1
|
Mujizah EY, Kuwana S, Matsumoto K, Gushiken T, Aoyama N, Ishikawa HO, Sasamura T, Umetsu D, Inaki M, Yamakawa T, Baron M, Matsuno K. Numb Suppresses Notch-Dependent Activation of Enhancer of split during Lateral Inhibition in the Drosophila Embryonic Nervous System. Biomolecules 2024; 14:1062. [PMID: 39334829 PMCID: PMC11429637 DOI: 10.3390/biom14091062] [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: 06/17/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 09/30/2024] Open
Abstract
The role of Drosophila numb in regulating Notch signaling and neurogenesis has been extensively studied, with a particular focus on its effects on the peripheral nervous system (PNS). Previous studies based on a single loss-of-function allele of numb, numb1, showed an antineurogenic effect on the peripheral nervous system (PNS), which revealed that the wild-type numb suppresses Notch signaling. In the current study, we examined whether this phenotype is consistently observed in loss-of-function mutations of numb. Two more numb alleles, numbEY03840 and numbEY03852, were shown to have an antineurogenic phenotype in the PNS. We also found that introducing a wild-type numb genomic fragment into numb1 homozygotes rescued their antineurogenic phenotype. These results demonstrated that loss-of-function mutations of numb universally induce this phenotype. Many components of Notch signaling are encoded by maternal effect genes, but no maternal effect of numb was observed in this study. The antineurogenic phenotype of numb was found to be dependent on the Enhancer of split (E(spl)), a downstream gene of Notch signaling. We found that the combination of E(spl) homozygous and numb1 homozygous suppressed the neurogenic phenotype of the embryonic central nervous system (CNS) associated with the E(spl) mutation. In the E(spl) allele, genes encoding basic helix-loop-helix proteins, such as m5, m6, m7, and m8, remain. Thus, in the E(spl) allele, derepression of Notch activity by numb mutation can rescue the neurogenic phenotype by increasing the expression of the remaining genes in the E(spl) complex. We also uncovered a role for numb in regulating neuronal projections. Our results further support an important role for numb in the suppression of Notch signaling during embryonic nervous system development.
Collapse
Affiliation(s)
- Elzava Yuslimatin Mujizah
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka 560-0043, Japan; (E.Y.M.)
| | - Satoshi Kuwana
- Graduate School of Arts and Sciences, University of Tokyo, Meguro 153-8902, Japan
| | - Kenjiroo Matsumoto
- Institute for Glyco-Core Research, Gifu University, Gifu 501-1193, Japan
| | - Takuma Gushiken
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka 560-0043, Japan; (E.Y.M.)
| | - Naoki Aoyama
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka 560-0043, Japan; (E.Y.M.)
| | | | - Takeshi Sasamura
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka 560-0043, Japan; (E.Y.M.)
| | - Daiki Umetsu
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka 560-0043, Japan; (E.Y.M.)
| | - Mikiko Inaki
- School of Science, Graduate School of Science, University of Hyogo, Ako 678-1297, Japan;
| | - Tomoko Yamakawa
- Department of Industrial Engineering, Chemistry, Bioengineering and Environmental Science Course, National Institute of Technology, Ibaraki College, Hitachinaka 312-8508, Japan
| | - Martin Baron
- School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Kenji Matsuno
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka 560-0043, Japan; (E.Y.M.)
| |
Collapse
|
2
|
Benchorin G, Cho RJ, Li MJ, Molotkova N, Kohwi M. Dan forms condensates in neuroblasts and regulates nuclear architecture and progenitor competence in vivo. Nat Commun 2024; 15:5097. [PMID: 38877037 PMCID: PMC11178893 DOI: 10.1038/s41467-024-49326-6] [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: 01/22/2024] [Accepted: 05/30/2024] [Indexed: 06/16/2024] Open
Abstract
Genome organization is thought to underlie cell type specific gene expression, yet how it is regulated in progenitors to produce cellular diversity is unknown. In Drosophila, a developmentally-timed genome reorganization in neural progenitors terminates competence to produce early-born neurons. These events require downregulation of Distal antenna (Dan), part of the conserved pipsqueak DNA-binding superfamily. Here we find that Dan forms liquid-like condensates with high protein mobility, and whose size and subnuclear distribution are balanced with its DNA-binding. Further, we identify a LARKS domain, a structural motif associated with condensate-forming proteins. Deleting just 13 amino acids from LARKS abrogates Dan's ability to retain the early-born neural fate gene, hunchback, in the neuroblast nuclear interior and maintain competence in vivo. Conversely, domain-swapping with LARKS from known phase-separating proteins rescues Dan's effects on competence. Together, we provide in vivo evidence for condensate formation and the regulation of progenitor nuclear architecture underlying neuronal diversification.
Collapse
Affiliation(s)
- Gillie Benchorin
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Richard Jangwon Cho
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Maggie Jiaqi Li
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Natalia Molotkova
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Minoree Kohwi
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
- Department of Neuroscience, Columbia University, New York, NY, USA.
- Kavli Institute for Brain Science, Columbia University, New York, NY, USA.
| |
Collapse
|
3
|
Sood C, Nahid MA, Branham KR, Pahl M, Doyle SE, Siegrist SE. Delta-dependent Notch activation closes the early neuroblast temporal program to promote lineage progression and neurogenesis termination in Drosophila. eLife 2024; 12:RP88565. [PMID: 38391176 PMCID: PMC10942576 DOI: 10.7554/elife.88565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Neuroblasts in Drosophila divide asymmetrically, sequentially expressing a series of intrinsic factors to generate a diversity of neuron types. These intrinsic factors known as temporal factors dictate timing of neuroblast transitions in response to steroid hormone signaling and specify early versus late temporal fates in neuroblast neuron progeny. After completing their temporal programs, neuroblasts differentiate or die, finalizing both neuron number and type within each neuroblast lineage. From a screen aimed at identifying genes required to terminate neuroblast divisions, we identified Notch and Notch pathway components. When Notch is knocked down, neuroblasts maintain early temporal factor expression longer, delay late temporal factor expression, and continue dividing into adulthood. We find that Delta, expressed in cortex glia, neuroblasts, and after division, their GMC progeny, regulates neuroblast Notch activity. We also find that Delta in neuroblasts is expressed high early, low late, and is controlled by the intrinsic temporal program: early factor Imp promotes Delta, late factors Syp/E93 reduce Delta. Thus, in addition to systemic steroid hormone cues, forward lineage progression is controlled by local cell-cell signaling between neuroblasts and their cortex glia/GMC neighbors: Delta transactivates Notch in neuroblasts bringing the early temporal program and early temporal factor expression to a close.
Collapse
Affiliation(s)
- Chhavi Sood
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | | | - Kendall R Branham
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Matt Pahl
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Susan E Doyle
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| | - Sarah E Siegrist
- Department of Biology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
4
|
Dillon NR, Manning L, Hirono K, Doe CQ. Seven-up acts in neuroblasts to specify adult central complex neuron identity and initiate neuroblast decommissioning. Development 2024; 151:dev202504. [PMID: 38230563 PMCID: PMC10906098 DOI: 10.1242/dev.202504] [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: 11/03/2023] [Accepted: 12/22/2023] [Indexed: 01/18/2024]
Abstract
An unanswered question in neurobiology is how are diverse neuron cell types generated from a small number of neural stem cells? In the Drosophila larval central brain, there are eight bilateral Type 2 neuroblast (T2NB) lineages that express a suite of early temporal factors followed by a different set of late temporal factors and generate the majority of the central complex (CX) neurons. The early-to-late switch is triggered by the orphan nuclear hormone receptor Seven-up (Svp), yet little is known about how this Svp-dependent switch is involved in specifying CX neuron identities. Here, we: (1) birth date the CX neurons P-EN and P-FN (early and late, respectively); (2) show that Svp is transiently expressed in all early T2NBs; and (3) show that loss of Svp expands the population of early born P-EN neurons at the expense of late born P-FN neurons. Furthermore, in the absence of Svp, T2NBs fail decommissioning and abnormally extend their lineage into week-old adults. We conclude that Svp is required to specify CX neuron identity, as well as to initiate T2NB decommissioning.
Collapse
Affiliation(s)
- Noah R. Dillon
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Laurina Manning
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Keiko Hirono
- 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
|
5
|
Busby L, Serrano Nájera G, Steventon BJ. Intrinsic and extrinsic cues time somite progenitor contribution to the vertebrate primary body axis. eLife 2024; 13:e90499. [PMID: 38193440 PMCID: PMC10834026 DOI: 10.7554/elife.90499] [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: 06/26/2023] [Accepted: 01/08/2024] [Indexed: 01/10/2024] Open
Abstract
During embryonic development, the timing of events at the cellular level must be coordinated across multiple length scales to ensure the formation of a well-proportioned body plan. This is clear during somitogenesis, where progenitors must be allocated to the axis over time whilst maintaining a progenitor population for continued elaboration of the body plan. However, the relative importance of intrinsic and extrinsic signals in timing progenitor addition at the single-cell level is not yet understood. Heterochronic grafts from older to younger embryos have suggested a level of intrinsic timing whereby later staged cells contribute to more posterior portions of the axis. To determine the precise step at which cells are delayed, we performed single-cell transcriptomic analysis on heterochronic grafts of somite progenitors in the chicken embryo. This revealed a previously undescribed cell state within which heterochronic grafted cells are stalled. The delayed exit of older cells from this state correlates with expression of posterior Hox genes. Using grafting and explant culture, we find that both Hox gene expression and the migratory capabilities of progenitor populations are intrinsically regulated at the population level. However, by grafting varied sizes of tissue, we find that small heterochronic grafts disperse more readily and contribute to more anterior portions of the body axis while still maintaining Hox gene expression. This enhanced dispersion is not replicated in explant culture, suggesting that it is a consequence of interaction between host and donor tissue and thus extrinsic to the donor tissue. Therefore, we demonstrate that the timing of cell dispersion and resulting axis contribution is impacted by a combination of both intrinsic and extrinsic cues.
