Temporal Progression of Drosophila Neural Stem Cell Promoting Neuronal Diversity by Noah Robert Dillon A dissertation accepted and approved in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biology Dissertation Committee: Dr. Adam Miller, Chair Dr. Chris Doe, Advisor Dr. Tory Herman, Core Member Dr. Kryn Stankunas, Core Member Dr. Matt Smear, Institutional Representative University of Oregon Fall 2024 2 © 2024 Noah Robert Dillon This work is openly licensed via CC BY-NC 4.0. 3 Dissertation Abstract Noah Robert Dillon Doctor of Philosophy in Biology Title: Temporal Progression of Drosophila Neural Stem Cells Promoting Neuronal Diversity How are complex nervous systems generated? During development, a small pool of neural stem cells generates a diverse array of cell type diversity that forms a functional brain. Remarkably, this neuronal diversity is generated in a predictable order. In this dissertation, I report my work in understanding how neural stem cells of the developing Drosophila melanogaster, known as neuroblasts, are temporally patterned. My work has established a single-cell RNA sequencing atlas of the early larval stages of neurogenesis that identified key regulators of how neuroblasts progress from a quiescent to a proliferative state. My subsequent studies focused on neuroblast lineages that generate the central brain of the adult. I show that the transcription factor Seven-up is required for switching the production of early to late neuron identities and progressing Type 2 neuroblasts to the end of their lineage (i.e. death). Finally, I show the temporal transcription factor Castor is required for specifying neuron identities born in early larval Type 2 neuroblast lineages. My work shows significant advancements in understanding how the fly brain is generated and provides fruitful future directions to pursue. This dissertation includes previously published and unpublished co-authored material. 4 Curriculum vitae Name of author: Noah Robert Dillon Graduate and undergraduate schools attended: University of Oregon, Eugene, OR University of Puget Sound, Tacoma, WA Degrees awarded: Doctor of Philosophy, Biology, 2024, University of Oregon Bachelor of Science, Biology, 2020, University of Puget Sound Areas of special interest: Developmental Biology Neurogenetics Stem cells Professional experience: Graduate Researcher, University of Oregon, 2021-2024 Laboratory of Dr. Chris Q. Doe Graduate Employee – Teaching Assistant, University of Oregon, 2020-2021 Associate Editor – Elements magazine, University of Puget Sound 2019-2020 Lab Accessibility and Biology Instructor Assistant, University of Puget Sound 2018-2020 Undergraduate Research Intern, University of Puget Sound, 2017-2020 Laboratory of Dr. Oscar Sosa, 2019-2020 Laboratory of Dr. Siddhartha Ramakrishnan, 2018-2019 Laboratory of Dr. Alyce DeMarais, 2017-2019 Museum Docent, Puget Sound Natural History Museum, 2016-2020 Director Dr. Peter Wimberger Grants, awards, and honors: 5 Talk Platform Prize, Seven-up acts in neuroblasts to specify adult Drosophila central complex neuron identity and initiate neuroblast decommissioning, Society for Developmental Biology, 2024 Developmental Biology Training Grant, University of Oregon / National Institute of Child Health and Human Disease, 2021-2024 University Enrichment Committee Funds for Research, University of Puget Sound, 2018 Mellam Independent Research Scholarship, University of Puget Sound, 2018 LIASE: Biodiversity in Borneo, Henry Luce Foundation, 2018 Publications Dillon, N. R. and Doe, C. Q. (2024). Castor is a temporal transcription factor that specifies early born central complex neuron identity. 2024.08.22.609207. Under review at Development. Dillon, N. R., Manning, L., Hirono, K. and Doe, C. Q. (2024). Seven-up acts in neuroblasts to specify adult central complex neuron identity and initiate neuroblast decommissioning. Development 151, dev202504. Epiney, D., Chaya, G. N. M., Dillon, N. R., Lai, S.-L. and Doe, C. Q. (2023). Transcriptional complexity in the insect central complex: single nuclei RNA sequencing of adult brain neurons derived from type 2 neuroblasts. bioRxiv. 2023.12.10.571022. Dillon, N., Cocanougher, B., Sood, C., Yuan, X., Kohn, A. B., Moroz, L. L., Siegrist, S. E., Zlatic, M. and Doe, C. Q. (2022). Single cell RNA-seq analysis reveals temporally- regulated and quiescence-regulated gene expression in Drosophila larval neuroblasts. Neural Development 17, 7. 6 Acknowledgements My adventure into the sciences has been an unexpected but marvelous journey. My original inspiration for pursing biology came from reading books by Sean B. Carroll and Richard Dawkins while ditching school in rural Wyoming. I have my high school science teachers to thank for keeping me from dropping all my classes. I am grateful to have found a home in the Pacific Northwest for the past eight years attending the University of Puget Sound and then the University of Oregon. My mentors during undergrad were gracious enough to steer me towards a path in scientific research. I am indebted to Dr. Alyce DeMarais, Dr. Siddhartha Ramakrishnan, Dr. Oscar Sosa, Dr. Peter Wimberger, and Dr. Joel Elliott (just to name a few). I thank Dr. Chris Q. Doe for his mentorship and guidance during my dissertation as I have worked in his lab pursing projects built on a passion for understanding developmental biology. The Doe Lab members (Sarah Ackerman, Nathan Anderson, Elena Barth, Ben Brissette, Arnaldo Carreira-Rosario, Kasey Drake, Derek Epiney, Katie Fisher, Josmarie Graciani, Janet Hanawalt, Emily Heckman, Keiko Hirono, Sen-Lin Lai, Kristen Lee, Laurina Manning, Gonzalo Morales Chaya, Jordan Munroe, Peter Newstein, Heather Pollington, Megan Radler, Tyler Ramos, Natalie Rico Carvajal, Rishi Sastry, Austin Seroka, Alanna Sowles, and Chundi Xu) have, over the past four years, been an incredible