1
|
Lee GG, Peterson AJ, Kim MJ, O’Connor MB, Park JH. Multiple isoforms of the Activin-like receptor baboon differentially regulate proliferation and conversion behaviors of neuroblasts and neuroepithelial cells in the Drosophila larval brain. PLoS One 2024; 19:e0305696. [PMID: 38913612 PMCID: PMC11195991 DOI: 10.1371/journal.pone.0305696] [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: 03/22/2024] [Accepted: 06/04/2024] [Indexed: 06/26/2024] Open
Abstract
In Drosophila coordinated proliferation of two neural stem cells, neuroblasts (NB) and neuroepithelial (NE) cells, is pivotal for proper larval brain growth that ultimately determines the final size and performance of an adult brain. The larval brain growth displays two phases based on behaviors of NB and NEs: the first one in early larval stages, influenced by nutritional status and the second one in the last larval stage, promoted by ecdysone signaling after critical weight checkpoint. Mutations of the baboon (babo) gene that produces three isoforms (BaboA-C), all acting as type-I receptors of Activin-type transforming growth factor β (TGF-β) signaling, cause a small brain phenotype due to severely reduced proliferation of the neural stem cells. In this study we show that loss of babo function severely affects proliferation of NBs and NEs as well as conversion of NEs from both phases. By analyzing babo-null and newly generated isoform-specific mutants by CRISPR mutagenesis as well as isoform-specific RNAi knockdowns in a cell- and stage-specific manner, our data support differential contributions of the isoforms for these cellular events with BaboA playing the major role. Stage-specific expression of EcR-B1 in the brain is also regulated primarily by BaboA along with function of the other isoforms. Blocking EcR function in both neural stem cells results in a small brain phenotype that is more severe than baboA-knockdown alone. In summary, our study proposes that the Babo-mediated signaling promotes proper behaviors of the neural stem cells in both phases and achieves this by acting upstream of EcR-B1 expression in the second phase.
Collapse
Affiliation(s)
- Gyunghee G. Lee
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Aidan J. Peterson
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Michael B. O’Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jae H. Park
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, United States of America
| |
Collapse
|
2
|
Nguyen PK, Cheng LY. Drosophila medulla neuroblast termination via apoptosis, differentiation, and gliogenic switch is scheduled by the depletion of the neuroepithelial stem cell pool. eLife 2024; 13:e96876. [PMID: 38905123 PMCID: PMC11262793 DOI: 10.7554/elife.96876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/20/2024] [Indexed: 06/23/2024] Open
Abstract
The brain is consisted of diverse neurons arising from a limited number of neural stem cells. Drosophila neural stem cells called neuroblasts (NBs) produces specific neural lineages of various lineage sizes depending on their location in the brain. In the Drosophila visual processing centre - the optic lobes (OLs), medulla NBs derived from the neuroepithelium (NE) give rise to neurons and glia cells of the medulla cortex. The timing and the mechanisms responsible for the cessation of medulla NBs are so far not known. In this study, we show that the termination of medulla NBs during early pupal development is determined by the exhaustion of the NE stem cell pool. Hence, altering NE-NB transition during larval neurogenesis disrupts the timely termination of medulla NBs. Medulla NBs terminate neurogenesis via a combination of apoptosis, terminal symmetric division via Prospero, and a switch to gliogenesis via Glial Cell Missing (Gcm); however, these processes occur independently of each other. We also show that temporal progression of the medulla NBs is mostly not required for their termination. As the Drosophila OL shares a similar mode of division with mammalian neurogenesis, understanding when and how these progenitors cease proliferation during development can have important implications for mammalian brain size determination and regulation of its overall function.
Collapse
Affiliation(s)
- Phuong-Khanh Nguyen
- Peter MacCallum Cancer CentreMelbourneAustralia
- Department of Anatomy and Physiology, The University of MelbourneMelbourneAustralia
| | - Louise Y Cheng
- Peter MacCallum Cancer CentreMelbourneAustralia
- Department of Anatomy and Physiology, The University of MelbourneMelbourneAustralia
- Sir Peter MacCallum Department of Oncology, The University of MelbourneMelbourneAustralia
| |
Collapse
|
3
|
Malin JA, Chen YC, Simon F, Keefer E, Desplan C. Spatial patterning controls neuron numbers in the Drosophila visual system. Dev Cell 2024; 59:1132-1145.e6. [PMID: 38531357 PMCID: PMC11078608 DOI: 10.1016/j.devcel.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/18/2023] [Accepted: 03/01/2024] [Indexed: 03/28/2024]
Abstract
Neurons must be made in the correct proportions to communicate with the appropriate synaptic partners and form functional circuits. In the Drosophila visual system, multiple subtypes of distal medulla (Dm) inhibitory interneurons are made in distinct, reproducible numbers-from 5 to 800 per optic lobe. These neurons are born from a crescent-shaped neuroepithelium called the outer proliferation center (OPC), which can be subdivided into specific domains based on transcription factor and growth factor expression. We fate mapped Dm neurons and found that more abundant neural types are born from larger neuroepithelial subdomains, while less abundant subtypes are born from smaller ones. Additionally, morphogenetic Dpp/BMP signaling provides a second layer of patterning that subdivides the neuroepithelium into smaller domains to provide more granular control of cell proportions. Apoptosis appears to play a minor role in regulating Dm neuron abundance. This work describes an underappreciated mechanism for the regulation of neuronal stoichiometry.
Collapse
Affiliation(s)
- Jennifer A Malin
- Department of Biology, New York University, New York, NY 10003, USA.
| | - Yen-Chung Chen
- Department of Biology, New York University, New York, NY 10003, USA
| | - Félix Simon
- Department of Biology, New York University, New York, NY 10003, USA
| | - Evelyn Keefer
- Department of Biology, New York University, New York, NY 10003, USA
| | - Claude Desplan
- Department of Biology, New York University, New York, NY 10003, USA.
| |
Collapse
|
4
|
Abstract
The Drosophila visual system has been a great model to study fundamental questions in neurobiology, such as neural fate specification, axon guidance, circuit formation, and information processing. The Drosophila visual system is composed of the compound eye and the optic lobe. The optic lobe is divided into four neuropils-namely, the lamina, medulla, lobula, and lobula plate. There are around 200 types of optic lobe neurons, which wire together to form a complex neural structure to processes visual information. These neurons are derived from two neuroepithelial structures-namely, the outer proliferation center (OPC) and the inner proliferation center (IPC), in the larval brain. Recent work on the Drosophila optic lobe has revealed basic principles underlying the development of this complex neural structure, and immunostaining has been a key tool in these studies. Here, we provide a brief overview of the Drosophila optic lobe structure and development, as revealed by immunostaining. First, we introduce the structure of the adult optic lobe. Then, we summarize recent advances in the study of neural fate specification during development of different parts of the optic lobe. Last, we briefly summarize general aspects of axon guidance and neuropil assembly in the optic lobe. With this review, we aim to familiarize readers with this complex neural structure and highlight the power of this great model to study neural development to facilitate further developmental and functional studies using this system.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Xin Li
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| |
Collapse
|
5
|
Martin M, Gutierrez-Avino F, Shaikh MN, Tejedor FJ. A novel proneural function of Asense is integrated with the sequential actions of Delta-Notch, L'sc and Su(H) to promote the neuroepithelial to neuroblast transition. PLoS Genet 2023; 19:e1010991. [PMID: 37871020 PMCID: PMC10621995 DOI: 10.1371/journal.pgen.1010991] [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: 04/09/2023] [Revised: 11/02/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023] Open
Abstract
In order for neural progenitors (NPs) to generate distinct populations of neurons at the right time and place during CNS development, they must switch from undergoing purely proliferative, self-renewing divisions to neurogenic, asymmetric divisions in a tightly regulated manner. In the developing Drosophila optic lobe, neuroepithelial (NE) cells of the outer proliferation center (OPC) are progressively transformed into neurogenic NPs called neuroblasts (NBs) in a medial to lateral proneural wave. The cells undergoing this transition express Lethal of Scute (L'sc), a proneural transcription factor (TF) of the Acheate Scute Complex (AS-C). Here we show that there is also a peak of expression of Asense (Ase), another AS-C TF, in the cells neighboring those with transient L'sc expression. These peak of Ase cells help to identify a new transitional stage as they have lost NE markers and L'sc, they receive a strong Notch signal and barely exhibit NB markers. This expression of Ase is necessary and sufficient to promote the NE to NB transition in a more robust and rapid manner than that of l'sc gain of function or Notch loss of function. Thus, to our knowledge, these data provide the first direct evidence of a proneural role for Ase in CNS neurogenesis. Strikingly, we found that strong Delta-Notch signaling at the lateral border of the NE triggers l'sc expression, which in turn induces ase expression in the adjacent cells through the activation of Delta-Notch signaling. These results reveal two novel non-conventional actions of Notch signaling in driving the expression of proneural factors, in contrast to the repression that Notch signaling exerts on them during classical lateral inhibition. Finally, Suppressor of Hairless (Su(H)), which seems to be upregulated late in the transitioning cells and in NBs, represses l'sc and ase, ensuring their expression is transient. Thus, our data identify a key proneural role of Ase that is integrated with the sequential activities of Delta-Notch signaling, L'sc, and Su(H), driving the progressive transformation of NE cells into NBs.