Collapse
Affiliation(s)
- Lara Busby
- Department of Genetics, University of CambridgeCambridgeUnited Kingdom
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | | | | |
Collapse
|
6
|
Liu T, Li T, Ke S. Role of the CASZ1 transcription factor in tissue development and disease. Eur J Med Res 2023; 28:562. [PMID: 38053207 DOI: 10.1186/s40001-023-01548-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023] Open
Abstract
The zinc finger transcription factor gene, CASZ1/Castor (Castor zinc finger 1), initially identified in Drosophila, plays a critical role in neural, cardiac, and cardiovascular development, exerting a complex, multifaceted influence on cell fate and tissue morphogenesis. During neurogenesis, CASZ1 exhibits dynamic expression from early embryonic development to the perinatal period, constituting a key regulator in this process. Additionally, CASZ1 controls the transition between neurogenesis and gliomagenesis. During human cardiovascular system development, CASZ1 is essential for cardiomyocyte differentiation, cardiac morphogenesis, and vascular morphology homeostasis and formation. The deletion or inactivation of CASZ1 mutations can lead to human developmental diseases or tumors, including congenital heart disease, cardiovascular disease, and neuroblastoma. CASZ1 can be used as a biomarker for disease prevention and diagnosis as well as a prognostic indicator for cancer. This review explores the unique functions of CASZ1 in tissue morphogenesis and associated diseases, offering new insights for elucidating the molecular mechanisms underlying diseases and identifying potential therapeutic targets for disease prevention and treatment.
Collapse
Affiliation(s)
- Tiantian Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, Henan, China.
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China.
| | - Tao Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou, 450002, China
| | - Shaorui Ke
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, 450046, Henan, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan, China
| |
Collapse
|
7
|
Dillon NR, Manning L, Hirono K, Doe CQ. Seven-up acts in neuroblasts to specify adult central complex neuron identity and initiate neuroblast decommissioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.02.565340. [PMID: 37961302 PMCID: PMC10635090 DOI: 10.1101/2023.11.02.565340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
An open question in neurobiology is how diverse neuron cell types are generated from a small number of neural stem cells. In the Drosophila larval central brain, there are eight bilateral Type 2 neuroblast (T2NB) lineages that express a suite of early temporal factors followed by a different set of late temporal factors and generate the majority of the central complex (CX) neurons. The early-to-late switch is triggered by the orphan nuclear hormone receptor Seven-up (Svp), yet little is known about this Svp-dependent switch in specifying CX neuron identities. Here, we (i) birthdate the CX neurons P-EN and P-FN (early and late, respectively); (ii) show that Svp is transiently expressed in all early T2NBs; and (iii) show that loss of Svp expands the population of early born P-EN neurons at the expense of late born P-FN neurons. Furthermore, in the absence of Svp, T2NBs fail decommissioning and abnormally extend their lineage into week-old adults. We conclude that Svp is required to specify CX neuron identity, as well as to initiate T2NB decommissioning.
Collapse
Affiliation(s)
- Noah R. Dillon
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| | - Laurina Manning
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| | - Keiko Hirono
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| | - Chris Q. Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| |
Collapse
|
8
|
González YR, Kamkar F, Jafar-Nejad P, Wang S, Qu D, Alvarez LS, Hawari D, Sonnenfeld M, Slack RS, Albert PR, Park DS, Joselin A. PFTK1 kinase regulates axogenesis during development via RhoA activation. BMC Biol 2023; 21:240. [PMID: 37907898 PMCID: PMC10617079 DOI: 10.1186/s12915-023-01732-w] [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: 09/26/2022] [Accepted: 10/11/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND PFTK1/Eip63E is a member of the cyclin-dependent kinases (CDKs) family and plays an important role in normal cell cycle progression. Eip63E expresses primarily in postnatal and adult nervous system in Drosophila melanogaster but its role in CNS development remains unknown. We sought to understand the function of Eip63E in the CNS by studying the fly ventral nerve cord during development. RESULTS Our results demonstrate that Eip63E regulates axogenesis in neurons and its deficiency leads to neuronal defects. Functional interaction studies performed using the same system identify an interaction between Eip63E and the small GTPase Rho1. Furthermore, deficiency of Eip63E homolog in mice, PFTK1, in a newly generated PFTK1 knockout mice results in increased axonal outgrowth confirming that the developmental defects observed in the fly model are due to defects in axogenesis. Importantly, RhoA phosphorylation and activity are affected by PFTK1 in primary neuronal cultures. We report that GDP-bound inactive RhoA is a substrate of PFTK1 and PFTK1 phosphorylation is required for RhoA activity. CONCLUSIONS In conclusion, our work establishes an unreported neuronal role of PFTK1 in axon development mediated by phosphorylation and activation of GDP-bound RhoA. The results presented add to our understanding of the role of Cdks in the maintenance of RhoA-mediated axon growth and its impact on CNS development and axonal regeneration.
Collapse
Affiliation(s)
| | - Fatemeh Kamkar
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Paymaan Jafar-Nejad
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Present Address: Ionis Pharmaceuticals Inc., Carlsbad, CA, 92010, USA
| | - Suzi Wang
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Dianbo Qu
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Leticia Sanchez Alvarez
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Dina Hawari
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Margaret Sonnenfeld
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Ruth S Slack
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute and Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - David S Park
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| | - Alvin Joselin
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
9
|
Thompson MK, Ceccarelli A, Ish-Horowicz D, Davis I. Dynamically regulated transcription factors are encoded by highly unstable mRNAs in the Drosophila larval brain. RNA (NEW YORK, N.Y.) 2023; 29:1020-1032. [PMID: 37041032 DOI: 10.1261/rna.079552.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
The level of each RNA species depends on the balance between its rates of production and decay. Although previous studies have measured RNA decay across the genome in tissue culture and single-celled organisms, few experiments have been performed in intact complex tissues and organs. It is therefore unclear whether the determinants of RNA decay found in cultured cells are preserved in an intact tissue, and whether they differ between neighboring cell types and are regulated during development. To address these questions, we measured RNA synthesis and decay rates genome wide via metabolic labeling of whole cultured Drosophila larval brains using 4-thiouridine. Our analysis revealed that decay rates span a range of more than 100-fold, and that RNA stability is linked to gene function, with mRNAs encoding transcription factors being much less stable than mRNAs involved in core metabolic functions. Surprisingly, among transcription factor mRNAs there was a clear demarcation between more widely used transcription factors and those that are expressed only transiently during development. mRNAs encoding transient transcription factors are among the least stable in the brain. These mRNAs are characterized by epigenetic silencing in most cell types, as shown by their enrichment with the histone modification H3K27me3. Our data suggest the presence of an mRNA destabilizing mechanism targeted to these transiently expressed transcription factors to allow their levels to be regulated rapidly with high precision. Our study also demonstrates a general method for measuring mRNA transcription and decay rates in intact organs or tissues, offering insights into the role of mRNA stability in the regulation of complex developmental programs.
Collapse
Affiliation(s)
- Mary Kay Thompson
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Arianna Ceccarelli
- Mathematical Institute, University of Oxford, Oxford OX1 3LB, United Kingdom
| | - David Ish-Horowicz
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Ilan Davis
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| |
Collapse
|
10
|
Santos-França PL, David LA, Kassem F, Meng XQ, Cayouette M. Time to see: How temporal identity factors specify the developing mammalian retina. Semin Cell Dev Biol 2023; 142:36-42. [PMID: 35760728 DOI: 10.1016/j.semcdb.2022.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022]
Abstract
Understanding how retinal progenitor cells (RPCs) give rise to the variety of neural cell types of the retina has been a question of major interest over the last few decades. While environmental cues and transcription factor networks have been shown to control specific cell fate decisions, how RPCs alter fate output over time to control proper histogenesis remains poorly understood. In recent years, the identification of "temporal identity factors (TIFs)", which control RPC competence states to ensure that the right cell types are produced at the right time, has contributed to increasing our understanding of temporal patterning in the retina. Here, we review the different TIFs identified to date in the mammalian retina and discuss the underlying mechanisms by which they are thought to operate. We conclude by speculating on how identification of temporal patterning mechanisms might support the development of new therapeutic approaches against visual impairments.