support structure that I have cherished in helping me produce work that I am proud to publish. I also thank Dr. Adam Miller, and the Miller Lab, for guidance when I first joined the program in setting me up for success the moment I started at the University of Oregon. I extend gratitude to the rest of my dissertation advisory committee members Dr. Tory Herman, Dr. Matt Smear, and Dr. Kryn Stankunas for their guidance. For the work in Chapter II, I thank Dr. Sen-Lin Lai, Dr. Cheng-Yu, and Dr. Sarah Ackerman for comments on the manuscript. Funding for this work was provided by NIH Training Grant 5-T32- HD07348, NIH HD27056, and Howard Hughes Medical Institute (Noah Dillon and Chris Doe). I thank my co-authors: Ben Cocanougher, Chhavi Sood, Xin Yuan, Andrea Kohn, Leonid Moroz, Dr. Sarah Siegrist, and Dr. Marta Zlatic for their help in making this work possible. Funding in part was also covered by NIH R35 GM141886 (Sarah Siegrist). 7 For work in Chapter III, I thank Dr. Kristen Lee, Peter Newstein, Dr. Megan Radler, and Dr. Chundi Xu for internal comments on the manuscript. I also thank Dr. Josie Clowny, Dr. Tzumin Lee and Dr. Mubarak Syed for feedback on the project. Funding for this work was provided by NIH Training Grant 5-T32-HD07348, NIH HD27056, and Howard Hughes Medical Institute (Noah Dillon and Chris Doe). I thank my co-authors Laurina Manning and Keiko Hirono as it was a privilege to work with these gifted scientists. For work in Chapter IV, I thank Dr. Tory Herman, Derek Epiney and Gonzalo Morales Chaya for comments on the manuscript. I thank Laurina Manning and Jordan Munroe for assistance on the EdU experiments and Dr. Sen-Lin Lai for advice on antibodies. Funding for this work was provided by NIH Training Grant 5-T32-HD07348, NIH HD27056, and Howard Hughes Medical Institute. I thank Chris Doe for motivating me in getting this last paper ready in record time. I would like to acknowledge the resource centers that make doing high quality science possible: FlyBase (http://flybase.org/) for a wonderful repository of Drosophila genetic information; Developmental Studies Hybridoma Bank, created by the NICHD of the NIH and maintained at the University of Iowa, Department of Biology, Iowa City, IA for antibodies; Bloomington Drosophila Stock Center (NIH P40OD018537); and Vienna Drosophila Resource Center for fly stocks; Institute of Molecular Biology Media Room at the University of Oregon for prepping the abundance of fly food used to maintain stocks and for cleaning the lab glassware. This work was only made possible by the contributions of many in supporting the pursuit of knowledge. I will forever be grateful and indebted to the people who have shown me kindness and respect through my scientific career. 8 Table of Contents Chapter Page I. An introduction to generating neuronal diversity and a complex brain in Drosophila ................................................................................................. 17 Patterning during Drosophila neurogenesis creates neural diversity ..................... 17 An introduction to neuroblasts and the developing nervous system ................ 17 The discovery of temporal transcription factors .............................................. 20 Temporal progression in the larval neuroblasts ............................................... 23 Establishing complexity within the Drosophila central brain ............................... 27 The diversity and function of the Central Complex ......................................... 27 Tools for accessing the Central Complex ........................................................ 30 Bridge ..................................................................................................................... 32 References .............................................................................................................. 33 II. A single-cell RNA-seq analysis reveals temporally-regulated and quiescence-regulated gene expression in larval neuroblasts ............................ 44 Author contributions .............................................................................................. 44 Introduction ............................................................................................................ 44 Results .................................................................................................................... 45 Larval atlas shows distinct cell identities and differentiating neural progenitor axis ................................................................................................. 45 Quiescent neuroblasts and associated glia are enriched for expression of genes regulating the TOR and insulin pathways ......................................... 49 Proliferating neuroblasts shows candidate novel markers and temporal transcription factors ......................................................................................... 52 INPs express candidate novel cell type markers .............................................. 56 GMCs, new‐born neurons and immature neurons express candidate novel cell type markers .................................................................................... 58 Mature neurons show temporally distinct groups of transcription factors and cell surface molecules ................................................................... 61 Discussion .............................................................................................................. 63 Quiescent neuroblasts and glial signaling ........................................................ 64 9 TTFs in type I and type II neuroblasts ............................................................. 65 Intermediate neural progenitors ....................................................................... 