Collapse
Affiliation(s)
- Mercedes Martin
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Francisco Gutierrez-Avino
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Mirja N. Shaikh
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| | - Francisco J. Tejedor
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernandez, Sant Joan d’Alacant, Spain
| |
Collapse
|
6
|
Pollington HQ, Seroka AQ, Doe CQ. From temporal patterning to neuronal connectivity in Drosophila type I neuroblast lineages. Semin Cell Dev Biol 2023; 142:4-12. [PMID: 35659165 PMCID: PMC9938700 DOI: 10.1016/j.semcdb.2022.05.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
The development of the central nervous system (CNS) in flies and mammals requires the production of distinct neurons in different locations and times. Here we review progress on how Drosophila stem cells (neuroblasts; NBs) generate distinct neurons over time. There are two types of NBs: type I and type II NBs (defined below); here we focus on type I NBs; type II NBs are reviewed elsewhere in this issue. Type I NBs generate neural diversity via the cascading expression of specific temporal transcription factors (TTFs). TTFs are sequentially expressed in neuroblasts and required for the identity of neurons born during each TTF expression window. In this way TTFs specify the "temporal identity" or birth-order dependent identity of neurons. Recent studies have shown that TTF expression in neuroblasts alter the identity of their progeny, including directing motor neurons to form proper connectivity to the proper muscle targets, independent of their birth-order. Similarly, optic lobe (OL) type I NBs express a series of TTFs that promote proper neuron morphology and targeting to the four OL neuropils. Together, these studies demonstrate how temporal identity is crucial in promoting proper circuit assembly within the Drosophila CNS. In addition, TTF orthologs in mouse are good candidates for specifying neuron types in the neocortex and retina. In this review we highlight the recent advances in understanding the role of TTFs in CNS circuit assembly in Drosophila and reflect on the conservation of these mechanisms in mammalian CNS development.
Collapse
Affiliation(s)
- Heather Q Pollington
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Austin Q Seroka
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
7
|
Sen SQ. Generating neural diversity through spatial and temporal patterning. Semin Cell Dev Biol 2023; 142:54-66. [PMID: 35738966 DOI: 10.1016/j.semcdb.2022.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
The nervous system consists of a vast diversity of neurons and glia that are accurately assembled into functional circuits. What are the mechanisms that generate these diverse cell types? During development, an epithelial sheet with neurogenic potential is initially regionalised into spatially restricted domains of gene expression. From this, pools of neural stem cells (NSCs) with distinct molecular profiles and the potential to generate different neuron types, are specified. These NSCs then divide asymmetrically to self-renew and generate post-mitotic neurons or glia. As NSCs age, they experience transitions in gene expression, which further allows them to generate different neurons or glia over time. Versions of this general template of spatial and temporal patterning operate during the development of different parts of different nervous systems. Here, I cover our current knowledge of Drosophila brain and optic lobe development as well as the development of the vertebrate cortex and spinal cord within the framework of this above template. I highlight where our knowledge is lacking, where mechanisms beyond these might operate, and how the emergence of new technologies might help address unanswered questions.
Collapse
Affiliation(s)
- Sonia Q Sen
- Tata Institute for Genetics and Society, UAS-GKVK Campus, Bellary Road, Bangalore, India.
| |
Collapse
|
8
|
Egger B. Neural Stem Cells and Brain Tumour Models in Drosophila. Neurogenetics 2023. [DOI: 10.1007/978-3-031-07793-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
9
|
Sato M, Suzuki T. Cutting edge technologies expose the temporal regulation of neurogenesis in the Drosophila nervous system. Fly (Austin) 2022; 16:222-232. [PMID: 35549651 PMCID: PMC9116403 DOI: 10.1080/19336934.2022.2073158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
During the development of the central nervous system (CNS), extremely large numbers of neurons are produced in a regular fashion to form precise neural circuits. During this process, neural progenitor cells produce different neurons over time due to their intrinsic gene regulatory mechanisms as well as extrinsic mechanisms. The Drosophila CNS has played an important role in elucidating the temporal mechanisms that control neurogenesis over time. It has been shown that a series of temporal transcription factors are sequentially expressed in neural progenitor cells and regulate the temporal specification of neurons in the embryonic CNS. Additionally, similar mechanisms are found in the developing optic lobe and central brain in the larval CNS. However, it is difficult to elucidate the function of numerous molecules in many different cell types solely by molecular genetic approaches. Recently, omics analysis using single-cell RNA-seq and other methods has been used to study the Drosophila nervous system on a large scale and is making a significant contribution to the understanding of the temporal mechanisms of neurogenesis. In this article, recent findings on the temporal patterning of neurogenesis and the contributions of cutting-edge technologies will be reviewed.
Collapse
Affiliation(s)
- Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative,Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
| | - Takumi Suzuki
- College of Science, Department of Science, Ibaraki University, Ibaraki, Japan
| |
Collapse
|
10
|
Valentino P, Erclik T. Spalt and disco define the dorsal-ventral neuroepithelial compartments of the developing Drosophila medulla. Genetics 2022; 222:iyac145. [PMID: 36135799 PMCID: PMC9630984 DOI: 10.1093/genetics/iyac145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/14/2022] [Indexed: 11/14/2022] Open
Abstract
Spatial patterning of neural stem cell populations is a powerful mechanism by which to generate neuronal diversity. In the developing Drosophila medulla, the symmetrically dividing neuroepithelial cells of the outer proliferation center crescent are spatially patterned by the nonoverlapping expression of 3 transcription factors: Vsx1 in the center, Optix in the adjacent arms, and Rx in the tips. These spatial genes compartmentalize the outer proliferation center and, together with the temporal patterning of neuroblasts, act to diversify medulla neuronal fates. The observation that the dorsal and ventral halves of the outer proliferation center also grow as distinct compartments, together with the fact that a subset of neuronal types is generated from only one half of the crescent, suggests that additional transcription factors spatially pattern the outer proliferation center along the dorsal-ventral axis. Here, we identify the spalt (salm and salr) and disco (disco and disco-r) genes as the dorsal-ventral patterning transcription factors of the outer proliferation center. Spalt and Disco are differentially expressed in the dorsal and ventral outer proliferation center from the embryo through to the third instar larva, where they cross-repress each other to form a sharp dorsal-ventral boundary. We show that hedgehog is necessary for Disco expression in the embryonic optic placode and that disco is subsequently required for the development of the ventral outer proliferation center and its neuronal progeny. We further demonstrate that this dorsal-ventral patterning axis acts independently of Vsx1-Optix-Rx and thus propose that Spalt and Disco represent a third outer proliferation center patterning axis that may act to further diversify medulla fates.
Collapse
Affiliation(s)
- Priscilla Valentino
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Ted Erclik
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
11
|
Nguyen PK, Cheng LY. Non-autonomous regulation of neurogenesis by extrinsic cues: a Drosophila perspective. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac004. [PMID: 38596708 PMCID: PMC10913833 DOI: 10.1093/oons/kvac004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 04/11/2024]
Abstract
The formation of a functional circuitry in the central nervous system (CNS) requires the correct number and subtypes of neural cells. In the developing brain, neural stem cells (NSCs) self-renew while giving rise to progenitors that in turn generate differentiated progeny. As such, the size and the diversity of cells that make up the functional CNS depend on the proliferative properties of NSCs. In the fruit fly Drosophila, where the process of neurogenesis has been extensively investigated, extrinsic factors such as the microenvironment of NSCs, nutrients, oxygen levels and systemic signals have been identified as regulators of NSC proliferation. Here, we review decades of work that explores how extrinsic signals non-autonomously regulate key NSC characteristics such as quiescence, proliferation and termination in the fly.
Collapse
Affiliation(s)
- Phuong-Khanh Nguyen
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
12
|
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: 21] [Impact Index Per Article: 10.5] [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
|
13
|
Cao X, Rojas M, Pastor-Pareja JC. Intrinsic and damage-induced JAK/STAT signaling regulate developmental timing by the Drosophila prothoracic gland. Dis Model Mech 2021; 15:273570. [PMID: 34842272 PMCID: PMC8807578 DOI: 10.1242/dmm.049160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/15/2021] [Indexed: 11/20/2022] Open
Abstract
Development involves tightly paced, reproducible sequences of events, yet it must adjust to conditions external to it, such as resource availability and organismal damage. A major mediator of damage-induced immune responses in vertebrates and insects is JAK/STAT signaling. At the same time, JAK/STAT activation by the Drosophila Upd cytokines is pleiotropically involved in normal development of multiple organs. Whether inflammatory and developmental JAK/STAT roles intersect is unknown. Here, we show that JAK/STAT is active during development of the prothoracic gland (PG), which controls metamorphosis onset through ecdysone production. Reducing JAK/STAT signaling decreased PG size and advanced metamorphosis. Conversely, JAK/STAT hyperactivation by overexpression of pathway components or SUMOylation loss caused PG hypertrophy and metamorphosis delay. Tissue damage and tumors, known to secrete Upd cytokines, also activated JAK/STAT in the PG and delayed metamorphosis, at least in part by inducing expression of the JAK/STAT target Apontic. JAK/STAT damage signaling, therefore, regulates metamorphosis onset by co-opting its developmental role in the PG. Our findings in Drosophila provide insights on how systemic effects of damage and cancer can interfere with hormonally controlled development and developmental transitions. Summary: Damage signaling from tumors mediated by JAK/STAT-activating Upd cytokines delays the Drosophila larva–pupa transition through co-option of a JAK/STAT developmental role in the prothoracic gland.
