neuropil-glia architecture in adult Drosophila. Development 2020; 147:dev184085. [PMID: 32051172 DOI: 10.1242/dev.184085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022]
Abstract
Neural remodeling is essential for the development of a functional nervous system and has been extensively studied in the metamorphosis of Drosophila Despite the crucial roles of glial cells in brain functions, including learning and behavior, little is known of how adult glial cells develop in the context of neural remodeling. Here, we show that the architecture of neuropil-glia in the adult Drosophila brain, which is composed of astrocyte-like glia (ALG) and ensheathing glia (EG), robustly develops from two different populations in the larva: the larval EG and glial cell missing-positive (gcm+ ) cells. Whereas gcm+ cells proliferate and generate adult ALG and EG, larval EG dedifferentiate, proliferate and redifferentiate into the same glial subtypes. Each glial lineage occupies a certain brain area complementary to the other, and together they form the adult neuropil-glia architecture. Both lineages require the FGF receptor Heartless to proliferate, and the homeoprotein Prospero to differentiate into ALG. Lineage-specific inhibition of gliogenesis revealed that each lineage compensates for deficiency in the proliferation of the other. Together, the lineages ensure the robust development of adult neuropil-glia, thereby ensuring a functional brain.
Collapse
Affiliation(s)
- Kentaro Kato
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| | - Minako Orihara-Ono
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| | - Takeshi Awasaki
- Department of Biology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka 181-8611, Tokyo, Japan
| |
Collapse
|
15
|
Li H, Li T, Horns F, Li J, Xie Q, Xu C, Wu B, Kebschull JM, McLaughlin CN, Kolluru SS, Jones RC, Vacek D, Xie A, Luginbuhl DJ, Quake SR, Luo L. Single-Cell Transcriptomes Reveal Diverse Regulatory Strategies for Olfactory Receptor Expression and Axon Targeting. Curr Biol 2020; 30:1189-1198.e5. [PMID: 32059767 DOI: 10.1016/j.cub.2020.01.049] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/20/2022]
Abstract
The regulatory mechanisms by which neurons coordinate their physiology and connectivity are not well understood. The Drosophila olfactory receptor neurons (ORNs) provide an excellent system to investigate this question. Each ORN type expresses a unique olfactory receptor, or a combination thereof, and sends their axons to a stereotyped glomerulus. Using single-cell RNA sequencing, we identified 33 transcriptomic clusters for ORNs and mapped 20 to their glomerular types, demonstrating that transcriptomic clusters correspond well with anatomically and physiologically defined ORN types. Each ORN type expresses hundreds of transcription factors. Transcriptome-instructed genetic analyses revealed that (1) one broadly expressed transcription factor (Acj6) only regulates olfactory receptor expression in one ORN type and only wiring specificity in another type, (2) one type-restricted transcription factor (Forkhead) only regulates receptor expression, and (3) another type-restricted transcription factor (Unplugged) regulates both events. Thus, ORNs utilize diverse strategies and complex regulatory networks to coordinate their physiology and connectivity.
Collapse
Affiliation(s)
- Hongjie Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| | - Tongchao Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Felix Horns
- Biophysics Graduate Program, Stanford University, Stanford, CA 94305, USA; Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Jiefu Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Qijing Xie
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Bing Wu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Justus M Kebschull
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Colleen N McLaughlin
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Sai Saroja Kolluru
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Robert C Jones
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - David Vacek
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Anthony Xie
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David J Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Stephen R Quake
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, Stanford, CA 94305, USA.
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
16
|
Li J, Han S, Li H, Udeshi ND, Svinkina T, Mani DR, Xu C, Guajardo R, Xie Q, Li T, Luginbuhl DJ, Wu B, McLaughlin CN, Xie A, Kaewsapsak P, Quake SR, Carr SA, Ting AY, Luo L. Cell-Surface Proteomic Profiling in the Fly Brain Uncovers Wiring Regulators. Cell 2020; 180:373-386.e15. [PMID: 31955847 DOI: 10.1016/j.cell.2019.12.029] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/12/2023]
Abstract
Molecular interactions at the cellular interface mediate organized assembly of single cells into tissues and, thus, govern the development and physiology of multicellular organisms. Here, we developed a cell-type-specific, spatiotemporally resolved approach to profile cell-surface proteomes in intact tissues. Quantitative profiling of cell-surface proteomes of Drosophila olfactory projection neurons (PNs) in pupae and adults revealed global downregulation of wiring molecules and upregulation of synaptic molecules in the transition from developing to mature PNs. A proteome-instructed in vivo screen identified 20 cell-surface molecules regulating neural circuit assembly, many of which belong to evolutionarily conserved protein families not previously linked to neural development. Genetic analysis further revealed that the lipoprotein receptor LRP1 cell-autonomously controls PN dendrite targeting, contributing to the formation of a precise olfactory map. These findings highlight the power of temporally resolved in situ cell-surface proteomic profiling in discovering regulators of brain wiring.
Collapse
Affiliation(s)
- Jiefu Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| | - Shuo Han
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Hongjie Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Namrata D Udeshi
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tanya Svinkina
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - D R Mani
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chuanyun Xu
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Guajardo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Qijing Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Tongchao Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David J Luginbuhl
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Bing Wu
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Colleen N McLaughlin
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Anthony Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Pornchai Kaewsapsak
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Stephen R Quake
- Departments of Bioengineering and Applied Physics, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Steven A Carr
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alice Y Ting
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA.
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
17
|
Shimozono M, Osaka J, Kato Y, Araki T, Kawamura H, Takechi H, Hakeda-Suzuki S, Suzuki T. Cell surface molecule, Klingon, mediates the refinement of synaptic specificity in the Drosophila visual system. Genes Cells 2019; 24:496-510. [PMID: 31124270 DOI: 10.1111/gtc.12703] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 05/04/2019] [Accepted: 05/19/2019] [Indexed: 11/29/2022]
Abstract
In the Drosophila brain, neurons form genetically specified synaptic connections with defined neuronal targets. It is proposed that each central nervous system neuron expresses specific cell surface proteins, which act as identification tags. Through an RNAi screen of cell surface molecules in the Drosophila visual system, we found that the cell adhesion molecule Klingon (Klg) plays an important role in repressing the ectopic formation of extended axons, preventing the formation of excessive synapses. Cell-specific manipulation of klg showed that Klg is required in both photoreceptors and the glia, suggesting that the balanced homophilic interaction between photoreceptor axons and the glia is required for normal synapse formation. Previous studies suggested that Klg binds to cDIP and our genetic analyses indicate that cDIP is required in glia for ectopic synaptic repression. These data suggest that Klg play a critical role together with cDIP in refining synaptic specificity and preventing unnecessary connections in the brain.