65 The transition from progenitor to post‐mitotic neurons ................................... 66 Mature neurons show novel temporal changes ................................................ 66 Conclusions ............................................................................................................ 67 Materials and methods .......................................................................................... 67 Single cell isolation and sequencing ................................................................ 67 scRNA‐seq analysis ......................................................................................... 68 Subclustering for further Seurat analysis ......................................................... 69 Data and code availability ................................................................................ 69 Protein localization .......................................................................................... 69 Supplementary information ............................................................................. 70 Bridge ..................................................................................................................... 71 References .............................................................................................................. 72 III. Seven-up acts in neuroblasts to specify adult central complex neuron identity and initiate neuroblast decommissioning ............................................ 81 Author contributions .............................................................................................. 81 Introduction ............................................................................................................ 81 Results .................................................................................................................... 84 Columnar neurons P-EN and P-FN are born from larval T2NBs in different temporal windows ......................................................................... 84 Svp is expressed transiently and asynchronously in all larval T2NB lineages between 18 h and 24 h ALH .............................................................. 85 Cut expression distinguishes molecular identities of adult P-EN neurons from P-FN neurons ............................................................................. 88 Loss of Svp decreases the number of late born P-FN adult neurons ............... 89 Loss of Svp extends the production of early born P-EN adult neurons ........... 91 Loss of Svp extends T2NB lineages into the adult .......................................... 95 Discussion .............................................................................................................. 97 Columnar neurons are born at different times in the T2NB lineage ................ 97 Svp expression in larval T2NB lineages .......................................................... 98 10 Svp is required for late born fates in the T2NB lineages ................................. 98 Svp specification of other CX neuron subtypes ............................................... 99 Svp-mediated regulation of Type 2 neuroblast temporal progression ............. 100 Conserved role of Svp in vertebrate temporal patterning ................................ 101 Materials and methods ........................................................................................... 101 Animal preparation .......................................................................................... 101 EdU experiments .............................................................................................. 102 Larval experiments ........................................................................................... 102 Adult experiments ............................................................................................ 102 Hybridization chain reaction (HCR) RNA fluorescent in situ hybridization .................................................................................................... 102 Larval brain sample preparation ...................................................................... 102 Adult brain sample preparation ........................................................................ 103 EdU adult brain sample preparation ................................................................ 103 Antibodies ........................................................................................................ 103 Confocal microscopy ....................................................................................... 103 Image processing and analysis ......................................................................... 103 Cell counting and neuropil target scoring ............................................ 104 Imaris neuropil reconstructions ............................................................ 104 Figure preparation ................................................................................ 104 Statistical analyses ........................................................................................... 104 Data availability ............................................................................................... 105 Supplementary information ............................................................................. 105 Bridge ..................................................................................................................... 106 References .............................................................................................................. 107 IV. Castor is a temporal transcription factor that specifies early born central complex neuron identity ......................................................................... 113 Author contributions .............................................................................................. 