Collapse
Affiliation(s)
- Xueya Cao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Marta Rojas
- School of Medicine, Tsinghua University, Beijing, China
| | - José Carlos Pastor-Pareja
- School of Life Sciences, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
14
|
Transcriptional and epigenetic regulation of temporal patterning in neural progenitors. Dev Biol 2021; 481:116-128. [PMID: 34666024 DOI: 10.1016/j.ydbio.2021.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/05/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022]
Abstract
During development, neural progenitors undergo temporal patterning as they age to sequentially generate differently fated progeny. Temporal patterning of neural progenitors is relatively well-studied in Drosophila. Temporal cascades of transcription factors or opposing temporal gradients of RNA-binding proteins are expressed in neural progenitors as they age to control the fates of the progeny. The temporal progression is mostly driven by intrinsic mechanisms including cross-regulations between temporal genes, but environmental cues also play important roles in certain transitions. Vertebrate neural progenitors demonstrate greater plasticity in response to extrinsic cues. Recent studies suggest that vertebrate neural progenitors are also temporally patterned by a combination of transcriptional and post-transcriptional mechanisms in response to extracellular signaling to regulate neural fate specification. In this review, we summarize recent advances in the study of temporal patterning of neural progenitors in Drosophila and vertebrates. We also discuss the involvement of epigenetic mechanisms, specifically the Polycomb group complexes and ATP-dependent chromatin remodeling complexes, in the temporal patterning of neural progenitors.
Collapse
|
15
|
Abstract
Notch signalling is a well-conserved signalling pathway that regulates cell fate through cell-cell communication. A typical feature of Notch signalling is ‘lateral inhibition’, whereby two neighbouring cells of equivalent state of differentiation acquire different cell fates. Recently, mathematical and computational approaches have addressed the Notch dynamics in Drosophila neural development. Typical examples of lateral inhibition are observed in the specification of neural stem cells in the embryo and sensory organ precursors in the thorax. In eye disc development, Notch signalling cooperates with other signalling pathways to define the evenly spaced positioning of the photoreceptor cells. The interplay between Notch and epidermal growth factor receptor signalling regulates the timing of neural stem cell differentiation in the optic lobe. In this review, we summarize the theoretical studies that have been conducted to elucidate the Notch dynamics in these systems and discuss the advantages of combining mathematical models with biological experiments.
Collapse
Affiliation(s)
- Tetsuo Yasugi
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa, Japan.,Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
16
|
Neural specification, targeting, and circuit formation during visual system assembly. Proc Natl Acad Sci U S A 2021; 118:2101823118. [PMID: 34183440 DOI: 10.1073/pnas.2101823118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Like other sensory systems, the visual system is topographically organized: Its sensory neurons, the photoreceptors, and their targets maintain point-to-point correspondence in physical space, forming a retinotopic map. The iterative wiring of circuits in the visual system conveniently facilitates the study of its development. Over the past few decades, experiments in Drosophila have shed light on the principles that guide the specification and connectivity of visual system neurons. In this review, we describe the main findings unearthed by the study of the Drosophila visual system and compare them with similar events in mammals. We focus on how temporal and spatial patterning generates diverse cell types, how guidance molecules distribute the axons and dendrites of neurons within the correct target regions, how vertebrates and invertebrates generate their retinotopic map, and the molecules and mechanisms required for neuronal migration. We suggest that basic principles used to wire the fly visual system are broadly applicable to other systems and highlight its importance as a model to study nervous system development.
Collapse
|
17
|
Abstract
AbstractIn the developing Drosophila CNS, two pools of neural stem cells, the symmetrically dividing progenitors in the neuroepithelium (NE) and the asymmetrically dividing neuroblasts (NBs) generate the majority of the neurons that make up the adult central nervous system (CNS). The generation of a correct sized brain depends on maintaining the fine balance between neural stem cell self-renewal and differentiation, which are regulated by cell-intrinsic and cell-extrinsic cues. In this review, we will discuss our current understanding of how self-renewal and differentiation are regulated in the two neural stem cell pools, and the consequences of the deregulation of these processes.
Collapse
Affiliation(s)
- Francesca Froldi
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
| | - Milán Szuperák
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
| | - Louise Y. Cheng
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
18
|
Intracellular trafficking of Notch orchestrates temporal dynamics of Notch activity in the fly brain. Nat Commun 2021; 12:2083. [PMID: 33828096 PMCID: PMC8027629 DOI: 10.1038/s41467-021-22442-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 03/16/2021] [Indexed: 12/03/2022] Open
Abstract
While Delta non-autonomously activates Notch in neighboring cells, it autonomously inactivates Notch through cis-inhibition, the molecular mechanism and biological roles of which remain elusive. The wave of differentiation in the Drosophila brain, the ‘proneural wave’, is an excellent model for studying Notch signaling in vivo. Here, we show that strong nonlinearity in cis-inhibition reproduces the second peak of Notch activity behind the proneural wave in silico. Based on this, we demonstrate that Delta expression induces a quick degradation of Notch in late endosomes and the formation of the twin peaks of Notch activity in vivo. Indeed, the amount of Notch is upregulated and the twin peaks are fused forming a single peak when the function of Delta or late endosomes is compromised. Additionally, we show that the second Notch peak behind the wavefront controls neurogenesis. Thus, intracellular trafficking of Notch orchestrates the temporal dynamics of Notch activity and the temporal patterning of neurogenesis. During Drosophila development, two peaks of Notch activity propagate across the neuroepithelium to generate neuroblasts. Here, the authors show Notch cis-inhibition under the control of intracellular Notch trafficking establishes these two peaks, which temporally control neurogenesis in the brain.
Collapse
|
19
|
Shard C, Luna-Escalante J, Schweisguth F. Tissue-wide coordination of epithelium-to-neural stem cell transition in the Drosophila optic lobe requires Neuralized. J Cell Biol 2021; 219:152101. [PMID: 32946560 PMCID: PMC7594497 DOI: 10.1083/jcb.202005035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/06/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
Abstract
Many tissues are produced by specialized progenitor cells emanating from epithelia via epithelial-to-mesenchymal transition (EMT). Most studies have so far focused on EMT involving single or isolated groups of cells. Here we describe an EMT-like process that requires tissue-level coordination. This EMT-like process occurs along a continuous front in the Drosophila optic lobe neuroepithelium to produce neural stem cells (NSCs). We find that emerging NSCs remain epithelial and apically constrict before dividing asymmetrically to produce neurons. Apical constriction is associated with contractile myosin pulses and involves RhoGEF3 and down-regulation of the Crumbs complex by the E3 ubiquitin ligase Neuralized. Anisotropy in Crumbs complex levels also results in accumulation of junctional myosin. Disrupting the regulation of Crumbs by Neuralized lowered junctional myosin and led to imprecision in the integration of emerging NSCs into the front. Thus, Neuralized promotes smooth progression of the differentiation front by coupling epithelium remodeling at the tissue level with NSC fate acquisition.
Collapse
Affiliation(s)
- Chloé Shard
- Institut Pasteur, Paris, France.,UMR3738, Centre National de la Recherche Scientifique, Paris, France
| | - Juan Luna-Escalante
- Institut Pasteur, Paris, France.,UMR3738, Centre National de la Recherche Scientifique, Paris, France.,Laboratoire de Physique, Ecole Normale Supérieure, Centre National de la Recherche Scientifique, Sorbonne Université, Université Paris Diderot, Paris, France
| | - François Schweisguth
- Institut Pasteur, Paris, France.,UMR3738, Centre National de la Recherche Scientifique, Paris, France
| |
Collapse
|
20
|
A continuation method for spatially discretized models with nonlocal interactions conserving size and shape of cells and lattices. J Math Biol 2020; 81:981-1028. [PMID: 32959067 PMCID: PMC7560951 DOI: 10.1007/s00285-020-01534-6] [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: 01/10/2020] [Revised: 07/15/2020] [Indexed: 11/18/2022]
Abstract
In this paper, we introduce a continuation method for the spatially discretized models, while conserving the size and shape of the cells and lattices. This proposed method is realized using the shift operators and nonlocal operators of convolution types. Through this method and using the shift operator, the nonlinear spatially discretized model on the uniform and nonuniform lattices can be systematically converted into a spatially continuous model; this renders both models point-wisely equivalent. Moreover, by the convolution with suitable kernels, we mollify the shift operator and approximate the spatially discretized models using the nonlocal evolution equations, rendering suitable for the application in both experimental and mathematical analyses. We also demonstrate that this approximation is supported by the singular limit analysis, and that the information of the lattice and cells is expressed in the shift and nonlocal operators. The continuous models designed using our method can successfully replicate the patterns corresponding to those of the original spatially discretized models obtained from the numerical simulations. Furthermore, from the observations of the isotropy of the Delta–Notch signaling system in a developing real fly brain, we propose a radially symmetric kernel for averaging the cell shape using our continuation method. We also apply our method for cell division and proliferation to spatially discretized models of the differentiation wave and describe the discrete models on the sphere surface. Finally, we demonstrate an application of our method in the linear stability analysis of the planar cell polarity model.
Collapse
|
21
|
Das P, Bhadra MP. Histone deacetylase (Rpd3) regulates Drosophila early brain development via regulation of Tailless. Open Biol 2020; 10:200029. [PMID: 32873153 PMCID: PMC7536075 DOI: 10.1098/rsob.200029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/29/2020] [Indexed: 11/29/2022] Open
Abstract
Tailless is a committed transcriptional repressor and principal regulator of the brain and eye development in Drosophila. Rpd3, the histone deacetylase, is an established repressor that interacts with co-repressors like Sin3a, Prospero, Brakeless and Atrophin. This study aims at deciphering the role of Rpd3 in embryonic segmentation and larval brain development in Drosophila. It delineates the mechanism of Tailless regulation by Rpd3, along with its interacting partners. There was a significant reduction in Tailless in Rpd3 heteroallelic mutant embryos, substantiating that Rpd3 is indispensable for the normal Tailless expression. The expression of the primary readout, Tailless was correlative to the expression of the neural cell adhesion molecule homologue, Fascilin2 (Fas2). Rpd3 also aids in the proper development of the mushroom body. Both Tailless and Fas2 expression are reported to be antagonistic to the epidermal growth factor receptor (EGFR) expression. The decrease in Tailless and Fas2 expression highlights that EGFR is upregulated in the larval mutants, hindering brain development. This study outlines the axis comprising Rpd3, dEGFR, Tailless and Fas2, which interact to fine-tune the early segmentation and larval brain development. Therefore, Rpd3 along with Tailless has immense significance in early embryogenesis and development of the larval brain.