Collapse
Affiliation(s)
- Mai Shimozono
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Jiro Osaka
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Yuya Kato
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Tomohiro Araki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Hinata Kawamura
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Hiroki Takechi
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Satoko Hakeda-Suzuki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| | - Takashi Suzuki
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokahama, Kanagawa, Japan
| |
Collapse
|
18
|
Temporal and spatial order of photoreceptor and glia projections into optic lobe in Drosophila. Sci Rep 2018; 8:12669. [PMID: 30140062 PMCID: PMC6107658 DOI: 10.1038/s41598-018-30415-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/25/2018] [Indexed: 11/10/2022] Open
Abstract
Photoreceptor (PR) axons project from the retina to the optic lobe in brain and form a precise retinotopic map in the Drosophila visual system. Yet the role of retinal basal glia in the retinotopic map formation is not previously known. We examined the formation of the retinotopic map by marking single PR pairs and following their axonal projections. In addition to confirming previous studies that the spatial information is preserved from the retina to the optic stalk and then to the optic lamina, we found that the young PR R3/4 axons transiently overshoot and then retract to their final destination, the lamina plexus. We then examined the process of wrapping glia (WG) membrane extension in the eye disc and showed that the WG membrane extensions also follow the retinotopic map. We show that the WG is important for the proper spatial distribution of PR axons in the optic stalk and lamina, suggesting an active role of wrapping glia in the retinotopic map formation.
Collapse
|
19
|
Li J, Guajardo R, Xu C, Wu B, Li H, Li T, Luginbuhl DJ, Xie X, Luo L. Stepwise wiring of the Drosophila olfactory map requires specific Plexin B levels. eLife 2018; 7:39088. [PMID: 30136927 PMCID: PMC6118820 DOI: 10.7554/elife.39088] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 08/22/2018] [Indexed: 01/13/2023] Open
Abstract
The precise assembly of a neural circuit involves many consecutive steps. The conflict between a limited number of wiring molecules and the complexity of the neural network impels each molecule to execute multiple functions at different steps. Here, we examined the cell-type specific distribution of endogenous levels of axon guidance receptor Plexin B (PlexB) in the developing antennal lobe, the first olfactory processing center in Drosophila. We found that different classes of olfactory receptor neurons (ORNs) express PlexB at different levels in two wiring steps – axonal trajectory choice and subsequent target selection. In line with its temporally distinct patterns, the proper levels of PlexB control both steps in succession. Genetic interactions further revealed that the effect of high-level PlexB is antagonized by its canonical partner Sema2b. Thus, PlexB plays a multifaceted role in instructing the assembly of the Drosophila olfactory circuit through temporally-regulated expression patterns and expression level-dependent effects.
Collapse
Affiliation(s)
- Jiefu Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Ricardo Guajardo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Chuanyun Xu
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Bing Wu
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Hongjie Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Tongchao Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - David J Luginbuhl
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Xiaojun Xie
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
20
|
Bazzi W, Cattenoz PB, Delaporte C, Dasari V, Sakr R, Yuasa Y, Giangrande A. Embryonic hematopoiesis modulates the inflammatory response and larval hematopoiesis in Drosophila. eLife 2018; 7:e34890. [PMID: 29992900 PMCID: PMC6040882 DOI: 10.7554/elife.34890] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/18/2018] [Indexed: 11/25/2022] Open
Abstract
Recent lineage tracing analyses have significantly improved our understanding of immune system development and highlighted the importance of the different hematopoietic waves. The current challenge is to understand whether these waves interact and whether this affects the function of the immune system. Here we report a molecular pathway regulating the immune response and involving the communication between embryonic and larval hematopoietic waves in Drosophila. Down-regulating the transcription factor Gcm specific to embryonic hematopoiesis enhances the larval phenotypes induced by over-expressing the pro-inflammatory Jak/Stat pathway or by wasp infestation. Gcm works by modulating the transduction of the Upd cytokines to the site of larval hematopoiesis and hence the response to chronic (Jak/Stat over-expression) and acute (wasp infestation) immune challenges. Thus, homeostatic interactions control the function of the immune system in physiology and pathology. Our data also indicate that a transiently expressed developmental pathway has a long-lasting effect on the immune response.
Collapse
Affiliation(s)
- Wael Bazzi
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Pierre B Cattenoz
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Claude Delaporte
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Vasanthi Dasari
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Rosy Sakr
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Yoshihiro Yuasa
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| | - Angela Giangrande
- Institut de Génétique et de Biologie Moléculaire et CellulaireIllkirchFrance
- UMR7104Centre National de la Recherche ScientifiqueIllkirchFrance
- U1258Institut National de la Santé et de la Recherche MédicaleIllkirchFrance
- Université de StrasbourgIllkirchFrance
| |
Collapse
|
21
|
Bao R, Dia SE, Issa HA, Alhusein D, Friedrich M. Comparative Evidence of an Exceptional Impact of Gene Duplication on the Developmental Evolution of Drosophila and the Higher Diptera. Front Ecol Evol 2018. [DOI: 10.3389/fevo.2018.00063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
22
|
Liou NF, Lin SH, Chen YJ, Tsai KT, Yang CJ, Lin TY, Wu TH, Lin HJ, Chen YT, Gohl DM, Silies M, Chou YH. Diverse populations of local interneurons integrate into the Drosophila adult olfactory circuit. Nat Commun 2018; 9:2232. [PMID: 29884811 PMCID: PMC5993751 DOI: 10.1038/s41467-018-04675-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/14/2018] [Indexed: 11/09/2022] Open
Abstract
Drosophila olfactory local interneurons (LNs) in the antennal lobe are highly diverse and variable. How and when distinct types of LNs emerge, differentiate, and integrate into the olfactory circuit is unknown. Through systematic developmental analyses, we found that LNs are recruited to the adult olfactory circuit in three groups. Group 1 LNs are residual larval LNs. Group 2 are adult-specific LNs that emerge before cognate sensory and projection neurons establish synaptic specificity, and Group 3 LNs emerge after synaptic specificity is established. Group 1 larval LNs are selectively reintegrated into the adult circuit through pruning and re-extension of processes to distinct regions of the antennal lobe, while others die during metamorphosis. Precise temporal control of this pruning and cell death shapes the global organization of the adult antennal lobe. Our findings provide a road map to understand how LNs develop and contribute to constructing the olfactory circuit. Local interneurons (LNs) in the Drosophila olfactory system are highly diverse. Here, the authors labeled different LN types and described how different LN subtypes are integrated into the developing circuit.