Collapse
Affiliation(s)
- Pedro L Santos-França
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada; Molecular Biology Program, Université de Montréal, Montréal, QC, Canada
| | - Luke Ajay David
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada; Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Fatima Kassem
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada; Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Xiang Qi Meng
- Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada; Molecular Biology Program, Université de Montréal, Montréal, QC, Canada; Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montréal, QC, Canada; Department of Medicine, Université de Montréal, QC, Canada; Department of Anatomy and Cell Biology and Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
11
|
Hamid A, Gutierrez A, Munroe J, Syed MH. The Drivers of Diversity: Integrated genetic and hormonal cues regulate neural diversity. Semin Cell Dev Biol 2023; 142:23-35. [PMID: 35915026 DOI: 10.1016/j.semcdb.2022.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/06/2022] [Accepted: 07/17/2022] [Indexed: 11/17/2022]
Abstract
Proper functioning of the nervous system relies not only on the generation of a vast repertoire of distinct neural cell types but also on the precise neural circuitry within them. How the generation of highly diverse neural populations is regulated during development remains a topic of interest. Landmark studies in Drosophila have identified the genetic and temporal cues regulating neural diversity and thus have provided valuable insights into our understanding of temporal patterning of the central nervous system. The development of the Drosophila central complex, which is mostly derived from type II neural stem cell (NSC) lineages, showcases how a small pool of NSCs can give rise to vast and distinct progeny. Similar to the human outer subventricular zone (OSVZ) neural progenitors, type II NSCs generate intermediate neural progenitors (INPs) to expand and diversify lineages that populate higher brain centers. Each type II NSC has a distinct spatial identity and timely regulated expression of many transcription factors and mRNA binding proteins. Additionally, INPs derived from them show differential expression of genes depending on their birth order. Together type II NSCs and INPs display a combinatorial temporal patterning that expands neural diversity of the central brain lineages. We cover advances in current understanding of type II NSC temporal patterning and discuss similarities and differences in temporal patterning mechanisms of various NSCs with a focus on how cell-intrinsic and extrinsic hormonal cues regulate temporal transitions in NSCs during larval development. Cell extrinsic ligands activate conserved signaling pathways and extrinsic hormonal cues act as a temporal switch that regulate temporal progression of the NSCs. We conclude by elaborating on how a progenitor's temporal code regulates the fate specification and identity of distinct neural types. At the end, we also discuss open questions in linking developmental cues to neural identity, circuits, and underlying behaviors in the adult fly.
Collapse
Affiliation(s)
- Aisha Hamid
- Department of Biology, University of New Mexico, Albuquerque, NM 87113, USA
| | - Andrew Gutierrez
- Department of Biology, University of New Mexico, Albuquerque, NM 87113, USA
| | - Jordan Munroe
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | | |
Collapse
|
12
|
Specification of the Drosophila Orcokinin A neurons by combinatorial coding. Cell Tissue Res 2023; 391:269-286. [PMID: 36512054 DOI: 10.1007/s00441-022-03721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
The central nervous system contains a daunting number of different cell types. Understanding how each cell acquires its fate remains a major challenge for neurobiology. The developing embryonic ventral nerve cord (VNC) of Drosophila melanogaster has been a powerful model system for unraveling the basic principles of cell fate specification. This pertains specifically to neuropeptide neurons, which typically are stereotypically generated in discrete subsets, allowing for unambiguous single-cell resolution in different genetic contexts. Here, we study the specification of the OrcoA-LA neurons, characterized by the expression of the neuropeptide Orcokinin A and located laterally in the A1-A5 abdominal segments of the VNC. We identified the progenitor neuroblast (NB; NB5-3) and the temporal window (castor/grainyhead) that generate the OrcoA-LA neurons. We also describe the role of the Ubx, abd-A, and Abd-B Hox genes in the segment-specific generation of these neurons. Additionally, our results indicate that the OrcoA-LA neurons are "Notch Off" cells, and neither programmed cell death nor the BMP pathway appears to be involved in their specification. Finally, we performed a targeted genetic screen of 485 genes known to be expressed in the CNS and identified nab, vg, and tsh as crucial determinists for OrcoA-LA neurons. This work provides a new neuropeptidergic model that will allow for addressing new questions related to neuronal specification mechanisms in the future.
Collapse
|
13
|
Javed A, Santos-França PL, Mattar P, Cui A, Kassem F, Cayouette M. Ikaros family proteins redundantly regulate temporal patterning in the developing mouse retina. Development 2023; 150:286611. [PMID: 36537580 DOI: 10.1242/dev.200436] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Temporal identity factors regulate competence of neural progenitors to generate specific cell types in a time-dependent manner, but how they operate remains poorly defined. In the developing mouse retina, the Ikaros zinc-finger transcription factor Ikzf1 regulates production of early-born cell types, except cone photoreceptors. In this study we show that, during early stages of retinal development, another Ikaros family protein, Ikzf4, functions redundantly with Ikzf1 to regulate cone photoreceptor production. Using CUT&RUN and functional assays, we show that Ikzf4 binds and represses genes involved in late-born rod photoreceptor specification, hence favoring cone production. At late stages, when Ikzf1 is no longer expressed in progenitors, we show that Ikzf4 re-localizes to target genes involved in gliogenesis and is required for Müller glia production. We report that Ikzf4 regulates Notch signaling genes and is sufficient to activate the Hes1 promoter through two Ikzf GGAA-binding motifs, suggesting a mechanism by which Ikzf4 may influence gliogenesis. These results uncover a combinatorial role for Ikaros family members during nervous system development and provide mechanistic insights on how they temporally regulate cell fate output.
Collapse
Affiliation(s)
- Awais Javed
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal H2W 1R7, Canada
- Molecular Biology Program, Université de Montréal, Montreal H3T 1J4, Canada
| | - Pedro L Santos-França
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal H2W 1R7, Canada
- Molecular Biology Program, Université de Montréal, Montreal H3T 1J4, Canada
| | - Pierre Mattar
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal H2W 1R7, Canada
| | - Allie Cui
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal H2W 1R7, Canada
| | - Fatima Kassem
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montreal H3A 0G4, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal H2W 1R7, Canada
- Molecular Biology Program, Université de Montréal, Montreal H3T 1J4, Canada
- Integrated Program in Neuroscience, McGill University, Montreal H3A 0G4, Canada
- Department of Medicine, Université de Montréal, Montreal H3T 1J4, Canada
- Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Montreal H3A 0G4, Canada
| |
Collapse
|
14
|
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
|
15
|
Ray A, Li X. A Notch-dependent transcriptional mechanism controls expression of temporal patterning factors in Drosophila medulla. eLife 2022; 11:e75879. [PMID: 36040415 PMCID: PMC9427115 DOI: 10.7554/elife.75879] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Temporal patterning is an important mechanism for generating a great diversity of neuron subtypes from a seemingly homogenous progenitor pool in both vertebrates and invertebrates. Drosophila neuroblasts are temporally patterned by sequentially expressed Temporal Transcription Factors (TTFs). These TTFs are proposed to form a transcriptional cascade based on mutant phenotypes, although direct transcriptional regulation between TTFs has not been verified in most cases. Furthermore, it is not known how the temporal transitions are coupled with the generation of the appropriate number of neurons at each stage. We use neuroblasts of the Drosophila optic lobe medulla to address these questions and show that the expression of TTFs Sloppy-paired 1/2 (Slp1/2) is directly regulated at the transcriptional level by two other TTFs and the cell-cycle dependent Notch signaling through two cis-regulatory elements. We also show that supplying constitutively active Notch can rescue the delayed transition into the Slp stage in cell cycle arrested neuroblasts. Our findings reveal a novel Notch-pathway dependent mechanism through which the cell cycle progression regulates the timing of a temporal transition within a TTF transcriptional cascade.
Collapse
Affiliation(s)
- Alokananda Ray
- Department of Cell and Developmental Biology, University of Illinois at Urbana-ChampaignUrbanaUnited States
| | - Xin Li
- Department of Cell and Developmental Biology, University of Illinois at Urbana-ChampaignUrbanaUnited States
| |
Collapse
|
16
|
Tang JLY, Hakes AE, Krautz R, Suzuki T, Contreras EG, Fox PM, Brand AH. NanoDam identifies Homeobrain (ARX) and Scarecrow (NKX2.1) as conserved temporal factors in the Drosophila central brain and visual system. Dev Cell 2022; 57:1193-1207.e7. [PMID: 35483359 PMCID: PMC9616798 DOI: 10.1016/j.devcel.2022.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 02/08/2022] [Accepted: 04/05/2022] [Indexed: 11/24/2022]
Abstract
Temporal patterning of neural progenitors is an evolutionarily conserved strategy for generating neuronal diversity. Type II neural stem cells in the Drosophila central brain produce transit-amplifying intermediate neural progenitors (INPs) that exhibit temporal patterning. However, the known temporal factors cannot account for the neuronal diversity in the adult brain. To search for missing factors, we developed NanoDam, which enables rapid genome-wide profiling of endogenously tagged proteins in vivo with a single genetic cross. Mapping the targets of known temporal transcription factors with NanoDam revealed that Homeobrain and Scarecrow (ARX and NKX2.1 orthologs) are also temporal factors. We show that Homeobrain and Scarecrow define middle-aged and late INP temporal windows and play a role in cellular longevity. Strikingly, Homeobrain and Scarecrow have conserved functions as temporal factors in the developing visual system. NanoDam enables rapid cell-type-specific genome-wide profiling with temporal resolution and is easily adapted for use in higher organisms.