Collapse
Affiliation(s)
- Paromita Das
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR) Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai 600 113, India
| | - Manika Pal Bhadra
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research (AcSIR) Training and Development Complex, CSIR Campus, CSIR Road, Taramani, Chennai 600 113, India
| |
Collapse
|
22
|
Baccino-Calace M, Prieto D, Cantera R, Egger B. Compartment and cell-type specific hypoxia responses in the developing Drosophila brain. Biol Open 2020; 9:9/8/bio053629. [PMID: 32816692 PMCID: PMC7449796 DOI: 10.1242/bio.053629] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Environmental factors such as the availability of oxygen are instructive cues that regulate stem cell maintenance and differentiation. We used a genetically encoded biosensor to monitor the hypoxic state of neural cells in the larval brain of Drosophila. The biosensor reveals brain compartment and cell-type specific levels of hypoxia. The values correlate with differential tracheolation that is observed throughout development between the central brain and the optic lobe. Neural stem cells in both compartments show the strongest hypoxia response while intermediate progenitors, neurons and glial cells reveal weaker responses. We demonstrate that the distance between a cell and the next closest tracheole is a good predictor of the hypoxic state of that cell. Our study indicates that oxygen availability appears to be the major factor controlling the hypoxia response in the developing Drosophila brain and that cell intrinsic and cell-type specific factors contribute to modulate the response in an unexpected manner. This article has an associated First Person interview with the first author of the paper. Summary: A fluorescent biosensor reveals cell type specific hypoxia levels in the Drosophila brain in unprecedented detail. It paves the way for further functional studies addressing the role of oxygen in neural stem cell maintenance and differentiation.
Collapse
Affiliation(s)
- Martin Baccino-Calace
- Developmental Neurobiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Daniel Prieto
- Developmental Neurobiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay
| | - Rafael Cantera
- Developmental Neurobiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo 11600, Uruguay.,Zoology Department, Stockholm University, Stockholm 106 91, Sweden
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg CH-1700, Switzerland
| |
Collapse
|
23
|
Liu C, Trush O, Han X, Wang M, Takayama R, Yasugi T, Hayashi T, Sato M. Dscam1 establishes the columnar units through lineage-dependent repulsion between sister neurons in the fly brain. Nat Commun 2020; 11:4067. [PMID: 32792493 PMCID: PMC7426427 DOI: 10.1038/s41467-020-17931-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 07/24/2020] [Indexed: 11/15/2022] Open
Abstract
The brain is organized morphologically and functionally into a columnar structure. According to the radial unit hypothesis, neurons from the same lineage form a radial unit that contributes to column formation. However, the molecular mechanisms that link neuronal lineage and column formation remain elusive. Here, we show that neurons from the same lineage project to different columns under control of Down syndrome cell adhesion molecule (Dscam) in the fly brain. Dscam1 is temporally expressed in newly born neuroblasts and is inherited by their daughter neurons. The transient transcription of Dscam1 in neuroblasts enables the expression of the same Dscam1 splice isoform within cells of the same lineage, causing lineage-dependent repulsion. In the absence of Dscam1 function, neurons from the same lineage project to the same column. When the splice diversity of Dscam1 is reduced, column formation is significantly compromised. Thus, Dscam1 controls column formation through lineage-dependent repulsion. Columns are the functional and morphological unit of the brain, but how neurons assemble into this structure was unclear. Here, the authors show that Dscam gene rewires neurons that derive from the same stem cell to establish columns through the process of lineage-dependent repulsion.
Collapse
Affiliation(s)
- Chuyan Liu
- Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Olena Trush
- Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Xujun Han
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Miaoxing Wang
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Rie Takayama
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Tetsuo Yasugi
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Takashi Hayashi
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Makoto Sato
- Laboratory of Developmental Neurobiology, Graduate School of Medical Sciences, 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.
| |
Collapse
|
24
|
Okamoto N, Yamanaka N. Steroid Hormone Entry into the Brain Requires a Membrane Transporter in Drosophila. Curr Biol 2020; 30:359-366.e3. [PMID: 31928869 DOI: 10.1016/j.cub.2019.11.085] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/14/2019] [Accepted: 11/28/2019] [Indexed: 02/08/2023]
Abstract
Steroid hormones control various aspects of brain development and behavior in metazoans, but how they enter the central nervous system (CNS) through the blood-brain barrier (BBB) remains poorly understood. It is generally believed that steroid hormones freely diffuse through the plasma membrane of the BBB cells to reach the brain [1], because of the predominant "simple diffusion" model of steroid hormone transport across cell membranes. Recently, however, we challenged the simple diffusion model by showing that a Drosophila organic anion-transporting polypeptide (OATP), which we named Ecdysone Importer (EcI), is required for cellular uptake of the primary insect steroid hormone ecdysone [2]. As ecdysone is first secreted into the hemolymph before reaching the CNS [3], our finding raised the question of how ecdysone enters the CNS through the BBB to exert its diverse role in Drosophila brain development. Here, we demonstrate in the Drosophila BBB that EcI is indispensable for ecdysone entry into the CNS to facilitate brain development. EcI is highly expressed in surface glial cells that form the BBB, and EcI knockdown in the BBB suppresses ecdysone signaling within the CNS and blocks ecdysone-mediated neuronal events during development. In an ex vivo culture system, the CNS requires EcI in the BBB to incorporate ecdysone from the culture medium. Our results suggest a transporter-mediated mechanism of steroid hormone entry into the CNS, which may provide important implications in controlling brain development and behavior by regulating steroid hormone permeability across the BBB.
Collapse
Affiliation(s)
- Naoki Okamoto
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA
| | - Naoki Yamanaka
- Department of Entomology, Institute for Integrative Genome Biology, University of California, Riverside, 900 University Ave., Riverside, CA 92521, USA.
| |
Collapse
|
25
|
Miszczak K, Egger B. Live Cell Imaging of Neural Stem Cells in the Drosophila Larval Brain. Methods Mol Biol 2020; 2047:153-160. [PMID: 31552654 DOI: 10.1007/978-1-4939-9732-9_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Live cell imaging gives valuable insights into the dynamic biological processes within and between cells. An important aspect of live cell imaging is to keep the cells under best physiological condition and to prevent abnormal cellular behavior, which might be caused by phototoxicity during microscopy. In this chapter we describe a protocol to visualize division patterns of neural stem cells in live whole mount brains of Drosophila larvae. We also present a newly developed live cell chamber that allows us to control the environmental air during live cell imaging. The protocol can be adapted to look at a wide range of cellular and tissue behavior in the Drosophila model system.
Collapse
Affiliation(s)
- Karolina Miszczak
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
26
|
Regulation of Proneural Wave Propagation Through a Combination of Notch-Mediated Lateral Inhibition and EGF-Mediated Reaction Diffusion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1218:77-91. [PMID: 32060872 DOI: 10.1007/978-3-030-34436-8_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Notch-mediated lateral inhibition regulates binary cell fate choice, resulting in salt-and-pepper pattern formation during various biological processes. In many cases, Notch signaling acts together with other signaling systems. However, it is not clear what happens when Notch signaling is combined with other signaling systems. Mathematical modeling and the use of a simple biological model system will be essential to address this uncertainty. A wave of differentiation in the Drosophila visual center, the "proneural wave," accompanies the activity of the Notch and EGF signaling pathways. Although all of the Notch signaling components required for lateral inhibition are involved in the proneural wave, no salt-and-pepper pattern is found during the progression of the proneural wave. Instead, Notch is activated along the wave front and regulates proneural wave progression. How does Notch signaling control wave propagation without forming a salt-and-pepper pattern? A mathematical model of the proneural wave, based on biological evidence, has demonstrated that Notch-mediated lateral inhibition is implemented within the proneural wave and that the diffusible action of EGF cancels salt-and-pepper pattern formation. The results from numerical simulation have been confirmed by genetic experiments in vivo and suggest that the combination of Notch-mediated lateral inhibition and EGF-mediated reaction diffusion enables a novel function of Notch signaling that regulates propagation of the proneural wave. Similar mechanisms may play important roles in diverse biological processes found in animal development and cancer pathogenesis.
Collapse
|
27
|
Zhou Y, Yang Y, Huang Y, Wang H, Wang S, Luo H. Broad Promotes Neuroepithelial Stem Cell Differentiation in the Drosophila Optic Lobe. Genetics 2019; 213:941-951. [PMID: 31530575 PMCID: PMC6827381 DOI: 10.1534/genetics.119.302421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/08/2019] [Indexed: 11/18/2022] Open
Abstract
Brain development requires the generation of the right number, and type, of neurons and glial cells at the right time. The Drosophila optic lobe, like mammalian brains, develops from simple neuroepithelia; they first divide symmetrically to expand the progenitor pool and then differentiate into neuroblasts, which divide asymmetrically to generate neurons and glial cells. Here, we investigate the mechanisms that control neuroepithelial growth and differentiation in the optic lobe. We find that the Broad/Tramtrack/Bric a brac-zinc finger protein Broad, which is dynamically expressed in the optic lobe neuroepithelia, promotes the transition of neuroepithelial cells to medulla neuroblasts. Loss of Broad function causes neuroepithelial cells to remain highly proliferative and delays neuroepithelial cell differentiation into neuroblasts, which leads to defective lamina and medulla. Conversely, Broad overexpression induces neuroepithelial cells to prematurely transform into medulla neuroblasts. We find that the ecdysone receptor is required for neuroepithelial maintenance and growth, and that Broad expression in neuroepithelial cells is repressed by the ecdysone receptor. Our studies identify Broad as an important cell-intrinsic transcription factor that promotes the neuroepithelial-cell-to-neuroblast transition.