Collapse
Affiliation(s)
- Nan-Fu Liou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Shih-Han Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Ying-Jun Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Kuo-Ting Tsai
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Chi-Jen Yang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Tzi-Yang Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan.,Research Institute of Molecular Pathology (IMP), Vienna Biocenter, Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Ting-Han Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Hsin-Ju Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Yuh-Tarng Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Daryl M Gohl
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, USA.,University of Minnesota Genomics Center, 1-210 CCRB, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - Marion Silies
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, USA.,European Neuroscience Institute, University Medical Center Göttingen, Grisebachstr. 5, 37077, Göttingen, Germany
| | - Ya-Hui Chou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan. .,Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
23
|
Jin EJ, Kiral FR, Ozel MN, Burchardt LS, Osterland M, Epstein D, Wolfenberg H, Prohaska S, Hiesinger PR. Live Observation of Two Parallel Membrane Degradation Pathways at Axon Terminals. Curr Biol 2018; 28:1027-1038.e4. [PMID: 29551411 PMCID: PMC5944365 DOI: 10.1016/j.cub.2018.02.032] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 01/24/2018] [Accepted: 02/14/2018] [Indexed: 01/04/2023]
Abstract
Neurons are highly polarized cells that require continuous turnover of membrane proteins at axon terminals to develop, function, and survive. Yet, it is still unclear whether membrane protein degradation requires transport back to the cell body or whether degradation also occurs locally at the axon terminal, where live observation of sorting and degradation has remained a challenge. Here, we report direct observation of two cargo-specific membrane protein degradation mechanisms at axon terminals based on a live-imaging approach in intact Drosophila brains. We show that different acidification-sensing cargo probes are sorted into distinct classes of degradative “hub” compartments for synaptic vesicle proteins and plasma membrane proteins at axon terminals. Sorting and degradation of the two cargoes in the separate hubs are molecularly distinct. Local sorting of synaptic vesicle proteins for degradation at the axon terminal is, surprisingly, Rab7 independent, whereas sorting of plasma membrane proteins is Rab7 dependent. The cathepsin-like protease CP1 is specific to synaptic vesicle hubs, and its delivery requires the vesicle SNARE neuronal synaptobrevin. Cargo separation only occurs at the axon terminal, whereas degradative compartments at the cell body are mixed. These data show that at least two local, molecularly distinct pathways sort membrane cargo for degradation specifically at the axon terminal, whereas degradation can occur both at the terminal and en route to the cell body.
Collapse
Affiliation(s)
- Eugene Jennifer Jin
- Division of Neurobiology, Freie Universität Berlin, Königin Luise Straße 1-3, 14195 Berlin, Germany; Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ferdi Ridvan Kiral
- Division of Neurobiology, Freie Universität Berlin, Königin Luise Straße 1-3, 14195 Berlin, Germany
| | - Mehmet Neset Ozel
- Division of Neurobiology, Freie Universität Berlin, Königin Luise Straße 1-3, 14195 Berlin, Germany; Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lara Sophie Burchardt
- Division of Neurobiology, Freie Universität Berlin, Königin Luise Straße 1-3, 14195 Berlin, Germany
| | - Marc Osterland
- Zuse Institute Berlin, Takustraße 7, 14195 Berlin, Germany
| | - Daniel Epstein
- Division of Neurobiology, Freie Universität Berlin, Königin Luise Straße 1-3, 14195 Berlin, Germany
| | - Heike Wolfenberg
- Division of Neurobiology, Freie Universität Berlin, Königin Luise Straße 1-3, 14195 Berlin, Germany
| | | | - Peter Robin Hiesinger
- Division of Neurobiology, Freie Universität Berlin, Königin Luise Straße 1-3, 14195 Berlin, Germany.
| |
Collapse
|
24
|
Plazaola-Sasieta H, Fernández-Pineda A, Zhu Q, Morey M. Untangling the wiring of the Drosophila visual system: developmental principles and molecular strategies. J Neurogenet 2017; 31:231-249. [DOI: 10.1080/01677063.2017.1391249] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Haritz Plazaola-Sasieta
- Department of Genetics, Microbiology and Statistics; School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Alejandra Fernández-Pineda
- Department of Genetics, Microbiology and Statistics; School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Qi Zhu
- Department of Genetics, Microbiology and Statistics; School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Marta Morey
- Department of Genetics, Microbiology and Statistics; School of Biology and Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| |
Collapse
|
25
|
Apitz H, Salecker I. Retinal determination genes coordinate neuroepithelial specification and neurogenesis modes in the Drosophila optic lobe. Development 2017; 143:2431-42. [PMID: 27381228 PMCID: PMC4958324 DOI: 10.1242/dev.135004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 05/18/2016] [Indexed: 12/15/2022]
Abstract
Differences in neuroepithelial patterning and neurogenesis modes contribute to area-specific diversifications of neural circuits. In the Drosophila visual system, two neuroepithelia, the outer (OPC) and inner (IPC) proliferation centers, generate neuron subtypes for four ganglia in several ways. Whereas neuroepithelial cells in the medial OPC directly convert into neuroblasts, in an IPC subdomain they generate migratory progenitors by epithelial-mesenchymal transition that mature into neuroblasts in a second proliferative zone. The molecular mechanisms that regulate the identity of these neuroepithelia, including their neurogenesis modes, remain poorly understood. Analysis of Polycomblike revealed that loss of Polycomb group-mediated repression of the Hox gene Abdominal-B (Abd-B) caused the transformation of OPC to IPC neuroepithelial identity. This suggests that the neuroepithelial default state is IPC-like, whereas OPC identity is derived. Ectopic Abd-B blocks expression of the highly conserved retinal determination gene network members Eyes absent (Eya), Sine oculis (So) and Homothorax (Hth). These factors are essential for OPC specification and neurogenesis control. Finally, eya and so are also sufficient to confer OPC-like identity, and, in parallel with hth, the OPC-specific neurogenesis mode on the IPC. Summary: Polycomb-mediated repression of the Abd-B Hox gene controls expression of retinal determination genes and hence identity of the Drosophila optic lobe neuroepithelia.
Collapse
Affiliation(s)
- Holger Apitz
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK
| | - Iris Salecker
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK
| |
Collapse
|
26
|
Mosca TJ, Luginbuhl DJ, Wang IE, Luo L. Presynaptic LRP4 promotes synapse number and function of excitatory CNS neurons. eLife 2017; 6. [PMID: 28606304 PMCID: PMC5469616 DOI: 10.7554/elife.27347] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/08/2017] [Indexed: 12/24/2022] Open
Abstract
Precise coordination of synaptic connections ensures proper information flow within circuits. The activity of presynaptic organizing molecules signaling to downstream pathways is essential for such coordination, though such entities remain incompletely known. We show that LRP4, a conserved transmembrane protein known for its postsynaptic roles, functions presynaptically as an organizing molecule. In the Drosophila brain, LRP4 localizes to the nerve terminals at or near active zones. Loss of presynaptic LRP4 reduces excitatory (not inhibitory) synapse number, impairs active zone architecture, and abolishes olfactory attraction - the latter of which can be suppressed by reducing presynaptic GABAB receptors. LRP4 overexpression increases synapse number in excitatory and inhibitory neurons, suggesting an instructive role and a common downstream synapse addition pathway. Mechanistically, LRP4 functions via the conserved kinase SRPK79D to ensure normal synapse number and behavior. This highlights a presynaptic function for LRP4, enabling deeper understanding of how synapse organization is coordinated.