113 Introduction ............................................................................................................ 113 Results .................................................................................................................... 116 E-PG neurons are born early in Type 2 neuroblast lineages ........................... 116 11 E-PG and P-EN neurons have distinct molecular identities ............................ 117 Seven-up is required to restrict E-PG production in early Type 2 neuroblast lineages .......................................................................................... 118 Castor expression is transient in the Type 2 larval neuroblast lineages ......... 120 Generating Type 2 lineage specific Castor knockout and misexpression lines .......................................................................................... 122 Castor is required to specify early born P-EN and E-PG adult neuron molecular identities .......................................................................................... 123 Castor is sufficient to produce ectopic adult P-EN neurons but not E-PG neurons ................................................................................................... 125 Discussion .............................................................................................................. 127 Castor expression in larval Type 2 neuroblast lineages .................................. 127 Castor is a narrowly expressed early temporal transcription factor in larval Type 2 lineages ...................................................................................... 128 Columnar neurons born in the same Type 2 neuroblast window have distinct adult molecular identities .................................................................... 129 Conserved role of Castor in vertebrate neurogenesis ...................................... 129 Materials and methods ........................................................................................... 130 Animal preparation .......................................................................................... 130 EdU experiment ............................................................................................... 130 Larval experiments........................................................................................... 130 Adult experiments............................................................................................. 130 Immunohistochemistry ..................................................................................... 131 Larval brain sample preparation ..................................................................... 131 Adult brain sample preparation ....................................................................... 131 EdU adult brain sample preparation ............................................................... 131 Confocal microscopy ........................................................................................ 132 Image processing and analysis ........................................................................ 132 Figure preparation ............................................................................... 132 Statistical analyses ............................................................................... 132 Supplementary information .............................................................................. 132 12 Data availability .............................................................................................. 133 Bridge ..................................................................................................................... 134 References .............................................................................................................. 135 V. Discussion ............................................................................................................... 140 Future directions in understanding neuroblast quiescence and reactivation .......... 140 Insulin signaling and cell cycle in quiescence ................................................. 140 Connecting quiescence to the temporal cascade ............................................. 142 Future directions in understanding larval neuroblast temporal progression .......... 143 Extrinsic signaling over time ........................................................................... 143 Switching factors and the shifting of competence ............................................ 146 Temporal factors and the Central Complex ..................................................... 147 Conservation of temporal patterning mechanisms in Drosophila and mammalian central nervous systems ..................................................................... 152 An introduction to mammalian neural stem cells that generate the cortex ..... 153 A brief introduction to mammalian retinal progenitor cells ............................ 165 A comparison between the fly and mammalian neurogenesis ......................... 165 Concluding remarks ............................................................................................... 158 References .............................................................................................................. 159 Appendices .................................................................................................................. 169 A. Chapter III supplementary figures and tables ................................................... 169 B. Chapter IV supplementary figures and tables ................................................... 178 13 List of Figures Figure Page 1.1 Spatial patterning of neuroblasts in a single hemisegment of the developing Drosophila embryonic ventral nerve cord ............................................................. 