Collapse
Affiliation(s)
- Yanna Zhou
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yuqin Yang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yanyi Huang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Hui Wang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Shengyu Wang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Hong Luo
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| |
Collapse
|
28
|
Plazaola-Sasieta H, Zhu Q, Gaitán-Peñas H, Rios M, Estévez R, Morey M. Drosophila ClC-a is required in glia of the stem cell niche for proper neurogenesis and wiring of neural circuits. Glia 2019; 67:2374-2398. [PMID: 31479171 PMCID: PMC6851788 DOI: 10.1002/glia.23691] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 01/01/2023]
Abstract
Glial cells form part of the neural stem cell niche and express a wide variety of ion channels; however, the contribution of these channels to nervous system development is poorly understood. We explored the function of the Drosophila ClC‐a chloride channel, since its mammalian ortholog CLCN2 is expressed in glial cells, and defective channel function results in leukodystrophies, which in humans are accompanied by cognitive impairment. We found that ClC‐a was expressed in the niche in cortex glia, which are closely associated with neurogenic tissues. Characterization of loss‐of‐function ClC‐a mutants revealed that these animals had smaller brains and widespread wiring defects. We showed that ClC‐a is required in cortex glia for neurogenesis in neuroepithelia and neuroblasts, and identified defects in a neuroblast lineage that generates guidepost glial cells essential for photoreceptor axon guidance. We propose that glia‐mediated ionic homeostasis could nonautonomously affect neurogenesis, and consequently, the correct assembly of neural circuits.
Collapse
Affiliation(s)
- Haritz Plazaola-Sasieta
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Qi Zhu
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Héctor Gaitán-Peñas
- Departament de Ciencies Fisiològiques, Genes, Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Martín Rios
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Raúl Estévez
- Departament de Ciencies Fisiològiques, Genes, Disease and Therapy Program IDIBELL-Institute of Neurosciences, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Morey
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Programa de Biologia Integrativa, Barcelona, Spain
| |
Collapse
|
29
|
Hakes AE, Brand AH. Neural stem cell dynamics: the development of brain tumours. Curr Opin Cell Biol 2019; 60:131-138. [PMID: 31330360 DOI: 10.1016/j.ceb.2019.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023]
Abstract
Determining the premalignant lesions that develop into malignant tumours remains a daunting task. Brain tumours are frequently characterised by a block in differentiation, implying that normal developmental pathways become hijacked during tumourigenesis. However, the heterogeneity of stem cells and their progenitors in the brain suggests there are many potential routes to tumour initiation. Studies in Drosophila melanogaster have enhanced our understanding of the tumourigenic potential of distinct cell types in the brain. Here we review recent studies that have improved our knowledge of neural stem cell behaviour during development and in brain tumour models.
Collapse
Affiliation(s)
- Anna E Hakes
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Andrea H Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK.
| |
Collapse
|
30
|
Contreras EG, Sierralta J, Oliva C. Novel Strategies for the Generation of Neuronal Diversity: Lessons From the Fly Visual System. Front Mol Neurosci 2019; 12:140. [PMID: 31213980 PMCID: PMC6554424 DOI: 10.3389/fnmol.2019.00140] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022] Open
Abstract
Among all organs of an adult animal, the central nervous system stands out because of its vast complexity and morphological diversity. During early development, the entire central nervous system develops from an apparently homogenous group of progenitors that differentiate into all neural cell types. Therefore, understanding the molecular and genetic mechanisms that give rise to the cellular and anatomical diversity of the brain is a key goal of the developmental neurobiology field. With this aim in mind, the development of the central nervous system of model organisms has been extensively studied. From more than a century, the mechanisms of neurogenesis have been studied in the fruit fly Drosophila melanogaster. The visual system comprises one of the major structures of the Drosophila brain. The visual information is collected by the eye-retina photoreceptors and then processed by the four optic lobe ganglia: the lamina, medulla, lobula and lobula plate. The molecular mechanisms that originate neuronal diversity in the optic lobe have been unveiled in the past decade. In this article, we describe the early development and differentiation of the lobula plate ganglion, from the formation of the optic placode and the inner proliferation center to the specification of motion detection neurons. We focused specifically on how the precise combination of signaling pathways and cell-specific transcription factors patterns the pool of neural stem cells that generates the different neurons of the motion detection system.
Collapse
Affiliation(s)
- Esteban G Contreras
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Jimena Sierralta
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Carlos Oliva
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
31
|
He L, Binari R, Huang J, Falo-Sanjuan J, Perrimon N. In vivo study of gene expression with an enhanced dual-color fluorescent transcriptional timer. eLife 2019; 8:46181. [PMID: 31140975 PMCID: PMC6660218 DOI: 10.7554/elife.46181] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/28/2019] [Indexed: 12/28/2022] Open
Abstract
Fluorescent transcriptional reporters are widely used as signaling reporters and biomarkers to monitor pathway activities and determine cell type identities. However, a large amount of dynamic information is lost due to the long half-life of the fluorescent proteins. To better detect dynamics, fluorescent transcriptional reporters can be destabilized to shorten their half-lives. However, applications of this approach in vivo are limited due to significant reduction of signal intensities. To overcome this limitation, we enhanced translation of a destabilized fluorescent protein and demonstrate the advantages of this approach by characterizing spatio-temporal changes of transcriptional activities in Drosophila. In addition, by combining a fast-folding destabilized fluorescent protein and a slow-folding long-lived fluorescent protein, we generated a dual-color transcriptional timer that provides spatio-temporal information about signaling pathway activities. Finally, we demonstrate the use of this transcriptional timer to identify new genes with dynamic expression patterns.
Collapse
Affiliation(s)
- Li He
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Richard Binari
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| | - Jiuhong Huang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing, China
| | | | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| |
Collapse
|
32
|
Panov AA. Proliferation and Differentiation: Two Sequential Stages of Proliferative Center Activity in Embryonic Mushroom Bodies of Three Orthopterans, Gryllus bimaculatus Deg., Acheta domesticus L., and Schistocerca gregaria Forsk. (Insecta: Orthoptera). BIOL BULL+ 2019. [DOI: 10.1134/s1062359019020110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Jörg DJ, Caygill EE, Hakes AE, Contreras EG, Brand AH, Simons BD. The proneural wave in the Drosophila optic lobe is driven by an excitable reaction-diffusion mechanism. eLife 2019; 8:e40919. [PMID: 30794154 PMCID: PMC6386523 DOI: 10.7554/elife.40919] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/08/2019] [Indexed: 11/13/2022] Open
Abstract
In living organisms, self-organised waves of signalling activity propagate spatiotemporal information within tissues. During the development of the largest component of the visual processing centre of the Drosophila brain, a travelling wave of proneural gene expression initiates neurogenesis in the larval optic lobe primordium and drives the sequential transition of neuroepithelial cells into neuroblasts. Here, we propose that this 'proneural wave' is driven by an excitable reaction-diffusion system involving epidermal growth factor receptor (EGFR) signalling interacting with the proneural gene l'sc. Within this framework, a propagating transition zone emerges from molecular feedback and diffusion. Ectopic activation of EGFR signalling in clones within the neuroepithelium demonstrates that a transition wave can be excited anywhere in the tissue by inducing signalling activity, consistent with a key prediction of the model. Our model illuminates the physical and molecular underpinnings of proneural wave progression and suggests a generic mechanism for regulating the sequential differentiation of tissues.
Collapse
Affiliation(s)
- David J Jörg
- Cavendish Laboratory, Department of PhysicsUniversity of CambridgeCambridgeUnited Kingdom
- The Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Elizabeth E Caygill
- The Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| | - Anna E Hakes
- The Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUnited Kingdom
| | - Esteban G Contreras
- The Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Andrea H Brand
- The Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Benjamin D Simons
- Cavendish Laboratory, Department of PhysicsUniversity of CambridgeCambridgeUnited Kingdom
- The Wellcome Trust/Cancer Research UK Gurdon InstituteUniversity of CambridgeCambridgeUnited Kingdom
- The Wellcome Trust/Medical Research Council Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
34
|
Herrera SC, Bach EA. JAK/STAT signaling in stem cells and regeneration: from Drosophila to vertebrates. Development 2019; 146:dev167643. [PMID: 30696713 PMCID: PMC6361132 DOI: 10.1242/dev.167643] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022]
Abstract
The JAK/STAT pathway is a conserved metazoan signaling system that transduces cues from extracellular cytokines into transcriptional changes in the nucleus. JAK/STAT signaling is best known for its roles in immunity. However, recent work has demonstrated that it also regulates critical homeostatic processes in germline and somatic stem cells, as well as regenerative processes in several tissues, including the gonad, intestine and appendages. Here, we provide an overview of JAK/STAT signaling in stem cells and regeneration, focusing on Drosophila and highlighting JAK/STAT pathway functions in proliferation, survival and cell competition that are conserved between Drosophila and vertebrates.