Collapse
Affiliation(s)
- Timothy J Mosca
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, United States.,Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - David J Luginbuhl
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| | - Irving E Wang
- Department of Neurobiology, Stanford University, Stanford, United States
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, United States
| |
Collapse
|
27
|
MRL proteins cooperate with activated Ras in glia to drive distinct oncogenic outcomes. Oncogene 2017; 36:4311-4322. [PMID: 28346426 PMCID: PMC5537612 DOI: 10.1038/onc.2017.68] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 01/16/2017] [Accepted: 02/10/2017] [Indexed: 12/13/2022]
Abstract
The Mig10/RIAM/Lpd (MRL) adapter protein Lpd regulates actin dynamics through interactions with Scar/WAVE and Ena/VASP proteins to promote the formation of cellular protrusions and to stimulate invasive migration. However, the ability of MRL proteins to interact with multiple actin regulators and to promote serum response factor (SRF) signalling has raised the question of whether MRL proteins employ alternative downstream mechanisms to drive oncogenic processes in a context-dependent manner. Here, using a Drosophila model, we show that overexpression of either human Lpd or its Drosophila orthologue Pico can promote growth and invasion of RasV12-induced cell tumours in the brain. Notably, effects were restricted to two populations of Repo-positive glial cells: an invasive population, characterized by JNK-dependent elevation of Mmp1 expression, and a hyperproliferative population lacking elevated JNK signalling. JNK activation was not triggered by reactive immune cell signalling, implicating the involvement of an intrinsic stress response. The ability to promote dissemination of RasV12-induced tumours was shared by a subset of actin regulators, including, most prominently, Chicadee/Profilin, which directly interacts with Pico, and, Mal, a cofactor for serum response factor that responds to changes in G:F actin dynamics. Suppression of Mal activity partially abrogated the ability of pico to promote invasion of RasV12 tumours. Furthermore, we found that larval glia are enriched for serum response factor expression, explaining the apparent sensitivity of glial cells to Pico/RasV12 overexpression. Taken together, our findings indicate that MRL proteins cooperate with oncogenic Ras to promote formation of glial tumours, and that, in this context, Mal/serum response factor activation is rate-limiting for tumour dissemination.
Collapse
|
28
|
Transcriptome Profiling Identifies Multiplexin as a Target of SAGA Deubiquitinase Activity in Glia Required for Precise Axon Guidance During Drosophila Visual Development. G3-GENES GENOMES GENETICS 2016; 6:2435-45. [PMID: 27261002 PMCID: PMC4978897 DOI: 10.1534/g3.116.031310] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Spt-Ada-Gcn5 Acetyltransferase (SAGA) complex is a transcriptional coactivator with histone acetylase and deubiquitinase activities that plays an important role in visual development and function. In Drosophila melanogaster, four SAGA subunits are required for the deubiquitination of monoubiquitinated histone H2B (ubH2B): Nonstop, Sgf11, E(y)2, and Ataxin 7. Mutations that disrupt SAGA deubiquitinase activity cause defects in neuronal connectivity in the developing Drosophila visual system. In addition, mutations in SAGA result in the human progressive visual disorder spinocerebellar ataxia type 7 (SCA7). Glial cells play a crucial role in both the neuronal connectivity defect in nonstop and sgf11 flies, and in the retinal degeneration observed in SCA7 patients. Thus, we sought to identify the gene targets of SAGA deubiquitinase activity in glia in the Drosophila larval central nervous system. To do this, we enriched glia from wild-type, nonstop, and sgf11 larval optic lobes using affinity-purification of KASH-GFP tagged nuclei, and then examined each transcriptome using RNA-seq. Our analysis showed that SAGA deubiquitinase activity is required for proper expression of 16% of actively transcribed genes in glia, especially genes involved in proteasome function, protein folding and axon guidance. We further show that the SAGA deubiquitinase-activated gene Multiplexin (Mp) is required in glia for proper photoreceptor axon targeting. Mutations in the human ortholog of Mp, COL18A1, have been identified in a family with a SCA7-like progressive visual disorder, suggesting that defects in the expression of this gene in SCA7 patients could play a role in the retinal degeneration that is unique to this ataxia.
Collapse
|
29
|
Cattenoz PB, Giangrande A. Revisiting the role of the Gcm transcription factor, from master regulator to Swiss army knife. Fly (Austin) 2016; 10:210-8. [PMID: 27434165 DOI: 10.1080/19336934.2016.1212793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Master genes are known to induce the differentiation of a multipotent cell into a specific cell type. These molecules are often transcription factors that switch on the regulatory cascade that triggers cell specification. Gcm was first described as the master gene of the glial fate in Drosophila as it induces the differentiation of neuroblasts into glia in the developing nervous system. Later on, Gcm was also shown to regulate the differentiation of blood, tendon and peritracheal cells as well as that of neuronal subsets. Thus, the glial master gene is used in at least 4 additional systems to promote differentiation. To understand the numerous roles of Gcm, we recently reported a genome-wide screen of Gcm direct targets in the Drosophila embryo. This screen provided new insight into the role and mode of action of this powerful transcription factor, notably on the interactions between Gcm and major differentiation pathways such as the Hedgehog, Notch and JAK/STAT. Here, we discuss the mode of action of Gcm in the different systems, we present new tissues that require Gcm and we revise the concept of 'master gene'.
Collapse
Affiliation(s)
- Pierre B Cattenoz
- a Department of Functional Genomics and Cancer , Institut de Génétique et de Biologie Moléculaire et Cellulaire , Illkirch , France
| | - Angela Giangrande
- a Department of Functional Genomics and Cancer , Institut de Génétique et de Biologie Moléculaire et Cellulaire , Illkirch , France
| |
Collapse
|
30
|
A Unique Class of Neural Progenitors in the Drosophila Optic Lobe Generates Both Migrating Neurons and Glia. Cell Rep 2016; 15:774-786. [PMID: 27149843 PMCID: PMC5154769 DOI: 10.1016/j.celrep.2016.03.061] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/25/2016] [Accepted: 03/15/2016] [Indexed: 02/04/2023] Open
Abstract
How neuronal and glial fates are specified from neural precursor cells is an important question for developmental neurobiologists. We address this question in the Drosophila optic lobe, composed of the lamina, medulla, and lobula complex. We show that two gliogenic regions posterior to the prospective lamina also produce lamina wide-field (Lawf) neurons, which share common progenitors with lamina glia. These progenitors express neither canonical neuroblast nor lamina precursor cell markers. They bifurcate into two sub-lineages in response to Notch signaling, generating lamina glia or Lawf neurons, respectively. The newly born glia and Lawfs then migrate tangentially over substantial distances to reach their target tissue. Thus, Lawf neurogenesis, which includes a common origin with glia, as well as neuronal migration, resembles several aspects of vertebrate neurogenesis.