Collapse
Affiliation(s)
- Jocelyn L Y Tang
- Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Anna E Hakes
- Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Robert Krautz
- Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Takumi Suzuki
- Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Esteban G Contreras
- Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Paul M Fox
- Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| | - Andrea H Brand
- Gurdon Institute and Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK.
| |
Collapse
|
17
|
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
|
18
|
Pérez E, Venkatanarayan A, Lundell MJ. Hunchback prevents notch-induced apoptosis in the serotonergic lineage of Drosophila Melanogaster. Dev Biol 2022; 486:109-120. [PMID: 35381219 DOI: 10.1016/j.ydbio.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/16/2022] [Accepted: 03/30/2022] [Indexed: 11/19/2022]
Abstract
The serotonergic lineage (NB7-3) in the Drosophila ventral nerve cord produces six cells during neurogenesis. Four of the cells differentiate into neurons: EW1, EW2, EW3 and GW. The other two cells undergo apoptosis. This simple lineage provides an opportunity to examine genes that are required to induce or repress apoptosis during cell specification. Previous studies have shown that Notch signaling induces apoptosis within the NB7-3 lineage. The three EW neurons are protected from Notch-induced apoptosis by asymmetric distribution of Numb protein, an inhibitor of Notch signaling. In a numb1 mutant EW2 and EW3 undergo apoptosis. The EW1 and GW neurons survive even in a numb1 mutant background suggesting that these cells are protected from Notch-induced apoptosis by some factor other than Numb. The EW1 and GW neurons are mitotic sister cells, and uniquely express the transcription factor Hunchback. We present evidence that Hunchback prevents apoptosis in the NB7-3 lineage during normal CNS development and can rescue the two apoptotic cells in the lineage when it is ectopically expressed. We show that hunchback overexpression produces ectopic cells that express markers similar to the EW2 neuron and changes the expression pattern of the EW3 neuron to a EW2 neuron In addition we show that hunchback overexpression can override apoptosis that is genetically induced by the pro-apoptotic genes grim and hid.
Collapse
Affiliation(s)
- Ernesto Pérez
- Department of Biology, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA
| | | | - Martha J Lundell
- Department of Biology, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
19
|
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
|
20
|
Hafer TL, Patra S, Tagami D, Kohwi M. Enhancer of trithorax/polycomb, Corto, regulates timing of hunchback gene relocation and competence in Drosophila neuroblasts. Neural Dev 2022; 17:3. [PMID: 35177098 PMCID: PMC8855600 DOI: 10.1186/s13064-022-00159-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/27/2022] [Indexed: 12/27/2022] Open
Abstract
Background Neural progenitors produce diverse cells in a stereotyped birth order, but can specify each cell type for only a limited duration. In the Drosophila embryo, neuroblasts (neural progenitors) specify multiple, distinct neurons by sequentially expressing a series of temporal identity transcription factors with each division. Hunchback (Hb), the first of the series, specifies early-born neuronal identity. Neuroblast competence to generate early-born neurons is terminated when the hb gene relocates to the neuroblast nuclear lamina, rendering it refractory to activation in descendent neurons. Mechanisms and trans-acting factors underlying this process are poorly understood. Here we identify Corto, an enhancer of Trithorax/Polycomb (ETP) protein, as a new regulator of neuroblast competence. Methods We used the GAL4/UAS system to drive persistent misexpression of Hb in neuroblast 7–1 (NB7-1), a model lineage for which the early competence window has been well characterized, to examine the role of Corto in neuroblast competence. We used immuno-DNA Fluorescence in situ hybridization (DNA FISH) in whole embryos to track the position of the hb gene locus specifically in neuroblasts across developmental time, comparing corto mutants to control embryos. Finally, we used immunostaining in whole embryos to examine Corto’s role in repression of Hb and a known target gene, Abdominal B (Abd-B). Results We found that in corto mutants, the hb gene relocation to the neuroblast nuclear lamina is delayed and the early competence window is extended. The delay in gene relocation occurs after hb transcription is already terminated in the neuroblast and is not due to prolonged transcriptional activity. Further, we find that Corto genetically interacts with Posterior Sex Combs (Psc), a core subunit of polycomb group complex 1 (PRC1), to terminate early competence. Loss of Corto does not result in derepression of Hb or its Hox target, Abd-B, specifically in neuroblasts. Conclusions These results show that in neuroblasts, Corto genetically interacts with PRC1 to regulate timing of nuclear architecture reorganization and support the model that distinct mechanisms of silencing are implemented in a step-wise fashion during development to regulate cell fate gene expression in neuronal progeny. Supplementary Information The online version contains supplementary material available at 10.1186/s13064-022-00159-3.
Collapse
Affiliation(s)
- Terry L Hafer
- Department of Neuroscience, Mortimer B. Zuckerman Institute Mind Brain Behavior, Columbia University, New York, NY, 10027, USA.,Present Address: Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA
| | - Sofiya Patra
- Department of Neuroscience, Mortimer B. Zuckerman Institute Mind Brain Behavior, Columbia University, New York, NY, 10027, USA
| | - Daiki Tagami
- Department of Neuroscience, Mortimer B. Zuckerman Institute Mind Brain Behavior, Columbia University, New York, NY, 10027, USA
| | - Minoree Kohwi
- Department of Neuroscience, Mortimer B. Zuckerman Institute Mind Brain Behavior, Columbia University, New York, NY, 10027, USA. .,Kavli Institute for Brain Science, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
21
|
Wu Q, Shichino Y, Abe T, Suetsugu T, Omori A, Kiyonari H, Iwasaki S, Matsuzaki F. Selective translation of epigenetic modifiers affects the temporal pattern and differentiation of neural stem cells. Nat Commun 2022; 13:470. [PMID: 35078993 PMCID: PMC8789897 DOI: 10.1038/s41467-022-28097-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/07/2022] [Indexed: 11/09/2022] Open
Abstract
The cerebral cortex is formed by diverse neurons generated sequentially from neural stem cells (NSCs). A clock mechanism has been suggested to underlie the temporal progression of NSCs, which is mainly defined by the transcriptome and the epigenetic state. However, what drives such a developmental clock remains elusive. We show that translational control of histone H3 trimethylation in Lys27 (H3K27me3) modifiers is part of this clock. We find that depletion of Fbl, an rRNA methyltransferase, reduces translation of both Ezh2 methyltransferase and Kdm6b demethylase of H3K27me3 and delays the progression of the NSC state. These defects are partially phenocopied by simultaneous inhibition of H3K27me3 methyltransferase and demethylase, indicating the role of Fbl in the genome-wide H3K27me3 pattern. Therefore, we propose that Fbl drives the intrinsic clock through the translational enhancement of the H3K27me3 modifiers that predominantly define the NSC state. The temporal development of tissues and organs may be defined by the genome-wide epigenetic and transcriptional state functioning as the clock. Here the authors found that Fbl, a ribosomal RNA methyltransferase, potentially behaves as a clock during neural stem cell (NSC) development by controlling translational efficiencies of epigenetic modifiers in the cerebral cortex primordium.
Collapse
|
22
|
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
|
23
|
Tidswell ORA, Benton MA, Akam M. The neuroblast timer gene nubbin exhibits functional redundancy with gap genes to regulate segment identity in Tribolium. Development 2021; 148:271199. [PMID: 34351412 PMCID: PMC8406537 DOI: 10.1242/dev.199719] [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: 04/20/2021] [Accepted: 07/23/2021] [Indexed: 12/05/2022]
Abstract
The neuroblast timer genes hunchback, Krüppel, nubbin and castor are expressed in temporal sequence in neural stem cells, and in corresponding spatial sequence along the Drosophila blastoderm. As canonical gap genes, hunchback and Krüppel play a crucial role in insect segmentation, but the roles of nubbin and castor in this process remain ambiguous. We have investigated the expression and functions of nubbin and castor during segmentation in the beetle Tribolium. We show that Tc-hunchback, Tc-Krüppel, Tc-nubbin and Tc-castor are expressed sequentially in the segment addition zone, and that Tc-nubbin regulates segment identity redundantly with two previously described gap/gap-like genes, Tc-giant and Tc-knirps. Simultaneous knockdown of Tc-nubbin, Tc-giant and Tc-knirps results in the formation of ectopic legs on abdominal segments. This homeotic transformation is caused by loss of abdominal Hox gene expression, likely due to expanded Tc-Krüppel expression. Our findings support the theory that the neuroblast timer series was co-opted for use in insect segment patterning, and contribute to our growing understanding of the evolution and function of the gap gene network outside of Drosophila. Summary:nubbin and the gap genes knirps and giant redundantly repress Krüppel expression during segmentation. Simultaneous knockdown of all three genes causes ectopic Krüppel expression and loss of abdominal segment identity.
Collapse
Affiliation(s)
| | - Matthew A Benton
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Michael Akam
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| |
Collapse
|
24
|
Abstract
The temporal coordination of events at cellular and tissue scales is essential for the proper development of organisms, and involves cell-intrinsic processes that can be coupled by local cellular signalling and instructed by global signalling, thereby creating spatial patterns of cellular states that change over time. The timing and structure of these patterns determine how an organism develops. Traditional developmental genetic methods have revealed the complex molecular circuits regulating these processes but are limited in their ability to predict and understand the emergent spatio-temporal dynamics. Increasingly, approaches from physics are now being used to help capture the dynamics of the system by providing simplified, generic descriptions. Combined with advances in imaging and computational power, such approaches aim to provide insight into timing and patterning in developing systems.