Collapse
Affiliation(s)
- Salvador C Herrera
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Erika A Bach
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
- Helen L. and Martin S. Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
35
|
Ramon-Cañellas P, Peterson HP, Morante J. From Early to Late Neurogenesis: Neural Progenitors and the Glial Niche from a Fly's Point of View. Neuroscience 2018; 399:39-52. [PMID: 30578972 DOI: 10.1016/j.neuroscience.2018.12.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 12/06/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022]
Abstract
Drosophila melanogaster is an important model organism used to study the brain development of organisms ranging from insects to mammals. The central nervous system in fruit flies is formed primarily in two waves of neurogenesis, one of which occurs in the embryo and one of which occurs during larval stages. In order to understand neurogenesis, it is important to research the behavior of progenitor cells that give rise to the neural networks which make up the adult nervous system. This behavior has been shown to be influenced by different factors including interactions with other cells within the progenitor niche, or local tissue microenvironment. Glial cells form a crucial part of this niche and play an active role in the development of the brain. Although in the early years of neuroscience it was believed that glia were simply scaffolding for neurons and passive components of the nervous system, their importance is nowadays recognized. Recent discoveries in progenitors and niche cells have led to new understandings of how the developing brain shapes its diverse regions. In this review, we attempt to summarize the distinct neural progenitors and glia in the Drosophila melanogaster central nervous system, from embryo to late larval stages, and make note of homologous features in mammals. We also outline the recent advances in this field in order to define the impact that glial cells have on progenitor cell niches, and we finally emphasize the importance of communication between glia and progenitor cells for proper brain formation.
Collapse
Affiliation(s)
- Pol Ramon-Cañellas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Hannah Payette Peterson
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| | - Javier Morante
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), and Universidad Miguel Hernández (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain.
| |
Collapse
|
36
|
Contreras EG, Egger B, Gold KS, Brand AH. Dynamic Notch signalling regulates neural stem cell state progression in the Drosophila optic lobe. Neural Dev 2018; 13:25. [PMID: 30466475 PMCID: PMC6251220 DOI: 10.1186/s13064-018-0123-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/13/2018] [Indexed: 11/10/2022] Open
Abstract
Background Neural stem cells generate all of the neurons and glial cells in the central nervous system, both during development and in the adult to maintain homeostasis. In the Drosophila optic lobe, neuroepithelial cells progress through two transient progenitor states, PI and PII, before transforming into neuroblasts. Here we analyse the role of Notch signalling in the transition from neuroepithelial cells to neuroblasts. Results We observed dynamic regulation of Notch signalling: strong activity in PI progenitors, low signalling in PII progenitors, and increased activity after neuroblast transformation. Ectopic expression of the Notch ligand Delta induced the formation of ectopic PI progenitors. Interestingly, we show that the E3 ubiquitin ligase, Neuralized, regulates Delta levels and Notch signalling activity at the transition zone. We demonstrate that the proneural transcription factor, Lethal of scute, is essential to induce expression of Neuralized and promote the transition from the PI progenitor to the PII progenitor state. Conclusions Our results show dynamic regulation of Notch signalling activity in the transition from neuroepithelial cells to neuroblasts. We propose a model in which Lethal of scute activates Notch signalling in a non-cell autonomous manner by regulating the expression of Neuralized, thereby promoting the progression between different neural stem cell states. Electronic supplementary material The online version of this article (10.1186/s13064-018-0123-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Esteban G Contreras
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Boris Egger
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK.,Present Address: Department of Biology, Zoology, University of Fribourg, Chemin du Musée 10, CH-1700, Fribourg, Switzerland
| | - Katrina S Gold
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | - Andrea H Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK.
| |
Collapse
|
37
|
The Scalloped and Nerfin-1 Transcription Factors Cooperate to Maintain Neuronal Cell Fate. Cell Rep 2018; 25:1561-1576.e7. [DOI: 10.1016/j.celrep.2018.10.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/14/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023] Open
|
38
|
Hakes AE, Otsuki L, Brand AH. A newly discovered neural stem cell population is generated by the optic lobe neuroepithelium during embryogenesis in Drosophila melanogaster. Development 2018; 145:145/18/dev166207. [PMID: 30254066 PMCID: PMC6176933 DOI: 10.1242/dev.166207] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022]
Abstract
Neural stem cells must balance symmetric and asymmetric cell divisions to generate a functioning brain of the correct size. In both the developing Drosophila visual system and mammalian cerebral cortex, symmetrically dividing neuroepithelial cells transform gradually into asymmetrically dividing progenitors that generate neurons and glia. As a result, it has been widely accepted that stem cells in these tissues switch from a symmetric, expansive phase of cell divisions to a later neurogenic phase of cell divisions. In the Drosophila optic lobe, this switch is thought to occur during larval development. However, we have found that neuroepithelial cells start to produce neuroblasts during embryonic development, demonstrating a much earlier role for neuroblasts in the developing visual system. These neuroblasts undergo neurogenic divisions, enter quiescence and are retained post-embryonically, together with neuroepithelial cells. Later in development, neuroepithelial cells undergo further cell divisions before transforming into larval neuroblasts. Our results demonstrate that the optic lobe neuroepithelium gives rise to neurons and glia over 60 h earlier than was thought previously. Highlighted Article: Two types of neural stem cell, with distinct proliferation modes, act side by side to generate the Drosophila optic lobe, challenging current dogma that they act sequentially.
Collapse
Affiliation(s)
- Anna E Hakes
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Leo Otsuki
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Andrea H Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| |
Collapse
|
39
|
Sato M, Yasugi T, Trush O. Temporal patterning of neurogenesis and neural wiring in the fly visual system. Neurosci Res 2018; 138:49-58. [PMID: 30227165 DOI: 10.1016/j.neures.2018.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/13/2018] [Accepted: 08/13/2018] [Indexed: 12/21/2022]
Abstract
During neural development, a wide variety of neurons are produced in a highly coordinated manner and form complex and highly coordinated neural circuits. Temporal patterning of neuron type specification plays very important roles in orchestrating the production and wiring of neurons. The fly visual system, which is composed of the retina and the optic lobe of the brain, is an outstanding model system to study temporal patterning and wiring of the nervous system. All of the components of the fly visual system are topographically connected, and each ommatidial unit in the retina corresponds to a columnar unit in the optic lobe. In the retina, the wave of differentiation follows the morphogenetic furrow, which progresses in a posterior-to-anterior direction. At the same time, differentiation of the optic lobe also accompanies the wave of differentiation or temporally coordinated neurogenesis. Thus, temporal patterning plays important roles in establishing topographic connections throughout the fly visual system. In this article, we review how neuronal differentiation and connectivity are orchestrated in the fly visual system by temporal patterning mechanisms.
Collapse
Affiliation(s)
- Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Japan; Lab of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Japan.
| | - Tetsuo Yasugi
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Japan
| | - Olena Trush
- Lab of Developmental Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Japan
| |
Collapse
|
40
|
Contreras EG, Palominos T, Glavic Á, Brand AH, Sierralta J, Oliva C. The transcription factor SoxD controls neuronal guidance in the Drosophila visual system. Sci Rep 2018; 8:13332. [PMID: 30190506 PMCID: PMC6127262 DOI: 10.1038/s41598-018-31654-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 08/23/2018] [Indexed: 01/21/2023] Open
Abstract
Precise control of neurite guidance during development is essential to ensure proper formation of neuronal networks and correct function of the central nervous system (CNS). How neuronal projections find their targets to generate appropriate synapses is not entirely understood. Although transcription factors are key molecules during neurogenesis, we do not know their entire function during the formation of networks in the CNS. Here, we used the Drosophila melanogaster optic lobe as a model for understanding neurite guidance during development. We assessed the function of Sox102F/SoxD, the unique Drosophila orthologue of the vertebrate SoxD family of transcription factors. SoxD is expressed in immature and mature neurons in the larval and adult lobula plate ganglia (one of the optic lobe neuropils), but is absent from glial cells, neural stem cells and progenitors of the lobula plate. SoxD RNAi knockdown in all neurons results in a reduction of the lobula plate neuropil, without affecting neuronal fate. This morphological defect is associated with an impaired optomotor response of adult flies. Moreover, knocking down SoxD only in T4/T5 neuronal types, which control motion vision, affects proper neurite guidance into the medulla and lobula. Our findings suggest that SoxD regulates neurite guidance, without affecting neuronal fate.
Collapse
Affiliation(s)
- Esteban G Contreras
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Independencia, 1027, Santiago, Chile.,Center for Genome Regulation, Faculty of Sciences, Universidad de Chile, Las Palmeras, 3425, Nuñoa, Santiago, Chile
| | - Tomás Palominos
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Av Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Álvaro Glavic
- Center for Genome Regulation, Faculty of Sciences, Universidad de Chile, Las Palmeras, 3425, Nuñoa, Santiago, Chile
| | - Andrea H Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, United Kingdom
| | - Jimena Sierralta
- Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Independencia, 1027, Santiago, Chile
| | - Carlos Oliva
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Av Libertador Bernardo O'Higgins 340, Santiago, Chile.
| |
Collapse
|
41
|
JAK/STAT guarantees robust neural stem cell differentiation by shutting off biological noise. Sci Rep 2018; 8:12484. [PMID: 30127451 PMCID: PMC6102247 DOI: 10.1038/s41598-018-30929-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 08/08/2018] [Indexed: 01/10/2023] Open
Abstract
Organismal development is precisely regulated by a sequence of gene functions even in the presence of biological noise. However, it is difficult to evaluate the effect of noise in vivo, and the mechanisms by which noise is filtered during development are largely unknown. To identify the noise-canceling mechanism, we used the fly visual system, in which the timing of differentiation of neural stem cells is spatio-temporally ordered. Our mathematical model predicts that JAK/STAT signaling contributes to noise canceling to guarantee the robust progression of the differentiation wave in silico. We further demonstrate that the suppression of JAK/STAT signaling causes stochastic and ectopic neural stem cell differentiation in vivo, suggesting an evolutionarily conserved function of JAK/STAT to regulate the robustness of stem cell differentiation.