Collapse
|
31
|
Suzuki T, Hasegawa E, Nakai Y, Kaido M, Takayama R, Sato M. Formation of Neuronal Circuits by Interactions between Neuronal Populations Derived from Different Origins in the Drosophila Visual Center. Cell Rep 2016; 15:499-509. [PMID: 27068458 DOI: 10.1016/j.celrep.2016.03.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/14/2016] [Accepted: 03/15/2016] [Indexed: 11/28/2022] Open
Abstract
A wide variety of neurons, including populations derived from different origins, are precisely arranged and correctly connected with their partner to establish a functional neural circuit during brain development. The molecular mechanisms that orchestrate the production and arrangement of these neurons have been obscure. Here, we demonstrate that cell-cell interactions play an important role in establishing the arrangement of neurons of different origins in the Drosophila visual center. Specific types of neurons born outside the medulla primordium migrate tangentially into the developing medulla cortex. During their tangential migration, these neurons express the repellent ligand Slit, and the two layers that the neurons intercalate between express the receptors Robo2 and Robo3. Genetic analysis suggests that Slit-Robo signaling may control the positioning of the layer cells or their processes to form a path for migration. Our results suggest that conserved axon guidance signaling is involved in the interactions between neurons of different origins during brain development.
Collapse
Affiliation(s)
- Takumi Suzuki
- Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| | - Eri Hasegawa
- Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| | - Yasuhiro Nakai
- Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan; CREST, JST, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Masako Kaido
- Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| | - Rie Takayama
- Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan; CREST, JST, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Makoto Sato
- Brain/Liver Interface Medicine Research Center, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan; Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan; Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8640, Japan; CREST, JST, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
32
|
Functional Conservation of the Glide/Gcm Regulatory Network Controlling Glia, Hemocyte, and Tendon Cell Differentiation in Drosophila. Genetics 2015; 202:191-219. [PMID: 26567182 PMCID: PMC4701085 DOI: 10.1534/genetics.115.182154] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 11/03/2015] [Indexed: 12/21/2022] Open
Abstract
High-throughput screens allow us to understand how transcription factors trigger developmental processes, including cell specification. A major challenge is identification of their binding sites because feedback loops and homeostatic interactions may mask the direct impact of those factors in transcriptome analyses. Moreover, this approach dissects the downstream signaling cascades and facilitates identification of conserved transcriptional programs. Here we show the results and the validation of a DNA adenine methyltransferase identification (DamID) genome-wide screen that identifies the direct targets of Glide/Gcm, a potent transcription factor that controls glia, hemocyte, and tendon cell differentiation in Drosophila. The screen identifies many genes that had not been previously associated with Glide/Gcm and highlights three major signaling pathways interacting with Glide/Gcm: Notch, Hedgehog, and JAK/STAT, which all involve feedback loops. Furthermore, the screen identifies effector molecules that are necessary for cell-cell interactions during late developmental processes and/or in ontogeny. Typically, immunoglobulin (Ig) domain-containing proteins control cell adhesion and axonal navigation. This shows that early and transiently expressed fate determinants not only control other transcription factors that, in turn, implement a specific developmental program but also directly affect late developmental events and cell function. Finally, while the mammalian genome contains two orthologous Gcm genes, their function has been demonstrated in vertebrate-specific tissues, placenta, and parathyroid glands, begging questions on the evolutionary conservation of the Gcm cascade in higher organisms. Here we provide the first evidence for the conservation of Gcm direct targets in humans. In sum, this work uncovers novel aspects of cell specification and sets the basis for further understanding of the role of conserved Gcm gene regulatory cascades.
Collapse
|
33
|
Altenhein B, Cattenoz PB, Giangrande A. The early life of a fly glial cell. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015. [DOI: 10.1002/wdev.200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | | | - Angela Giangrande
- Department of Functional Genomics and Cancer; IGBMC; Illkirch France
| |
Collapse
|
34
|
Characterization of tailless functions during Drosophila optic lobe formation. Dev Biol 2015; 405:202-13. [PMID: 26111972 DOI: 10.1016/j.ydbio.2015.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/09/2015] [Accepted: 06/11/2015] [Indexed: 11/21/2022]
Abstract
Brain development goes through phases of proliferative growth and differentiation to ensure the formation of correct number and variety of neurons. How and when naïve neuroepithelial cells decide to enter a differentiation pathway remains poorly understood. In the Drosophila visual system, four optic ganglia emerge from neuroepithelia of the inner (IPC) and outer (OPC) proliferation centers. Here we demonstrate that the orphan nuclear receptor Tailless (Tll) is a key factor for the development of all optic ganglia. We describe tll expression during larval optic lobe development in unprecedented detail and find a spatiotemporally dynamic pattern. In the larval OPC, symmetrically dividing neuroepithelial cells transform into asymmetrically dividing medulla neuroblast and into lamina precursor cells in a precisely regulated fashion. Using genetic manipulations we found that tll is required for proper neuroepithelium morphology and neuroepithelial cell survival. We show that tll regulates the precise timing of the transition from neuroepithelial cells to medulla neuroblasts. In particular, however, we demonstrate that tll has a crucial role for the specification of lamina precursor cells. We propose that the Tll/Tlx transcription factors have an evolutionary conserved role in regulating neural precursor cell states in the Drosophila optic lobe and in the mammalian retina.
Collapse
|
35
|
Tastan ÖY, Liu JL. CTP Synthase Is Required for Optic Lobe Homeostasis in Drosophila. J Genet Genomics 2015; 42:261-74. [PMID: 26059773 PMCID: PMC4458259 DOI: 10.1016/j.jgg.2015.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 04/09/2015] [Accepted: 04/10/2015] [Indexed: 10/31/2022]
Abstract
CTP synthase (CTPsyn) is a metabolic enzyme responsible for the de novo synthesis of the nucleotide CTP. Several recent studies have shown that CTPsyn forms filamentous subcellular structures known as cytoophidia in bacteria, yeast, fruit flies and humans. However, it remains elusive whether and how CTPsyn and cytoophidia play a role during development. Here, we show that cytoophidia are abundant in the neuroepithelial stem cells in Drosophila optic lobes. Optic lobes are underdeveloped in CTPsyn mutants as well as in CTPsyn RNAi. Moreover, overexpressing CTPsyn impairs the development of optic lobes, specifically by blocking the transition from neuroepithelium to neuroblast. Taken together, our results indicate that CTPsyn is critical for optic lobe homeostasis in Drosophila.
Collapse
Affiliation(s)
- Ömür Y Tastan
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Ji-Long Liu
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom.
| |
Collapse
|
36
|
Stratoulias V, Heino TI. Analysis of the conserved neurotrophic factor MANF in the Drosophila adult brain. Gene Expr Patterns 2015; 18:8-15. [PMID: 25917377 DOI: 10.1016/j.gep.2015.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/30/2015] [Accepted: 04/09/2015] [Indexed: 11/29/2022]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an evolutionarily conserved neurotrophic factor that supports and protects dopaminergic neurons. The Drosophila MANF (DmMANF) null mutant animals die during early development, and DmMANF is required for the maintenance of dopamine positive neurites. The aim of this study was to investigate the role of DmMANF during later developmental stages. Here we report that DmMANF expression in the adult brain is much wider than in the embryonic and larval stages. It is expressed in both glia and neurons including dopaminergic neurons. Clonal analysis showed that DmMANF is not required cell-autonomously for the differentiation of either glia or dopaminergic neurons. In addition, DmMANF overexpression resulted in no apparent abnormal dopaminergic phenotype while DmMANF silencing in glia resulted in prolonged larval stage.