17 1.2 Division patterns of Drosophila neuroblast lineages ............................................. 18 1.3 Spatial positioning of Type 2 neuroblasts and cell lineage markers ...................... 19 1.4 The canonical temporal transcription factor cascade of embryonic Type 1 neuroblasts in the ventral nerve cord ......................................................... 21 1.5 The candidate temporal transcription factor cascade of larval Type 2 intermediate neural progenitors ............................................................................. 22 1.6 Neuroblast progression of neurogenesis across fly development .......................... 23 1.7 Temporal patterning of the Type 2 neuroblast in larval stages .............................. 25 1.8 The adult Central Complex neuropils and neurons ................................................ 27 2.1 Larval atlas shows distinct cell identities and differentiating neural progenitor axis ....................................................................................................... 46 2.2 Quiescent neuroblasts and glial cells show enriched markers for regulating the TOR and insulin pathway ............................................................... 50 2.3 Type I neuroblasts shows candidate novel markers and temporal transcription factors ............................................................................................... 53 2.4 Type II progenitor cluster contains type II neuroblasts that show candidate temporal transcription factors ................................................................ 68 2.5 INPs show candidate novel markers ...................................................................... 57 2.6 GMCs, new-born neurons and immature neurons show candidate novel markers ......................................................................................................... 59 2.7 Mature neuron conclusion ...................................................................................... 62 3.1 Columnar neuron subtypes are born from larval T2NBs at distinct temporal windows .................................................................................................. 82 3.2 Svp is expressed early in all larval T2NB lineages ................................................ 86 3.3 Cut expression distinguishes P-EN and P-FN molecular identities ....................... 89 3.4 Svp is required for the late born P-FN neuron identity .......................................... 90 3.5 Svp restricts the early born P-EN neuron molecular identity and 14 birth window .......................................................................................................... 92 3.6 Svp regulates the early born P-EN adult neuron morphology ............................... 94 3.7 Svp is required for timely onset of T2NB decommissioning ................................ 96 4.1 E-PG neurons are born early in Type 2 neuroblast lineages .................................. 115 4.2 E-PG and P-EN neurons have distinct molecular identities .................................. 118 4.3 Seven-up is required to restrict E-PG production in early Type 2 neuroblast lineages ................................................................................................. 119 4.4 Castor expression is transient in Type 2 neuroblast lineages ................................. 121 4.5 Castor is required to specify early born P-EN and E-PG adult neuron molecular identities ................................................................................................ 124 4.6 Castor is sufficient to produce ectopic adult P-EN neurons but not EPG neurons .......................................................................................................... 126 5.1 Model of temporal progression in larval neuroblasts by regulation from extrinsic signaling pathways. ....................................................... 145 5.2 Model for Type 2 neuroblast generation of Central Complex neuron identities ..................................................................................................... 149 5.3 Current understanding of temporal patterning in the Type 2 lineages generating Central Complex neurons ..................................................................... 151 S3.1 Svp mRNA is expressed early in all larval T2NB lineages similar to protein expression ............................................................................................ 170 S3.2 Svp CRISPR/Cas9 knockout Svp in T2NBs and prevents temporal expression of the late factor E93 .......................................................................... 171 S3.3 Adult P-EN and P-FN neurons do not express Svp ............................................. 172 S3.4 Svp in T2NBs regulates adult CX neuropil development ................................... 174 S4.1 Loss of Seven-up in Type 2 neuroblasts leads to altered E-PG ellipsoid body morphology ................................................................................. 178 S4.2 Generating Type 2 lineage specific Castor knockout and misexpression lines ............................................................................................. 179 S4.3 Castor and Seven-up do not cross regulate in Type 2 neuroblasts ....................... 180 15 List of Tables Table Page 1.1 Markers distinguishing the Type 2 neuroblast lineage progenitor cell types ......... 20 2.1 Validated markers for progenitors and young neurons .......................................... 48 2.2 Validated markers for glial cell types ..................................................................... 48 2.3 Validated markers for mature neuron cell types .................................................... 49 S3.1 Transgenes and Drosophila melanogaster stock lines used in Dillon et al., 2024................................................................................................ 