Collapse
|
42
|
Apitz H, Salecker I. Spatio-temporal relays control layer identity of direction-selective neuron subtypes in Drosophila. Nat Commun 2018; 9:2295. [PMID: 29895891 PMCID: PMC5997761 DOI: 10.1038/s41467-018-04592-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/04/2018] [Indexed: 11/09/2022] Open
Abstract
Visual motion detection in sighted animals is essential to guide behavioral actions ensuring their survival. In Drosophila, motion direction is first detected by T4/T5 neurons. Their axons innervate one of the four lobula plate layers. How T4/T5 neurons with layer-specific representation of motion-direction preferences are specified during development is unknown. We show that diffusible Wingless (Wg) between adjacent neuroepithelia induces its own expression to form secondary signaling centers. These activate Decapentaplegic (Dpp) signaling in adjacent lateral tertiary neuroepithelial domains dedicated to producing layer 3/4-specific T4/T5 neurons. T4/T5 neurons derived from the core domain devoid of Dpp signaling adopt the default layer 1/2 fate. Dpp signaling induces the expression of the T-box transcription factor Optomotor-blind (Omb), serving as a relay to postmitotic neurons. Omb-mediated repression of Dachshund transforms layer 1/2- into layer 3/4-specific neurons. Hence, spatio-temporal relay mechanisms, bridging the distances between neuroepithelial domains and their postmitotic progeny, implement T4/T5 neuron-subtype identity.
Collapse
Affiliation(s)
- Holger Apitz
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK
| | - Iris Salecker
- The Francis Crick Institute, Visual Circuit Assembly Laboratory, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
43
|
Abstract
A small pool of neural progenitors generates the vast diversity of cell types in the CNS. Spatial patterning specifies progenitor identity, followed by temporal patterning within progenitor lineages to expand neural diversity. Recent work has shown that in Drosophila, all neural progenitors (neuroblasts) sequentially express temporal transcription factors (TTFs) that generate molecular and cellular diversity. Embryonic neuroblasts use a lineage-intrinsic cascade of five TTFs that switch nearly every neuroblast cell division; larval optic lobe neuroblasts also use a rapid cascade of five TTFs, but the factors are completely different. In contrast, larval central brain neuroblasts undergo a major molecular transition midway through larval life, and this transition is regulated by a lineage-extrinsic cue (ecdysone hormone signaling). Overall, every neuroblast lineage uses a TTF cascade to generate diversity, illustrating the widespread importance of temporal patterning.
Collapse
Affiliation(s)
- Chris Q Doe
- Institute of Neuroscience, Institute of Molecular Biology, and Howard Hughes Medical Institute (HHMI), University of Oregon, Eugene, Oregon 97403;
| |
Collapse
|
44
|
Caygill EE, Brand AH. miR-7 Buffers Differentiation in the Developing Drosophila Visual System. Cell Rep 2018; 20:1255-1261. [PMID: 28793250 PMCID: PMC5561169 DOI: 10.1016/j.celrep.2017.07.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/23/2017] [Accepted: 07/18/2017] [Indexed: 01/21/2023] Open
Abstract
The 40,000 neurons of the medulla, the largest visual processing center of the Drosophila brain, derive from a sheet of neuroepithelial cells. During larval development, a wave of differentiation sweeps across the neuroepithelium, converting neuroepithelial cells into neuroblasts that sequentially express transcription factors specifying different neuronal cell fates. The switch from neuroepithelial cells to neuroblasts is controlled by a complex gene regulatory network and is marked by the expression of the proneural gene l’sc. We discovered that microRNA miR-7 is expressed at the transition between neuroepithelial cells and neuroblasts. We showed that miR-7 promotes neuroepithelial cell-to-neuroblast transition by targeting downstream Notch effectors to limit Notch signaling. miR-7 acts as a buffer to ensure that a precise and stereotypical pattern of transition is maintained, even under conditions of environmental stress, echoing the role that miR-7 plays in the eye imaginal disc. This common mechanism reflects the importance of robust visual system development. miR-7 promotes neuroblast formation during optic lobe development miR-7 targets the Notch pathway miR-7 buffers the effects of environmental stress Without miR-7, timely neuroblast production is disrupted
Collapse
Affiliation(s)
- Elizabeth E Caygill
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Andrea H Brand
- The Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK.
| |
Collapse
|
45
|
Shaikh MN, Tejedor FJ. Mnb/Dyrk1A orchestrates a transcriptional network at the transition from self-renewing neurogenic progenitors to postmitotic neuronal precursors. J Neurogenet 2018; 32:37-50. [PMID: 29495936 DOI: 10.1080/01677063.2018.1438427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The Down syndrome and microcephaly related gene Mnb/Dyrk1A encodes an evolutionary conserved protein kinase subfamily that plays important roles in neurodevelopment. minibrain (mnb) mutants of Drosophila melanogaster (Dm) exhibit reduced adult brains due to neuronal deficits generated during larval development. These deficits are the consequence of the apoptotic cell death of numerous neuronal precursors that fail to properly exit the cell cycle and differentiate. We have recently found that in both the Dm larval brain and the embryonic vertebrate central nervous system (CNS), a transient expression of Mnb/Dyrk1A promotes the cell cycle exit of newborn neuronal precursors by upregulating the expression of the cyclin-dependent kinase inhibitor p27kip1 (called Dacapo in Dm). In the larval brain, Mnb performs this action by regulating the expression of three transcription factors, Asense (Ase), Deadpan (Dpn) and Prospero (Pros), which are key regulators of the self-renewal, proliferation, and terminal differentiation of neural progenitor cells. We have here studied in detail the cellular/temporal expression pattern of Ase, Dpn, Pros and Mnb, and have analyzed possible regulatory effects among them at the transitions from neurogenic progenitors to postmitotic neuronal precursors in the Dm larval brain. The emerging picture of this analysis reveals an intricate regulatory network in which Mnb appears to play a pivotal role helping to delineate the dynamics of the expression patterns of Ase, Dpn and Pros, as well as their specific functions in the aforementioned transitions. Our results also show that Ase, Dpn and Pros perform several cross-regulatory actions and contribute to shape the precise cellular/temporal expression pattern of Mnb. We propose that Mnb/Dyrk1A plays a central role in CNS neurogenesis by integrating molecular mechanisms that regulate progenitor self-renewal, cell cycle progression and neuronal differentiation.
Collapse
Affiliation(s)
- Mirja N Shaikh
- a Instituto de Neurociencias , CSIC and Universidad Miguel Hernandez , Alicante , Spain
| | - Francisco J Tejedor
- a Instituto de Neurociencias , CSIC and Universidad Miguel Hernandez , Alicante , Spain
| |
Collapse
|
46
|
Hara Y, Sudo T, Togane Y, Akagawa H, Tsujimura H. Cell death in neural precursor cells and neurons before neurite formation prevents the emergence of abnormal neural structures in the Drosophila optic lobe. Dev Biol 2018; 436:28-41. [PMID: 29447906 DOI: 10.1016/j.ydbio.2018.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/15/2018] [Accepted: 02/07/2018] [Indexed: 11/30/2022]
Abstract
Programmed cell death is a conserved strategy for neural development both in vertebrates and invertebrates and is recognized at various developmental stages in the brain from neurogenesis to adulthood. To understand the development of the central nervous system, it is essential to reveal not only molecular mechanisms but also the role of neural cell death (Pinto-Teixeira et al., 2016). To understand the role of cell death in neural development, we investigated the effect of inhibition of cell death on optic lobe development. Our data demonstrate that, in the optic lobe of Drosophila, cell death occurs in neural precursor cells and neurons before neurite formation and functions to prevent various developmental abnormalities. When neuronal cell death was inhibited by an effector caspase inhibitor, p35, multiple abnormal neuropil structures arose during optic lobe development-e.g., enlarged or fused neuropils, misrouted neurons and abnormal neurite lumps. Inhibition of cell death also induced morphogenetic defects in the lamina and medulla development-e.g., failures in the separation of the lamina and medulla cortices and the medulla rotation. These defects were reproduced in the mutant of an initiator caspase, dronc. If cell death was a mechanism for removing the abnormal neuropil structures, we would also expect to observe them in mutants defective for corpse clearance. However, they were not observed in these mutants. When dead cell-membranes were visualized with Apoliner, they were observed only in cortices and not in neuropils. These results suggest that the cell death occurs before mature neurite formation. Moreover, we found that inhibition of cell death induced ectopic neuroepithelial cells, neuroblasts and ganglion mother cells in late pupal stages, at sites where the outer and inner proliferation centers were located at earlier developmental stages. Caspase-3 activation was observed in the neuroepithelial cells and neuroblasts in the proliferation centers. These results indicate that cell death is required for elimination of the precursor cells composing the proliferation centers. This study substantiates an essential role of early neural cell death for ensuring normal development of the central nervous system.
Collapse
Affiliation(s)
- Yusuke Hara
- Developmental Biology, Tokyo University of Agriculture and Technology, Japan; Graduate School of Life Sciences, Tohoku University, Japan.
| | - Tatsuya Sudo
- Developmental Biology, Tokyo University of Agriculture and Technology, Japan
| | - Yu Togane
- Developmental Biology, Tokyo University of Agriculture and Technology, Japan
| | - Hiromi Akagawa
- Developmental Biology, Tokyo University of Agriculture and Technology, Japan
| | - Hidenobu Tsujimura
- Developmental Biology, Tokyo University of Agriculture and Technology, Japan
| |
Collapse
|
47
|
Dillard C, Narbonne-Reveau K, Foppolo S, Lanet E, Maurange C. Two distinct mechanisms silence chinmo in Drosophila neuroblasts and neuroepithelial cells to limit their self-renewal. Development 2018; 145:dev.154534. [PMID: 29361557 DOI: 10.1242/dev.154534] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/18/2017] [Indexed: 02/03/2023]
Abstract
Whether common principles regulate the self-renewing potential of neural stem cells (NSCs) throughout the developing central nervous system is still unclear. In the Drosophila ventral nerve cord and central brain, asymmetrically dividing NSCs, called neuroblasts (NBs), progress through a series of sequentially expressed transcription factors that limits self-renewal by silencing a genetic module involving the transcription factor Chinmo. Here, we find that Chinmo also promotes neuroepithelium growth in the optic lobe during early larval stages by boosting symmetric self-renewing divisions while preventing differentiation. Neuroepithelium differentiation in late larvae requires the transcriptional silencing of chinmo by ecdysone, the main steroid hormone, therefore allowing coordination of neural stem cell self-renewal with organismal growth. In contrast, chinmo silencing in NBs is post-transcriptional and does not require ecdysone. Thus, during Drosophila development, humoral cues or tissue-intrinsic temporal specification programs respectively limit self-renewal in different types of neural progenitors through the transcriptional and post-transcriptional regulation of the same transcription factor.