Collapse
|
25
|
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
|
26
|
Busby L, Steventon B. Tissue tectonics and the multi-scale regulation of developmental timing. Interface Focus 2021; 11:20200057. [PMID: 34055304 PMCID: PMC8086930 DOI: 10.1098/rsfs.2020.0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Development encompasses processes that occur at multiple length scales, including gene-regulatory interactions, cell movements and reorganization, cell signalling and growth. It is essential that the timing of events in all of these different processes is coordinated to generate well-patterned tissues and organs. However, how the timing of intrinsic cell state changes is coordinated with events occurring at the multi-tissue and whole-organism level is unknown. Here, we argue that an important mechanism that accounts for the integration of timing across levels of organization is provided by tissue tectonics, i.e. how morphogenetic events driving tissue shape changes result in the relative displacement of signalling and responding tissues and coordinate developmental timing across scales. In doing so, tissue tectonics provides a mechanism by which the cell specification events intrinsic to cells can be modulated by the temporal exposure to extracellular signals. This exposure is in turn regulated by higher-order properties of the embryo, such as their physical properties, rates of growth and the combination of dynamic cell behaviours, impacting tissue morphogenesis. Tissue tectonics creates a downward flow of information from higher to lower levels of biological organization, providing an instance of downward causation in development.
Collapse
Affiliation(s)
- Lara Busby
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|
27
|
Rossi AM, Jafari S, Desplan C. Integrated Patterning Programs During Drosophila Development Generate the Diversity of Neurons and Control Their Mature Properties. Annu Rev Neurosci 2021; 44:153-172. [PMID: 33556251 DOI: 10.1146/annurev-neuro-102120-014813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
During the approximately 5 days of Drosophila neurogenesis (late embryogenesis to the beginning of pupation), a limited number of neural stem cells produce approximately 200,000 neurons comprising hundreds of cell types. To build a functional nervous system, neuronal types need to be produced in the proper places, appropriate numbers, and correct times. We discuss how neural stem cells (neuroblasts) obtain so-called area codes for their positions in the nervous system (spatial patterning) and how they keep time to sequentially produce neurons with unique fates (temporal patterning). We focus on specific examples that demonstrate how a relatively simple patterning system (Notch) can be used reiteratively to generate different neuronal types. We also speculate on how different modes of temporal patterning that operate over short versus long time periods might be linked. We end by discussing how specification programs are integrated and lead to the terminal features of different neuronal types.
Collapse
Affiliation(s)
- Anthony M Rossi
- Department of Biology, New York University, New York, NY 10003, USA; .,Department of Neurobiology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shadi Jafari
- 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
|
28
|
Sood C, Doyle SE, Siegrist SE. Steroid hormones, dietary nutrients, and temporal progression of neurogenesis. CURRENT OPINION IN INSECT SCIENCE 2021; 43:70-77. [PMID: 33127508 PMCID: PMC8058227 DOI: 10.1016/j.cois.2020.10.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/10/2020] [Accepted: 10/16/2020] [Indexed: 05/13/2023]
Abstract
Temporal patterning of neural progenitors, in which different factors are sequentially expressed, is an evolutionarily conserved strategy for generating neuronal diversity during development. In the Drosophila embryo, mechanisms that mediate temporal patterning of neural stem cells (neuroblasts) are largely cell-intrinsic. However, after embryogenesis, neuroblast temporal patterning relies on extrinsic cues as well, as freshly hatched larvae seek out nutrients and other key resources in varying natural environments. We recap current understanding of neuroblast-intrinsic temporal programs and discuss how neuroblast extrinsic cues integrate and coordinate with neuroblast intrinsic programs to control numbers and types of neurons produced. One key emerging extrinsic factor that impacts temporal patterning of neuroblasts and their daughters as well as termination of neurogenesis is the steroid hormone, ecdysone, a known regulator of large-scale developmental transitions in insects and arthropods. Lastly, we consider evolutionary conservation and discuss recent work on thyroid hormone signaling in early vertebrate brain development.
Collapse
Affiliation(s)
- Chhavi Sood
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Susan E Doyle
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA
| | - Sarah E Siegrist
- Department of Biology, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
29
|
Lopes A, Magrinelli E, Telley L. Emerging Roles of Single-Cell Multi-Omics in Studying Developmental Temporal Patterning. Int J Mol Sci 2020; 21:E7491. [PMID: 33050604 PMCID: PMC7589732 DOI: 10.3390/ijms21207491] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 01/16/2023] Open
Abstract
The complexity of brain structure and function is rooted in the precise spatial and temporal regulation of selective developmental events. During neurogenesis, both vertebrates and invertebrates generate a wide variety of specialized cell types through the expansion and specification of a restricted set of neuronal progenitors. Temporal patterning of neural progenitors rests on fine regulation between cell-intrinsic and cell-extrinsic mechanisms. The rapid emergence of high-throughput single-cell technologies combined with elaborate computational analysis has started to provide us with unprecedented biological insights related to temporal patterning in the developing central nervous system (CNS). Here, we present an overview of recent advances in Drosophila and vertebrates, focusing both on cell-intrinsic mechanisms and environmental influences. We then describe the various multi-omics approaches that have strongly contributed to our current understanding and discuss perspectives on the various -omics approaches that hold great potential for the future of temporal patterning research.
Collapse
Affiliation(s)
| | | | - Ludovic Telley
- Department of Basic Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland; (A.L.); (E.M.)
| |
Collapse
|
30
|
Maurange C. Temporal patterning in neural progenitors: from Drosophila development to childhood cancers. Dis Model Mech 2020; 13:dmm044883. [PMID: 32816915 PMCID: PMC7390627 DOI: 10.1242/dmm.044883] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The developing central nervous system (CNS) is particularly prone to malignant transformation, but the underlying mechanisms remain unresolved. However, periods of tumor susceptibility appear to correlate with windows of increased proliferation, which are often observed during embryonic and fetal stages and reflect stereotypical changes in the proliferative properties of neural progenitors. The temporal mechanisms underlying these proliferation patterns are still unclear in mammals. In Drosophila, two decades of work have revealed a network of sequentially expressed transcription factors and RNA-binding proteins that compose a neural progenitor-intrinsic temporal patterning system. Temporal patterning controls both the identity of the post-mitotic progeny of neural progenitors, according to the order in which they arose, and the proliferative properties of neural progenitors along development. In addition, in Drosophila, temporal patterning delineates early windows of cancer susceptibility and is aberrantly regulated in developmental tumors to govern cellular hierarchy as well as the metabolic and proliferative heterogeneity of tumor cells. Whereas recent studies have shown that similar genetic programs unfold during both fetal development and pediatric brain tumors, I discuss, in this Review, how the concept of temporal patterning that was pioneered in Drosophila could help to understand the mechanisms of initiation and progression of CNS tumors in children.
Collapse
Affiliation(s)
- Cédric Maurange
- Aix Marseille University, CNRS, IBDM, Equipe Labellisée LIGUE Contre le Cancer, Marseille 13009, France
| |
Collapse
|
31
|
Mira H, Morante J. Neurogenesis From Embryo to Adult - Lessons From Flies and Mice. Front Cell Dev Biol 2020; 8:533. [PMID: 32695783 PMCID: PMC7339912 DOI: 10.3389/fcell.2020.00533] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Abstract
The human brain is composed of billions of cells, including neurons and glia, with an undetermined number of subtypes. During the embryonic and early postnatal stages, the vast majority of these cells are generated from neural progenitors and stem cells located in all regions of the neural tube. A smaller number of neurons will continue to be generated throughout our lives, in localized neurogenic zones, mainly confined at least in rodents to the subependymal zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus. During neurogenesis, a combination of extrinsic cues interacting with temporal and regional intrinsic programs are thought to be critical for increasing neuronal diversity, but their underlying mechanisms need further elucidation. In this review, we discuss the recent findings in Drosophila and mammals on the types of cell division and cell interactions used by neural progenitors and stem cells to sustain neurogenesis, and how they are influenced by glia.
Collapse
Affiliation(s)
- Helena Mira
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Javier Morante
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernandez, Alicante, Spain
| |
Collapse
|
32
|
Does Birth Trigger Cell Death in the Developing Brain? eNeuro 2020; 7:ENEURO.0517-19.2020. [PMID: 32015098 PMCID: PMC7031855 DOI: 10.1523/eneuro.0517-19.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/27/2022] Open
Abstract
Developmental cell death eliminates half of the neurons initially generated in the mammalian brain, and occurs perinatally in many species. It is possible that the timing of neuronal cell death is developmentally programmed, and only coincidentally associated with birth. Alternatively, birth may play a role in shaping cell death. To test these competing hypotheses, we experimentally advanced or delayed birth by 1 d in mice (within the normal range of gestation for the species) and examined effects on the temporal pattern and magnitude (amount) of neuronal cell death, using immunohistochemical detection of activated caspase-3 as a cell death marker. In order to detect effects of subtle changes in birth timing, we focused on brain areas that exhibit sharp postnatal peaks in cell death. We find that advancing birth advances peak cell death, supporting the hypothesis that birth triggers cell death. However, a delay of birth does not delay cell death. Thus, birth can advance cell death, but if postponed, a developmental program governs. Advancing or delaying birth also caused region-specific changes in the overall magnitude of cell death. Our findings shed light on the long-standing question of what controls the timing and magnitude of developmental neuronal cell death, and position birth as an orchestrator of brain development. Because humans across the world now routinely alter birth timing, these findings may have implications for current obstetric practices.