Collapse
Affiliation(s)
| | - Tapio I Heino
- Department of Biosciences, University of Helsinki, FI-00014 Helsinki, Finland.
| |
Collapse
|
37
|
Haynes PR, Christmann BL, Griffith LC. A single pair of neurons links sleep to memory consolidation in Drosophila melanogaster. eLife 2015; 4:e03868. [PMID: 25564731 PMCID: PMC4305081 DOI: 10.7554/elife.03868] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 01/07/2015] [Indexed: 12/17/2022] Open
Abstract
Sleep promotes memory consolidation in humans and many other species, but the physiological and anatomical relationships between sleep and memory remain unclear. Here, we show the dorsal paired medial (DPM) neurons, which are required for memory consolidation in Drosophila, are sleep-promoting inhibitory neurons. DPMs increase sleep via release of GABA onto wake-promoting mushroom body (MB) α'/β' neurons. Functional imaging demonstrates that DPM activation evokes robust increases in chloride in MB neurons, but is unable to cause detectable increases in calcium or cAMP. Downregulation of α'/β' GABAA and GABABR3 receptors results in sleep loss, suggesting these receptors are the sleep-relevant targets of DPM-mediated inhibition. Regulation of sleep by neurons necessary for consolidation suggests that these brain processes may be functionally interrelated via their shared anatomy. These findings have important implications for the mechanistic relationship between sleep and memory consolidation, arguing for a significant role of inhibitory neurotransmission in regulating these processes.
Collapse
Affiliation(s)
- Paula R Haynes
- Department of Biology, Volen Center for Complex Systems, National Center for Behavioral Genomics, Brandeis University, Waltham, United States
| | - Bethany L Christmann
- Department of Biology, Volen Center for Complex Systems, National Center for Behavioral Genomics, Brandeis University, Waltham, United States
| | - Leslie C Griffith
- Department of Biology, Volen Center for Complex Systems, National Center for Behavioral Genomics, Brandeis University, Waltham, United States
| |
Collapse
|
38
|
Apitz H, Salecker I. A region-specific neurogenesis mode requires migratory progenitors in the Drosophila visual system. Nat Neurosci 2015; 18:46-55. [PMID: 25501037 PMCID: PMC4338547 DOI: 10.1038/nn.3896] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 11/13/2014] [Indexed: 12/14/2022]
Abstract
Brain areas each generate specific neuron subtypes during development. However, underlying regional variations in neurogenesis strategies and regulatory mechanisms remain poorly understood. In Drosophila, neurons in four optic lobe ganglia originate from two neuroepithelia, the outer (OPC) and inner (IPC) proliferation centers. Using genetic manipulations, we found that one IPC neuroepithelial domain progressively transformed into migratory progenitors that matured into neural stem cells (neuroblasts) in a second domain. Progenitors emerged by an epithelial-mesenchymal transition-like mechanism that required the Snail-family member Escargot and, in subdomains, Decapentaplegic signaling. The proneural factors Lethal of scute and Asense differentially controlled progenitor supply and maturation into neuroblasts. These switched expression from Asense to a third proneural protein, Atonal. Dichaete and Tailless mediated this transition, which was essential for generating two neuron populations at defined positions. We propose that this neurogenesis mode is central for setting up a new proliferative zone to facilitate spatio-temporal matching of neurogenesis and connectivity across ganglia.
Collapse
Affiliation(s)
- Holger Apitz
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, London, UK
| | - Iris Salecker
- Division of Molecular Neurobiology, MRC National Institute for Medical Research, London, UK
| |
Collapse
|
39
|
Cattenoz PB, Giangrande A. New insights in the clockwork mechanism regulating lineage specification: Lessons from the Drosophila nervous system. Dev Dyn 2014; 244:332-41. [PMID: 25399853 DOI: 10.1002/dvdy.24228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Powerful transcription factors called fate determinants induce robust differentiation programs in multipotent cells and trigger lineage specification. These factors guarantee the differentiation of specific tissues/organs/cells at the right place and the right moment to form a fully functional organism. Fate determinants are activated by temporal, positional, epigenetic, and post-transcriptional cues, hence integrating complex and dynamic developmental networks. In turn, they activate specific transcriptional/epigenetic programs that secure novel molecular landscapes. RESULTS In this review, we use the Drosophila Gcm glial determinant as a model to discuss the mechanisms that allow lineage specification in the nervous system. The dynamic regulation of Gcm via interlocked loops has recently emerged as a key event in the establishment of stable identity. Gcm induces gliogenesis while triggering its own extinction, thus preventing the appearance of metastable states and neoplastic processes. CONCLUSIONS Using simple animal models that allow in vivo manipulations provides a key tool to disentangle the complex regulation of cell fate determinants.
Collapse
Affiliation(s)
- Pierre B Cattenoz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, Illkirch, France; Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
40
|
Jones BW. Characterization of missense alleles of the glial cells missing gene of Drosophila. Genesis 2014; 52:864-9. [PMID: 25044731 DOI: 10.1002/dvg.22801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 11/12/2022]
Abstract
Glial cells missing (Gcm) is the primary regulator of glial cell fate in Drosophila. Gcm belongs to a small family of transcriptional regulators involved in fundamental developmental processes found in diverse animal phyla including vertebrates. Gcm proteins contain the highly conserved DNA-binding GCM domain, which recognizes an octamer DNA sequence. To date, studies in Drosophila have primarily relied on gcm alleles caused by P-element induced DNA deletions at the gcm locus, as well as a null allele caused by a single base pair substitution in the GCM domain that completely abolishes DNA binding. Here I characterize two hypomorphic missense alleles of gcm with intermediate glial cells missing phenotypes. In embryos homozygous for either of these gcm alleles the number of glial cells in the central nervous cystem (CNS) is reduced approximately in half. Both alleles have single amino acid changes in the GCM domain. These results suggest that Gcm protein activities in these mutant alleles have been attenuated such that they are operating at threshold levels, and trigger glial cell differentiation in neural precursors in the CNS in a stochastic fashion. These hypomorphic alleles provide additional genetic resources for understanding Gcm functions and structure in Drosophila and other species.