Collapse
Affiliation(s)
- Caroline Dillard
- Aix Marseille Univ, CNRS, IBDM, Campus de Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Karine Narbonne-Reveau
- Aix Marseille Univ, CNRS, IBDM, Campus de Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Sophie Foppolo
- Aix Marseille Univ, CNRS, IBDM, Campus de Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Elodie Lanet
- Aix Marseille Univ, CNRS, IBDM, Campus de Luminy Case 907, 13288 Marseille Cedex 9, France
| | - Cédric Maurange
- Aix Marseille Univ, CNRS, IBDM, Campus de Luminy Case 907, 13288 Marseille Cedex 9, France
| |
Collapse
|
48
|
A Conserved Developmental Mechanism Builds Complex Visual Systems in Insects and Vertebrates. Curr Biol 2017; 26:R1001-R1009. [PMID: 27780043 DOI: 10.1016/j.cub.2016.08.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The visual systems of vertebrates and many other bilaterian clades consist of complex neural structures guiding a wide spectrum of behaviors. Homologies at the level of cell types and even discrete neural circuits have been proposed, but many questions of how the architecture of visual neuropils evolved among different phyla remain open. In this review we argue that the profound conservation of genetic and developmental steps generating the eye and its target neuropils in fish and fruit flies supports a homology between some core elements of bilaterian visual circuitries. Fish retina and tectum, and fly optic lobe, develop from a partitioned, unidirectionally proliferating neurectodermal domain that combines slowly dividing neuroepithelial stem cells and rapidly amplifying progenitors with shared genetic signatures to generate large numbers and different types of neurons in a temporally ordered way. This peculiar 'conveyor belt neurogenesis' could play an essential role in generating the topographically ordered circuitry of the visual system.
Collapse
|
49
|
Rujano MA, Cannata Serio M, Panasyuk G, Péanne R, Reunert J, Rymen D, Hauser V, Park JH, Freisinger P, Souche E, Guida MC, Maier EM, Wada Y, Jäger S, Krogan NJ, Kretz O, Nobre S, Garcia P, Quelhas D, Bird TD, Raskind WH, Schwake M, Duvet S, Foulquier F, Matthijs G, Marquardt T, Simons M. Mutations in the X-linked ATP6AP2 cause a glycosylation disorder with autophagic defects. J Exp Med 2017; 214:3707-3729. [PMID: 29127204 PMCID: PMC5716037 DOI: 10.1084/jem.20170453] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 08/01/2017] [Accepted: 09/22/2017] [Indexed: 12/25/2022] Open
Abstract
Rujano et al. report mutations in ATP6AP2 leading to liver disease, immunodeficiency, and psychomotor impairment. ATP6AP2 deficiency impairs the assembly and function of the V-ATPase proton pump, causing defects in protein glycosylation and autophagy. The biogenesis of the multi-subunit vacuolar-type H+-ATPase (V-ATPase) is initiated in the endoplasmic reticulum with the assembly of the proton pore V0, which is controlled by a group of assembly factors. Here, we identify two hemizygous missense mutations in the extracellular domain of the accessory V-ATPase subunit ATP6AP2 (also known as the [pro]renin receptor) responsible for a glycosylation disorder with liver disease, immunodeficiency, cutis laxa, and psychomotor impairment. We show that ATP6AP2 deficiency in the mouse liver caused hypoglycosylation of serum proteins and autophagy defects. The introduction of one of the missense mutations into Drosophila led to reduced survival and altered lipid metabolism. We further demonstrate that in the liver-like fat body, the autophagic dysregulation was associated with defects in lysosomal acidification and mammalian target of rapamycin (mTOR) signaling. Finally, both ATP6AP2 mutations impaired protein stability and the interaction with ATP6AP1, a member of the V0 assembly complex. Collectively, our data suggest that the missense mutations in ATP6AP2 lead to impaired V-ATPase assembly and subsequent defects in glycosylation and autophagy.
Collapse
Affiliation(s)
- Maria A Rujano
- Laboratory of Epithelial Biology and Disease, Imagine Institute, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Magda Cannata Serio
- Laboratory of Epithelial Biology and Disease, Imagine Institute, Paris, France.,Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Ganna Panasyuk
- Institut Necker-Enfants Malades, Paris, France.,Institut National de la Santé et de la Recherche Medicale U1151/Centre National de la Recherche Scientifique UMR 8253, Paris, France
| | - Romain Péanne
- University of Leuven (KU Leuven), Center for Human Genetics, Leuven, Belgium
| | - Janine Reunert
- Universitätsklinikum Münster, Klinik für Kinder- und Jugendmedizin, Münster, Germany
| | - Daisy Rymen
- University of Leuven (KU Leuven), Center for Human Genetics, Leuven, Belgium
| | - Virginie Hauser
- Laboratory of Epithelial Biology and Disease, Imagine Institute, Paris, France.,Institut National de la Santé et de la Recherche Medicale U1151/Centre National de la Recherche Scientifique UMR 8253, Paris, France
| | - Julien H Park
- Universitätsklinikum Münster, Klinik für Kinder- und Jugendmedizin, Münster, Germany
| | - Peter Freisinger
- Kreiskliniken Reutlingen, Klinik für Kinder- und Jugendmedizin, Klinikum am Steinenberg, Reutlingen, Germany
| | - Erika Souche
- University of Leuven (KU Leuven), Center for Human Genetics, Leuven, Belgium
| | - Maria Clara Guida
- Laboratory of Epithelial Biology and Disease, Imagine Institute, Paris, France.,Institut National de la Santé et de la Recherche Medicale U1151/Centre National de la Recherche Scientifique UMR 8253, Paris, France
| | - Esther M Maier
- Dr. von Haunersches Kinderspital der Universität München, München, Germany
| | - Yoshinao Wada
- Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Stefanie Jäger
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA
| | - Oliver Kretz
- Centre for Biological Signaling Studies BIOSS, University of Freiburg, Freiburg, Germany
| | - Susana Nobre
- Metabolic Reference Center, Coimbra University Hospital Center, Coimbra, Portugal
| | - Paula Garcia
- Metabolic Reference Center, Coimbra University Hospital Center, Coimbra, Portugal
| | - Dulce Quelhas
- Biochemical Genetics Unit, Centro de Genética Médica Doutor Jacinto Magalhães, Centro Hospitalar do Porto, Abel Salazar Institute of Biomedical Sciences, University of Porto, Porto, Portugal
| | - Thomas D Bird
- Department of Neurology, University of Washington, Seattle, WA.,Geriatric Research Center, Veterans Administration Medical Center, Seattle, WA
| | - Wendy H Raskind
- Department of Medicine, University of Washington, Seattle, WA
| | - Michael Schwake
- Faculty of Chemistry/Biochemistry III, University Bielefeld, Bielefeld, Germany
| | - Sandrine Duvet
- Université Lille, Centre National de la Recherche Scientifique UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Francois Foulquier
- Université Lille, Centre National de la Recherche Scientifique UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Gert Matthijs
- University of Leuven (KU Leuven), Center for Human Genetics, Leuven, Belgium
| | - Thorsten Marquardt
- Universitätsklinikum Münster, Klinik für Kinder- und Jugendmedizin, Münster, Germany
| | - Matias Simons
- Laboratory of Epithelial Biology and Disease, Imagine Institute, Paris, France .,Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France
| |
Collapse
|
50
|
Suzuki T, Sato M. Inter-progenitor pool wiring: An evolutionarily conserved strategy that expands neural circuit diversity. Dev Biol 2017; 431:101-110. [PMID: 28958816 DOI: 10.1016/j.ydbio.2017.09.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/15/2017] [Accepted: 09/23/2017] [Indexed: 11/28/2022]
Abstract
Diversification of neuronal types is key to establishing functional variations in neural circuits. The first critical step to generate neuronal diversity is to organize the compartmental domains of developing brains into spatially distinct neural progenitor pools. Neural progenitors in each pool then generate a unique set of diverse neurons through specific spatiotemporal specification processes. In this review article, we focus on an additional mechanism, 'inter-progenitor pool wiring', that further expands the diversity of neural circuits. After diverse types of neurons are generated in one progenitor pool, a fraction of these neurons start migrating toward a remote brain region containing neurons that originate from another progenitor pool. Finally, neurons of different origins are intermingled and eventually form complex but precise neural circuits. The developing cerebral cortex of mammalian brains is one of the best examples of inter-progenitor pool wiring. However, Drosophila visual system development has revealed similar mechanisms in invertebrate brains, suggesting that inter-progenitor pool wiring is an evolutionarily conserved strategy that expands neural circuit diversity. Here, we will discuss how inter-progenitor pool wiring is accomplished in mammalian and fly brain systems.
Collapse
Affiliation(s)
- Takumi Suzuki
- Lab of Developmental Neurobiology, Graduate School of Medical Sciences, Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8640, Japan
| | - Makoto Sato
- Lab of Developmental Neurobiology, Graduate School of Medical Sciences, Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University, 13-1, Takaramachi, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|