Collapse
|
33
|
Javed A, Mattar P, Lu S, Kruczek K, Kloc M, Gonzalez-Cordero A, Bremner R, Ali RR, Cayouette M. Pou2f1 and Pou2f2 cooperate to control the timing of cone photoreceptor production in the developing mouse retina. Development 2020; 147:dev.188730. [DOI: 10.1242/dev.188730] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 08/19/2020] [Indexed: 12/27/2022]
Abstract
Multipotent retinal progenitor cells (RPCs) generate various cell types in a precise chronological order, but how exactly cone photoreceptor production is restricted to early stages remains unclear. Here, we show that the POU-homeodomain factors Pou2f1/Pou2f2, the homologs of Drosophila temporal identity factors nub/pdm2, regulate the timely production of cones in mice. Forcing sustained expression of Pou2f1 or Pou2f2 in RPCs expands the period of cone production, whereas misexpression in late-stage RPCs triggers ectopic cone production at the expense of late-born fates. Mechanistically, we report that Pou2f1 induces Pou2f2 expression, which binds to a POU motif in the promoter of the rod-inducing factor Nrl to repress its expression. Conversely, conditional inactivation of Pou2f2 in RPCs increases Nrl expression and reduces cone production. Finally, we provide evidence that Pou2f1 is part of a cross-regulatory cascade with the other temporal identity factors Ikzf1 and Casz1. These results uncover Pou2f1/2 as regulators of the temporal window for cone genesis and, given their widespread expression in the nervous system, raise the possibility of a general role in temporal patterning.
Collapse
Affiliation(s)
- Awais Javed
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montreal (IRCM), Canada
- Molecular Biology Program, Université de Montréal, Canada
| | - Pierre Mattar
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montreal (IRCM), Canada
| | - Suying Lu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada. Department of Ophthalmology and Vision Science, Department of Lab Medicine and Pathobiology, University of Toronto
| | | | | | | | - Rod Bremner
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada. Department of Ophthalmology and Vision Science, Department of Lab Medicine and Pathobiology, University of Toronto
| | - Robin R. Ali
- UCL Institute of Ophthalmology, London, UK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montreal (IRCM), Canada
- Molecular Biology Program, Université de Montréal, Canada
- Department of Medicine, Université de Montréal, Canada
- Department of Anatomy and Cell Biology; Division of Experimental Medicine, McGill University, Canada
| |
Collapse
|
34
|
Meng JL, Marshall ZD, Lobb-Rabe M, Heckscher ES. How prolonged expression of Hunchback, a temporal transcription factor, re-wires locomotor circuits. eLife 2019; 8:46089. [PMID: 31502540 PMCID: PMC6754208 DOI: 10.7554/elife.46089] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022] Open
Abstract
How circuits assemble starting from stem cells is a fundamental question in developmental neurobiology. We test the hypothesis that, in neuronal stem cells, temporal transcription factors predictably control neuronal terminal features and circuit assembly. Using the Drosophila motor system, we manipulate expression of the classic temporal transcription factor Hunchback (Hb) specifically in the NB7-1 stem cell, which produces U motor neurons (MNs), and then we monitor dendrite morphology and neuromuscular synaptic partnerships. We find that prolonged expression of Hb leads to transient specification of U MN identity, and that embryonic molecular markers do not accurately predict U MN terminal features. Nonetheless, our data show Hb acts as a potent regulator of neuromuscular wiring decisions. These data introduce important refinements to current models, show that molecular information acts early in neurogenesis as a switch to control motor circuit wiring, and provide novel insight into the relationship between stem cell and circuit.
Collapse
Affiliation(s)
- Julia L Meng
- Department of Molecular Genetics and Cell Biology, Grossman Institute for Neuroscience, Program in Cell and Molecular Biology, University of Chicago, Chicago, United States.,Program in Cell and Molecular Biology, University of Chicago, Chicago, United States
| | - Zarion D Marshall
- Department of Molecular Genetics and Cell Biology, Grossman Institute for Neuroscience, Program in Cell and Molecular Biology, University of Chicago, Chicago, United States
| | - Meike Lobb-Rabe
- Department of Molecular Genetics and Cell Biology, Grossman Institute for Neuroscience, Program in Cell and Molecular Biology, University of Chicago, Chicago, United States.,Program in Cell and Molecular Biology, University of Chicago, Chicago, United States
| | - Ellie S Heckscher
- Department of Molecular Genetics and Cell Biology, Grossman Institute for Neuroscience, Program in Cell and Molecular Biology, University of Chicago, Chicago, United States
| |
Collapse
|
35
|
Spirov AV, Myasnikova EM. Evolutionary Stability of Gene Regulatory Networks That Define the Temporal Identity of Neuroblasts. Mol Biol 2019. [DOI: 10.1134/s0026893319020158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
36
|
Oberst P, Agirman G, Jabaudon D. Principles of progenitor temporal patterning in the developing invertebrate and vertebrate nervous system. Curr Opin Neurobiol 2019; 56:185-193. [PMID: 30999235 DOI: 10.1016/j.conb.2019.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/27/2019] [Accepted: 03/01/2019] [Indexed: 10/27/2022]
Abstract
During the development of the central nervous system, progenitors successively generate distinct types of neurons which assemble into the circuits that underlie our ability to interact with the environment. Spatial and temporal patterning mechanisms are partially evolutionarily conserved processes that allow generation of neuronal diversity from a limited set of progenitors. Here, we review examples of temporal patterning in neuronal progenitors in the Drosophila ventral nerve cord and in the mammalian cerebral cortex. We discuss cell-autonomous mechanisms and environmental influences on the temporal transitions of neuronal progenitors. Identifying the principles controlling the temporal specification of progenitors across species, as highlighted here, may help understand the evolutionary constraints over brain circuit design and function.
Collapse
Affiliation(s)
- Polina Oberst
- Department of Basic Neurosciences, University of Geneva, Switzerland
| | - Gulistan Agirman
- Department of Basic Neurosciences, University of Geneva, Switzerland; GIGA-Neurosciences, University of Liège, C.H.U. Sart-Tilman, Liège, Belgium
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Switzerland; Department of Neurology, Geneva University Hospital, Geneva, Switzerland.
| |
Collapse
|
37
|
Wu D, Wu L, An H, Bao H, Guo P, Zhang B, Zheng H, Zhang F, Ge W, Cai Y, Xi Y, Yang X. RanGAP-mediated nucleocytoplasmic transport of Prospero regulates neural stem cell lifespan in Drosophila larval central brain. Aging Cell 2019; 18:e12854. [PMID: 30549175 PMCID: PMC6351831 DOI: 10.1111/acel.12854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/24/2018] [Accepted: 07/30/2018] [Indexed: 11/28/2022] Open
Abstract
By the end of neurogenesis in Drosophila pupal brain neuroblasts (NBs), nuclear Prospero (Pros) triggers cell cycle exit and terminates NB lifespan. Here, we reveal that in larval brain NBs, an intrinsic mechanism facilitates import and export of Pros across the nuclear envelope via a Ran‐mediated nucleocytoplasmic transport system. In rangap mutants, the export of Pros from the nucleus to cytoplasm is impaired and the nucleocytoplasmic transport of Pros becomes one‐way traffic, causing an early accumulation of Pros in the nuclei of the larval central brain NBs. This nuclear Pros retention initiates NB cell cycle exit and leads to a premature decrease of total NB numbers. Our data indicate that RanGAP plays a crucial role in this intrinsic mechanism that controls NB lifespan during neurogenesis. Our study may provide insights into understanding the lifespan of neural stem cells during neurogenesis in other organisms.
Collapse
Affiliation(s)
- Di Wu
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
- College of Life Sciences Zhejiang University Hangzhou China
| | - Litao Wu
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
- College of Life Sciences Zhejiang University Hangzhou China
| | - Huanping An
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
- College of Life Sciences Zhejiang University Hangzhou China
| | - Hongcun Bao
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
- College of Life Sciences Zhejiang University Hangzhou China
| | - Pengfei Guo
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
- College of Life Sciences Zhejiang University Hangzhou China
| | - Bei Zhang
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
- College of Life Sciences Zhejiang University Hangzhou China
| | - Huimei Zheng
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
| | - Fan Zhang
- College of Life Sciences Zhejiang University Hangzhou China
| | - Wanzhong Ge
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
| | - Yu Cai
- Temasek Life Sciences Laboratory National University of Singapore Singapore
- Department of Biological Sciences National University of Singapore Singapore
| | - Yongmei Xi
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
| | - Xiaohang Yang
- Division of Human Reproduction and Developmental Genetics The Women's Hospital School of Medicine Zhejiang University Hangzhou China
- Institute of Genetics Zhejiang University Hangzhou China
- Department of Genetics, School of Medicine Zhejiang University Hangzhou China
- Joint Institute of Genetics and Genomic Medicine between Zhejiang University and University of Toronto, Zhejiang University Hangzhou China
| |
Collapse
|
38
|
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
|
39
|
Averbukh I, Lai SL, Doe CQ, Barkai N. A repressor-decay timer for robust temporal patterning in embryonic Drosophila neuroblast lineages. eLife 2018; 7:38631. [PMID: 30526852 PMCID: PMC6303102 DOI: 10.7554/elife.38631] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 12/10/2018] [Indexed: 11/13/2022] Open
Abstract
Biological timers synchronize patterning processes during embryonic development. In the Drosophila embryo, neural progenitors (neuroblasts; NBs) produce a sequence of unique neurons whose identities depend on the sequential expression of temporal transcription factors (TTFs). The stereotypy and precision of NB lineages indicate reproducible TTF timer progression. We combine theory and experiments to define the timer mechanism. The TTF timer is commonly described as a relay of activators, but its regulatory circuit is also consistent with a repressor-decay timer, where TTF expression begins when its repressor decays. Theory shows that repressor-decay timers are more robust to parameter variations than activator-relay timers. This motivated us to experimentally compare the relative importance of the relay and decay interactions in vivo. Comparing WT and mutant NBs at high temporal resolution, we show that the TTF sequence progresses primarily by repressor-decay. We suggest that need for robust performance shapes the evolutionary-selected designs of biological circuits.