Collapse
Affiliation(s)
- Bradley W Jones
- Department of Biology, The University of Mississippi, 122 Shoemaker Hall, University, Mississippi
| |
Collapse
|
41
|
Apitz H, Salecker I. A Challenge of Numbers and Diversity: Neurogenesis in theDrosophilaOptic Lobe. J Neurogenet 2014; 28:233-49. [DOI: 10.3109/01677063.2014.922558] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
42
|
Multiple mechanisms modulate distinct cellular susceptibilities toward apoptosis in the developing Drosophila eye. Dev Cell 2014; 30:48-60. [PMID: 24981611 DOI: 10.1016/j.devcel.2014.05.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/09/2014] [Accepted: 05/10/2014] [Indexed: 02/06/2023]
Abstract
Although apoptosis is mechanistically well understood, a comprehensive understanding of how cells modulate their susceptibility toward apoptosis in a developing tissue is lacking. Here, we reveal striking dynamics in the apoptotic susceptibilities of different cell types in the Drosophila retina over a period of only 24 hr. Mitotic cells are extremely susceptible to apoptotic signals, while postmitotic cells have developed several strategies to promote survival. For example, photoreceptor neurons accumulate the inhibitor of apoptosis, Diap1. In unspecified cells, Cullin-3-mediated degradation keeps Diap1 levels low. These cells depend on EGFR signaling for survival. As development proceeds, developmentally older photoreceptors degrade Diap1, resulting in increased apoptosis susceptibility. Finally, R8 photoreceptors have very efficient survival mechanisms independent of EGFR or Diap1. These examples illustrate how complex cellular susceptibility toward apoptosis is regulated in a developing organ. Similar complexities may regulate apoptosis susceptibilities in mammalian development, and tumor cells may take advantage of it.
Collapse
|
43
|
Schwabe T, Borycz JA, Meinertzhagen IA, Clandinin TR. Differential adhesion determines the organization of synaptic fascicles in the Drosophila visual system. Curr Biol 2014; 24:1304-1313. [PMID: 24881879 PMCID: PMC4500537 DOI: 10.1016/j.cub.2014.04.047] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/20/2014] [Accepted: 04/24/2014] [Indexed: 01/20/2023]
Abstract
BACKGROUND Neuronal circuits in worms, flies, and mammals are organized so as to minimize wiring length for a functional number of synaptic connections, a phenomenon called wiring optimization. However, the molecular mechanisms that establish optimal wiring during development are unknown. We addressed this question by studying the role of N-cadherin in the development of optimally wired neurite fascicles in the peripheral visual system of Drosophila. RESULTS Photoreceptor axons surround the dendrites of their postsynaptic targets, called lamina cells, within a concentric fascicle called a cartridge. N-cadherin is expressed at higher levels in lamina cells than in photoreceptors, and all genetic manipulations that invert these relative differences displace lamina cells to the periphery and relocate photoreceptor axon terminals into the center. CONCLUSIONS Differential expression of a single cadherin is both necessary and sufficient to determine cartridge structure because it positions the most-adhesive elements that make the most synapses at the core and the less-adhesive elements that make fewer synapses at the periphery. These results suggest a general model by which differential adhesion can be utilized to determine the relative positions of axons and dendrites to establish optimal wiring.
Collapse
Affiliation(s)
- Tina Schwabe
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | - Jolanta A Borycz
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Ian A Meinertzhagen
- Department of Psychology and Neuroscience, Life Sciences Centre, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Thomas R Clandinin
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
44
|
Piñeiro C, Lopes CS, Casares F. A conserved transcriptional network regulates lamina development in the Drosophila visual system. Development 2014; 141:2838-47. [PMID: 24924198 DOI: 10.1242/dev.108670] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The visual system of insects is a multilayered structure composed externally by the compound eye and internally by the three ganglia of the optic lobe: lamina, medulla and the lobula complex. The differentiation of lamina neurons depends heavily on Hedgehog (Hh) signaling, which is delivered by the incoming photoreceptor axons, and occurs in a wave-like fashion. Despite the primary role of lamina neurons in visual perception, it is still unclear how these neurons are specified from neuroepithelial (NE) progenitors. Here we show that a homothorax (hth)-eyes absent (eya)-sine oculis (so)-dachshund (dac) gene regulatory cassette is involved in this specification. Lamina neurons differentiate from NE progenitors that express hth, eya and so. One of the first events in the differentiation of lamina neurons is the upregulation of dac expression in response to Hh signaling. We show that this dac upregulation, which marks the transition from NE progenitors into lamina precursors, also requires Eya/So, the expression of which is locked in by mutual feedback. dac expression is crucial for lamina differentiation because it ensures repression of hth, a negative regulator of single-minded, and thus dac allows further lamina neuron differentiation. Therefore, the specification of lamina neurons is controlled by coupling the cell-autonomous hth-eya-so-dac regulatory cassette to Hh signaling.
Collapse
Affiliation(s)
- Cristina Piñeiro
- CABD (Andalusian Centre for Developmental Biology), CSIC-UPO-JA, Seville 41013, Spain
| | - Carla S Lopes
- CABD (Andalusian Centre for Developmental Biology), CSIC-UPO-JA, Seville 41013, Spain
| | - Fernando Casares
- CABD (Andalusian Centre for Developmental Biology), CSIC-UPO-JA, Seville 41013, Spain
| |
Collapse
|
45
|
Generation of mice encoding a conditional null allele of Gcm2. Transgenic Res 2014; 23:631-41. [PMID: 24736975 DOI: 10.1007/s11248-014-9799-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
Abstract
Glial cells missing homolog 2 (GCM2) is a transcription factor that is expressed predominately in the pharyngeal pouches and, at later stages, in the developing and mature parathyroid glands. In humans, loss of GCM2 function, either through recessive apomorphic mutations or dominant inhibitor mutations in the human GCM2 gene, leads to isolated hypoparathyroidism. In mice, homozygous disruption of Gcm2 by conventional gene targeting results in parathyroid aplasia and hypoparathyroidism. In this study, we report the generation and functional characterization of mice encoding a conditional null allele of Gcm2. We demonstrate the functional integrity of the conditional Gcm2 allele and report successful in vivo deletion of exon 2 using Cre recombinase. The mice with conditional deletion of Gcm2 displayed phenotypes similar to those previously described for a conventional Gcm2 knockout, including perinatal lethality, hypocalemia, low or undetectable serum levels of parathyroid hormone, and absent parathyroid glands. The production of a conditional mutant allele for Gcm2 represents a valuable resource for the study of the temporal- and spatial-specific roles for Gcm2, and for understanding the postnatal activities of GCM2 protein.
Collapse
|
46
|
Freeman MR, Rowitch DH. Evolving concepts of gliogenesis: a look way back and ahead to the next 25 years. Neuron 2013; 80:613-23. [PMID: 24183014 DOI: 10.1016/j.neuron.2013.10.034] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glial cells are present in all organisms with a CNS and, with increasing brain complexity, glial cells have undergone substantive increases in cell number, diversity, and functions. Invertebrates, such as Drosophila, possess glial subtypes with similarity to mammalian astrocytes in their basic morphology and function, representing fertile ground for unraveling fundamental aspects of glial biology. Although glial subtypes in simple organisms may be relatively homogenous, emerging evidence suggests the possibility that mammalian astrocytes might be highly diversified to match the needs of local neuronal subtypes. In this Perspective, we review classic and new roles identified for astrocytes and oligodendrocytes by recent studies. We propose that delineating genetic and developmental programs across species will be essential to understand the core functions of glia that allow enhanced neuronal function and to achieve new insights into glial roles in higher-order brain function and neurological disease.