Collapse
Affiliation(s)
- Inna Averbukh
- Department of Molecular Genetics, Weizmann institute of science, Rehovot, Israel
| | - Sen-Lin Lai
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| | - Chris Q Doe
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| | - Naama Barkai
- Department of Molecular Genetics, Weizmann institute of science, Rehovot, Israel
| |
Collapse
|
40
|
Harding K, White K. Drosophila as a Model for Developmental Biology: Stem Cell-Fate Decisions in the Developing Nervous System. J Dev Biol 2018; 6:E25. [PMID: 30347666 PMCID: PMC6315890 DOI: 10.3390/jdb6040025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/25/2022] Open
Abstract
Stem cells face a diversity of choices throughout their lives. At specific times, they may decide to initiate cell division, terminal differentiation, or apoptosis, or they may enter a quiescent non-proliferative state. Neural stem cells in the Drosophila central nervous system do all of these, at stereotypical times and anatomical positions during development. Distinct populations of neural stem cells offer a unique system to investigate the regulation of a particular stem cell behavior, while comparisons between populations can lead us to a broader understanding of stem cell identity. Drosophila is a well-described and genetically tractable model for studying fundamental stem cell behavior and the mechanisms that underlie cell-fate decisions. This review will focus on recent advances in our understanding of the factors that contribute to distinct stem cell-fate decisions within the context of the Drosophila nervous system.
Collapse
Affiliation(s)
- Katherine Harding
- Massachusetts General Hospital Cutaneous Biology Research Center, Harvard Medical School, Boston, MA 02129, USA
| | - Kristin White
- Massachusetts General Hospital Cutaneous Biology Research Center, Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
41
|
Ross J, Kuzin A, Brody T, Odenwald WF. Mutational analysis of a Drosophila neuroblast enhancer governing nubbin expression during CNS development. Genesis 2018; 56:e23237. [PMID: 30005136 PMCID: PMC6175444 DOI: 10.1002/dvg.23237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/07/2018] [Accepted: 06/22/2018] [Indexed: 11/17/2022]
Abstract
While developmental studies of Drosophila neural stem cell lineages have identified transcription factors (TFs) important to cell identity decisions, currently only an incomplete understanding exists of the cis‐regulatory elements that control the dynamic expression of these TFs. Our previous studies have identified multiple enhancers that regulate the POU‐domain TF paralogs nubbin and pdm‐2 genes. Evolutionary comparative analysis of these enhancers reveals that they each contain multiple conserved sequence blocks (CSBs) that span TF DNA‐binding sites for known regulators of neuroblast (NB) gene expression in addition to novel sequences. This study functionally analyzes the conserved DNA sequence elements within a NB enhancer located within the nubbin gene and highlights a high level of complexity underlying enhancer structure. Mutational analysis has revealed CSBs that are important for enhancer activation and silencing in the developing CNS. We have also observed that adjusting the number and relative positions of the TF binding sites within these CSBs alters enhancer function.
Collapse
Affiliation(s)
- Jermaine Ross
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland
| | - Alexander Kuzin
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland
| | - Thomas Brody
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland
| | - Ward F Odenwald
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland
| |
Collapse
|
42
|
Abstract
Biology is dynamic. Timescales range from frenetic sub-second ion fluxes and enzymatic reactions to the glacial millions of years of evolutionary change. Falling somewhere in the middle of this range are the processes we usually study in development: cell division and differentiation, gene expression, cell-cell signalling, and morphogenesis. But what sets the tempo and manages the order of developmental events? Are the order and tempo different between species? How is the sequence of multiple events coordinated? Here, we discuss the importance of time for developing embryos, highlighting the necessity for global as well as cell-autonomous control. New reagents and tools in imaging and genomic engineering, combined with in vitro culture, are beginning to offer fresh perspectives and molecular insight into the origin and mechanisms of developmental time.
Collapse
Affiliation(s)
- Miki Ebisuya
- RIKEN Center for Biosystems Dynamics Research (RIKEN BDR), 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - James Briscoe
- The Francis Crick Institute, 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
|
Bothma JP, Norstad MR, Alamos S, Garcia HG. LlamaTags: A Versatile Tool to Image Transcription Factor Dynamics in Live Embryos. Cell 2018; 173:1810-1822.e16. [PMID: 29754814 DOI: 10.1016/j.cell.2018.03.069] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/28/2018] [Accepted: 03/27/2018] [Indexed: 11/18/2022]
Abstract
Embryonic cell fates are defined by transcription factors that are rapidly deployed, yet attempts to visualize these factors in vivo often fail because of slow fluorescent protein maturation. Here, we pioneer a protein tag, LlamaTag, which circumvents this maturation limit by binding mature fluorescent proteins, making it possible to visualize transcription factor concentration dynamics in live embryos. Implementing this approach in the fruit fly Drosophila melanogaster, we discovered stochastic bursts in the concentration of transcription factors that are correlated with bursts in transcription. We further used LlamaTags to show that the concentration of protein in a given nucleus heavily depends on transcription of that gene in neighboring nuclei; we speculate that this inter-nuclear signaling is an important mechanism for coordinating gene expression to delineate straight and sharp boundaries of gene expression. Thus, LlamaTags now make it possible to visualize the flow of information along the central dogma in live embryos.
Collapse
Affiliation(s)
- Jacques P Bothma
- Department of Molecular & Cell Biology, UC Berkeley, Berkeley, CA 94720, USA
| | - Matthew R Norstad
- Department of Molecular & Cell Biology, UC Berkeley, Berkeley, CA 94720, USA
| | - Simon Alamos
- Department of Plant and Microbial Biology, UC Berkeley, Berkeley, CA 94720, USA
| | - Hernan G Garcia
- Department of Molecular & Cell Biology, UC Berkeley, Berkeley, CA 94720, USA; Department of Physics, UC Berkeley, Berkeley, CA 94720, USA; Biophysics Graduate Group, UC Berkeley, Berkeley, CA 94720, USA; Institute for Quantitative Biosciences-QB3, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
45
|
Álvarez JA, Díaz-Benjumea FJ. Origin and specification of type II neuroblasts in the Drosophila embryo. Development 2018; 145:dev.158394. [PMID: 29567672 DOI: 10.1242/dev.158394] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 03/12/2018] [Indexed: 12/16/2022]
Abstract
In Drosophila, neural stem cells or neuroblasts (NBs) acquire different identities according to their site of origin in the embryonic neuroectoderm. Their identity determines the number of times they will divide and the types of daughter cells they will generate. All NBs divide asymmetrically, with type I NBs undergoing self-renewal and generating another cell that will divide only once more. By contrast, a small set of NBs in the larval brain, type II NBs, divides differently, undergoing self-renewal and generating an intermediate neural progenitor (INP) that continues to divide asymmetrically several more times, generating larger lineages. In this study, we have analysed the origin of type II NBs and how they are specified. Our results indicate that these cells originate in three distinct clusters in the dorsal protocerebrum during stage 12 of embryonic development. Moreover, it appears that their specification requires the combined action of EGFR signalling and the activity of the related genes buttonhead and Drosophila Sp1 In addition, we also show that the INPs generated in the embryo enter quiescence at the end of embryogenesis, resuming proliferation during the larval stage.
Collapse
Affiliation(s)
- José-Andrés Álvarez
- Centro de Biología Molecular-Severo Ochoa (CSIC-UAM), c/ Nicolas Cabrera 1, Universidad Autónoma, 28049 Madrid, Spain
| | - Fernando J Díaz-Benjumea
- Centro de Biología Molecular-Severo Ochoa (CSIC-UAM), c/ Nicolas Cabrera 1, Universidad Autónoma, 28049 Madrid, Spain
| |
Collapse
|
46
|
Torres-Oliva M, Schneider J, Wiegleb G, Kaufholz F, Posnien N. Dynamic genome wide expression profiling of Drosophila head development reveals a novel role of Hunchback in retinal glia cell development and blood-brain barrier integrity. PLoS Genet 2018; 14:e1007180. [PMID: 29360820 PMCID: PMC5796731 DOI: 10.1371/journal.pgen.1007180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 02/02/2018] [Accepted: 01/01/2018] [Indexed: 01/01/2023] Open
Abstract
Drosophila melanogaster head development represents a valuable process to study the developmental control of various organs, such as the antennae, the dorsal ocelli and the compound eyes from a common precursor, the eye-antennal imaginal disc. While the gene regulatory network underlying compound eye development has been extensively studied, the key transcription factors regulating the formation of other head structures from the same imaginal disc are largely unknown. We obtained the developmental transcriptome of the eye-antennal discs covering late patterning processes at the late 2nd larval instar stage to the onset and progression of differentiation at the end of larval development. We revealed the expression profiles of all genes expressed during eye-antennal disc development and we determined temporally co-expressed genes by hierarchical clustering. Since co-expressed genes may be regulated by common transcriptional regulators, we combined our transcriptome dataset with publicly available ChIP-seq data to identify central transcription factors that co-regulate genes during head development. Besides the identification of already known and well-described transcription factors, we show that the transcription factor Hunchback (Hb) regulates a significant number of genes that are expressed during late differentiation stages. We confirm that hb is expressed in two polyploid subperineurial glia cells (carpet cells) and a thorough functional analysis shows that loss of Hb function results in a loss of carpet cells in the eye-antennal disc. Additionally, we provide for the first time functional data indicating that carpet cells are an integral part of the blood-brain barrier. Eventually, we combined our expression data with a de novo Hb motif search to reveal stage specific putative target genes of which we find a significant number indeed expressed in carpet cells. The development of different cell types must be tightly coordinated, and the eye-antennal imaginal discs of Drosophila melanogaster represent an excellent model to study the molecular mechanisms underlying this coordination. These imaginal discs contain the anlagen of nearly all adult head structures, such as the antennae, the head cuticle, the ocelli and the compound eyes. While large scale screens have been performed to unravel the gene regulatory network underlying compound eye development, a comprehensive understanding of genome wide expression dynamics throughout head development is still missing to date. We studied the genome wide gene expression dynamics during eye-antennal disc development in D. melanogaster to identify new central regulators of the underlying gene regulatory network. Expression based gene clustering and transcription factor motif enrichment analyses revealed a central regulatory role of the transcription factor Hunchback (Hb). We confirmed that hb is expressed in two polyploid retinal subperineurial glia cells (carpet cells). Our functional analysis shows that Hb is necessary for carpet cell development and we show for the first time that the carpet cells are an integral part of the blood-brain barrier.
Collapse
Affiliation(s)
- Montserrat Torres-Oliva
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Julia Schneider
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Gordon Wiegleb
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Felix Kaufholz
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
| | - Nico Posnien
- Universität Göttingen, Johann-Friedrich-Blumenbach-Institut für Zoologie und Anthropologie, Abteilung für Entwicklungsbiologie, GZMB Ernst-Caspari-Haus, Göttingen, Germany
- * E-mail:
| |
Collapse
|
47
|
Perry M, Konstantinides N, Pinto-Teixeira F, Desplan C. Generation and Evolution of Neural Cell Types and Circuits: Insights from the Drosophila Visual System. Annu Rev Genet 2017; 51:501-527. [PMID: 28961025 DOI: 10.1146/annurev-genet-120215-035312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The Drosophila visual system has become a premier model for probing how neural diversity is generated during development. Recent work has provided deeper insight into the elaborate mechanisms that control the range of types and numbers of neurons produced, which neurons survive, and how they interact. These processes drive visual function and influence behavioral preferences. Other studies are beginning to provide insight into how neuronal diversity evolved in insects by adding new cell types and modifying neural circuits. Some of the most powerful comparisons have been those made to the Drosophila visual system, where a deeper understanding of molecular mechanisms allows for the generation of hypotheses about the evolution of neural anatomy and function. The evolution of new neural types contributes additional complexity to the brain and poses intriguing questions about how new neurons interact with existing circuitry. We explore how such individual changes in a variety of species might play a role over evolutionary timescales. Lessons learned from the fly visual system apply to other neural systems, including the fly central brain, where decisions are made and memories are stored.
Collapse
Affiliation(s)
- Michael Perry
- Department of Biology, New York University, New York, NY 10003, USA;
| | | | - Filipe Pinto-Teixeira
- Department of Biology, New York University, New York, NY 10003, USA; .,Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Claude Desplan
- Department of Biology, New York University, New York, NY 10003, USA; .,Center for Genomics and Systems Biology, New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| |
Collapse
|
48
|
Syed MH, Mark B, Doe CQ. Playing Well with Others: Extrinsic Cues Regulate Neural Progenitor Temporal Identity to Generate Neuronal Diversity. Trends Genet 2017; 33:933-942. [PMID: 28899597 DOI: 10.1016/j.tig.2017.08.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 11/27/2022]
Abstract
During neurogenesis, vertebrate and Drosophila progenitors change over time as they generate a diverse population of neurons and glia. Vertebrate neural progenitors have long been known to use both progenitor-intrinsic and progenitor-extrinsic cues to regulate temporal patterning. In contrast, virtually all temporal patterning mechanisms discovered in Drosophila neural progenitors (neuroblasts) involve progenitor-intrinsic temporal transcription factor cascades. Recent results, however, have revealed several extrinsic pathways that regulate Drosophila neuroblast temporal patterning: nutritional cues regulate the timing of neuroblast proliferation/quiescence and a steroid hormone cue that is required for temporal transcription factor expression. Here, we discuss newly discovered extrinsic cues regulating neural progenitor temporal identity in Drosophila, highlight conserved mechanisms, and raise open questions for the future.
Collapse
Affiliation(s)
- Mubarak Hussain Syed
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Brandon Mark
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Chris Q Doe
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
49
|
Syed MH, Mark B, Doe CQ. Steroid hormone induction of temporal gene expression in Drosophila brain neuroblasts generates neuronal and glial diversity. eLife 2017; 6:26287. [PMID: 28394252 PMCID: PMC5403213 DOI: 10.7554/elife.26287] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 04/09/2017] [Indexed: 12/14/2022] Open
Abstract
An important question in neuroscience is how stem cells generate neuronal diversity. During Drosophila embryonic development, neural stem cells (neuroblasts) sequentially express transcription factors that generate neuronal diversity; regulation of the embryonic temporal transcription factor cascade is lineage-intrinsic. In contrast, larval neuroblasts generate longer ~50 division lineages, and currently only one mid-larval molecular transition is known: Chinmo/Imp/Lin-28+ neuroblasts transition to Syncrip+ neuroblasts. Here we show that the hormone ecdysone is required to down-regulate Chinmo/Imp and activate Syncrip, plus two late neuroblast factors, Broad and E93. We show that Seven-up triggers Chinmo/Imp to Syncrip/Broad/E93 transition by inducing expression of the Ecdysone receptor in mid-larval neuroblasts, rendering them competent to respond to the systemic hormone ecdysone. Importantly, late temporal gene expression is essential for proper neuronal and glial cell type specification. This is the first example of hormonal regulation of temporal factor expression in Drosophila embryonic or larval neural progenitors.
Collapse
Affiliation(s)
- Mubarak Hussain Syed
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| | - Brandon Mark
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| | - Chris Q Doe
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, United States
| |
Collapse
|
50
|
Hirono K, Kohwi M, Clark MQ, Heckscher ES, Doe CQ. The Hunchback temporal transcription factor establishes, but is not required to maintain, early-born neuronal identity. Neural Dev 2017; 12:1. [PMID: 28137283 PMCID: PMC5282720 DOI: 10.1186/s13064-017-0078-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/26/2017] [Indexed: 01/01/2023] Open
Abstract
Background Drosophila and mammalian neural progenitors typically generate a diverse family of neurons in a stereotyped order. Neuronal diversity can be generated by the sequential expression of temporal transcription factors. In Drosophila, neural progenitors (neuroblasts) sequentially express the temporal transcription factors Hunchback (Hb), Kruppel, Pdm, and Castor. Hb is necessary and sufficient to specify early-born neuronal identity in multiple lineages, and is maintained in the post-mitotic neurons produced during each neuroblast expression window. Surprisingly, nothing is currently known about whether Hb acts in neuroblasts or post-mitotic neurons (or both) to specify first-born neuronal identity. Methods Here we selectively remove Hb from post-mitotic neurons, and assay the well-characterized NB7-1 and NB1-1 lineages for defects in neuronal identity and function. Results We find that loss of Hb from embryonic and larval post-mitotic neurons does not affect neuronal identity. Furthermore, removing Hb from post-mitotic neurons throughout the entire CNS has no effect on larval locomotor velocity, a sensitive assay for motor neuron and pre-motor neuron function. Conclusions We conclude that Hb functions in progenitors (neuroblasts/GMCs) to establish heritable neuronal identity that is maintained by a Hb-independent mechanism. We suggest that Hb acts in neuroblasts to establish an epigenetic state that is permanently maintained in early-born neurons. Electronic supplementary material The online version of this article (doi:10.1186/s13064-017-0078-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Keiko Hirono
- Howard Hughes Medical Institute, Eugene, 97403, USA.,Institute of Molecular Biology, Eugene, 97403, USA.,Institute of Neuroscience, University of Oregon, Eugene, 97403, USA
| | - Minoree Kohwi
- Department of Neuroscience, Columbia University Medical Center, New York, NY, 10032, USA
| | - Matt Q Clark
- Howard Hughes Medical Institute, Eugene, 97403, USA.,Institute of Molecular Biology, Eugene, 97403, USA.,Institute of Neuroscience, University of Oregon, Eugene, 97403, USA
| | - Ellie S Heckscher
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Chris Q Doe
- Howard Hughes Medical Institute, Eugene, 97403, USA. .,Institute of Molecular Biology, Eugene, 97403, USA. .,Institute of Neuroscience, University of Oregon, Eugene, 97403, USA.
| |
Collapse
|