Collapse
Affiliation(s)
- Marc R Freeman
- University of Massachusetts, and Howard Hughes Medical Institute, Worcester, MA 01605, USA.
| | | |
Collapse
|
47
|
Cherry S, Jin EJ, Ozel MN, Lu Z, Agi E, Wang D, Jung WH, Epstein D, Meinertzhagen IA, Chan CC, Hiesinger PR. Charcot-Marie-Tooth 2B mutations in rab7 cause dosage-dependent neurodegeneration due to partial loss of function. eLife 2013; 2:e01064. [PMID: 24327558 PMCID: PMC3857549 DOI: 10.7554/elife.01064] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The small GTPase Rab7 is a key regulator of endosomal maturation in eukaryotic cells. Mutations in rab7 are thought to cause the dominant neuropathy Charcot-Marie-Tooth 2B (CMT2B) by a gain-of-function mechanism. Here we show that loss of rab7, but not overexpression of rab7 CMT2B mutants, causes adult-onset neurodegeneration in a Drosophila model. All CMT2B mutant proteins retain 10–50% function based on quantitative imaging, electrophysiology, and rescue experiments in sensory and motor neurons in vivo. Consequently, expression of CMT2B mutants at levels between 0.5 and 10-fold their endogenous levels fully rescues the neuropathy-like phenotypes of the rab7 mutant. Live imaging reveals that CMT2B proteins are inefficiently recruited to endosomes, but do not impair endosomal maturation. These findings are not consistent with a gain-of-function mechanism. Instead, they indicate a dosage-dependent sensitivity of neurons to rab7-dependent degradation. Our results suggest a therapeutic approach opposite to the currently proposed reduction of mutant protein function. DOI:http://dx.doi.org/10.7554/eLife.01064.001 Charcot-Marie-Tooth disease is an inherited disorder of the nervous system with symptoms that typically begin in adolescence or early adulthood. The sensory and motor nerves gradually degenerate, causing muscles to waste away and leading to the loss of touch sensation across the body. One subtype of the disease—Charcot-Marie-Tooth 2B—is caused by mutations in a gene called rab7, which codes for a protein that helps to regulate the breakdown of waste proteins inside cells. Charcot-Marie-Tooth 2B is described as a genetically dominant disorder because all patients have one wild type copy and one mutant copy of the rab7 gene. Overexpression of the mutant gene in cells grown in culture alters many of the signaling pathways inside the cells, but it is unclear whether these alterations cause the pathology seen in the disease. Now, Cherry et al. have obtained new insights into the genetics of Charcot-Marie-Tooth 2B by creating the first animal model of the disorder. Fruit flies that did not have the rab7 gene in the light-sensitive sensory neurons in their eyes were used to compare normal and mutant cells. While the two cell types were initially similar, the mutant cells gradually degenerated in the adult animal. By contrast, cells that overexpressed a mutant form of the rab7 gene continued to function normally throughout adulthood. Moreover, when mutant Rab7 proteins were introduced into the cells that lacked the rab7 gene, the proteins restored the cells’ sensitivity to light. These results suggest that mutant Rab7 proteins do not cause degeneration; instead, it is the loss of normal Rab7 function that causes problems. At present, most research into treatment is aimed at finding ways to reduce the activity of mutant Rab7 proteins. However, the work of Cherry et al. suggests that increasing the activity of normal Rab7 proteins—or increasing the activity of alternative pathways that degrade waste proteins—may help to restore nerve function in this, and possibly other, neurodegenerative diseases. DOI:http://dx.doi.org/10.7554/eLife.01064.002
Collapse
Affiliation(s)
- Smita Cherry
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Schwabe T, Neuert H, Clandinin TR. A network of cadherin-mediated interactions polarizes growth cones to determine targeting specificity. Cell 2013; 154:351-64. [PMID: 23870124 DOI: 10.1016/j.cell.2013.06.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 04/02/2013] [Accepted: 06/07/2013] [Indexed: 01/17/2023]
Abstract
Neuronal growth cones select synaptic partners through interactions with multiple cell surfaces in their environment. Many of these interactions are adhesive, yet it is unclear how growth cones integrate adhesive cues to direct their movements. Here, we examine the mechanisms that enable photoreceptors in the Drosophila visual system to choose synaptic partners. We demonstrate that the classical cadherin, N-cadherin, and an atypical cadherin, Flamingo, act redundantly to instruct the targeting choices made by every photoreceptor axon. These molecules gradually bias the spatial distribution of growth cone filopodia, polarizing each growth cone toward its future synaptic target before direct contact with the target occurs. We demonstrate that these molecules are localized to distinct subcellular domains and create a network of adhesive interactions distributed across many growth cones. Because this network comprises multiple redundant interactions, a complex wiring diagram can be constructed with extraordinary fidelity, suggesting a general principle.
Collapse
Affiliation(s)
- Tina Schwabe
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA
| | | | | |
Collapse
|
49
|
Abstract
The evolutionarily conserved JAK/STAT pathway plays important roles in development and disease processes in humans. Although the signaling process has been well established, we know relatively little about what the relevant target genes are that mediate JAK/STAT activation during development. Here, we have used genome-wide microarrays to identify JAK/STAT targets in the optic lobes of the Drosophila brain and identified 47 genes that are positively regulated by JAK/STAT. About two-thirds of the genes encode proteins that have orthologs in humans. The STAT targets in the optic lobe appear to be different from the targets identified in other tissues, suggesting that JAK/STAT signaling may regulate different target genes in a tissue-specific manner. Functional analysis of Nop56, a cell-autonomous STAT target, revealed an essential role for this gene in the growth and proliferation of neuroepithelial stem cells in the optic lobe and an inhibitory role in lamina neurogenesis.
Collapse
|
50
|
Cattenoz PB, Giangrande A. Lineage specification in the fly nervous system and evolutionary implications. Cell Cycle 2013; 12:2753-9. [PMID: 23966161 DOI: 10.4161/cc.25918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Over the last decades, it has become clear that glia are multifunctional and plastic cells endowed with key regulatory roles. They control the response to developmental and/or pathological signals, thereby affecting neural proliferation, remodeling, survival, and regeneration. It is, therefore, important to understand the biology of these cells and the molecular mechanisms controlling their development/activity. The fly community has made major breakthroughs by characterizing the bases of gliogenesis and function. Here we describe the regulation and the role of the fly glial determinant. Then, we discuss the impact of the determinant in cell plasticity and differentiation. Finally, we address the conservation of this pathway across evolution.
Collapse
Affiliation(s)
- Pierre B Cattenoz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire; IGBMC/CNRS/INSERM/UDS; Strasbourg, France
| | | |
Collapse
|