175 S3.2 Genetic crosses for each experiment in Dillon et al., 2024 ................................. 176 S3.3 Antibodies used in Dillon et al., 2024.................................................................. 177 S4.1 Transgenes and Drosophila melanogaster stock lines used in Dillon and Doe 2024 ........................................................................................... 181 S4.2 Genetic crosses for each experiment in Dillon and Doe 2024 ............................. 182 S4.3 Antibodies used in Dillon and Doe 2024. ............................................................ 184 16 Chapter I An introduction to generating neuronal diversity and a complex brain in Drosophila Patterning during Drosophila neurogenesis creates neural diversity Animal behaviors require complex neural circuits, which requires a diverse set of neuronal cell types, to function. An open question in developmental neurobiology remains: How are diverse neurons generated from a small pool of neural stem cells? The primary focus of this chapter will be on the developing Drosophila central nervous system. The secondary focus will shift briefly to the matured adult central brain and how it is generated during development. Primary attention is placed on the developing ventral nerve cord and brain lobes with brief mentions of the optic lobes when relevant. The following chapters will cover my contributions to the field. The last chapter will discuss future directions and connect work done in Drosophila to mammalian neural development. An introduction to neuroblasts and the developing nervous system The neural stem cells in Drosophila are known as neuroblasts (NBs). NBs delaminate from the neuroectoderm with each NB having a distinct identity (Broadus et al., 1995; Doe, 1992; Hartenstein and Campos-Ortega, 1984; Wheeler, 1891; Wheeler, 1893) (reviewed in: Hartenstein and Wodarz, 2013; Skeath and Thor, 2003). Early lineage tracing studies show that each NB generates distinct and reproducible cell lineages (Bossing et al., 1996; Doe, 1992; Schmid et al., 1999; Schmidt et al., 1997). The identity of each NB in the ventral nerve cord is controlled by spatial factors that impart a unique molecular profile based on a ‘column and row’ logic (reviewed in Skeath and Thor, 2003) (Fig 1.1). For example, the row factor Gsb is required for row 5 NB identities (Bhat, 1996; Skeath et al., 1995) (Fig 1.1). The column factor Msh is required for the lateral most column of NB identities (Isshiki et al., 1997) (Fig 1.1). Recent work has shown these combinations of spatial factors to establish NB identity-specific chromatin accessibility (Sen et al., 2019). It remains to be seen if this NB identity chromatin profile is required for establishing unique cell lineages. Similar spatial patterning mechanisms have been seen in the optic lobe NBs (reviewed Rossi et al., 2021). These findings established the first level of generating diverse neuronal cell types in that each NB has a unique identity and produces a 17 distinct lineage with a defined set of cell types. Fig. 1.1 Spatial patterning of neuroblasts in a single hemisegment of the developing Drosophila embryonic ventral nerve cord. Neuroblasts have unique spatial identities determined by cross-regulating spatial factors in a row and column logic. Row spatial factors include: Hh, En, Wg, and Gsb (Bhat, 1996; Chu-LaGraff and Doe, 1993; Deshpande et al., 2001; Matsuzaki and Saigo, 1996; McDonald and Doe, 1997; Skeath et al., 1995). Column spatial factors include: Vnd, Ind, and Msh (Isshiki et al., 1997; McDonald et al., 1998; von Ohlen and Doe, 2000; Weiss et al., 1998). A, Anterior; P, Posterior; M, Medial; L, Lateral. All NBs undergo asymmetric cell divisions to generate smaller progeny while self-renewing for subsequent divisions. There are three NB division patterns (Fig 1.2). Type 0 NBs produce a progeny cell that directly differentiates into its postmitotic neuronal fate (Baumgardt et al., 2014) (Fig 1.2, left). These Type 0 divisions have not been extensively studied and therefore will not be discussed further. Type 1 NBs (T1NBs) produce a transitional progeny cell type known as a ganglion mother cell (GMC) that will divide once to produce a pair of postmitotic cells, neurons or glia (Fig 1.2, middle). Type 2 NBs (T2NBs) have a more complex division with each NB division generating an intermediate neural progenitor (INP) that will asymmetrically divide to produce 4-6 GMCs that each will divide once to produce a pair of postmitotic cells (Bello et al., 2008; Boone and Doe, 2008; Bowman et al., 2008) (Fig 1.2, right). Thus, T2NBs have more neurogenic potential from a single stem cell as each T2NB division will generate a total of 8-12 postmitotic cells compared to the 2 cells generated each T1NB division. 18 Fig. 1.2 Division patterns of Drosophila neuroblasts. Type 0 division (left) is a mode of direct neurogenesis with the NB producing one progeny cell per division. Type 1 division (middle) is indirect neurogenesis with the NB producing one GMC that will divide once to produce two progeny cells per NB division. Type 2 division (right) has the most neurogenic potential with the production of INPs that produces 4-6 GMCs that in total generate 8-12 progeny cells per T2NB division. NB, Neuroblast; GMC, Ganglion Mother Cell; INP, intermediate neural progenitor. T1NBs have been extensively studied as they comprise the majority of NB lineages in the central nervous system. The ventral nerve cord is divided into 14 repeated segments known are hemisegments that are bilaterally symmetrical across the midline and contain ~30 NBs each for a total of ~420 NBs per ventral nerve cord (Broadus et al., 1995; Hartenstein and Campos-Ortega, 1984; Hartenstein et al., 1994). The central brain lobes contain ~100 T1NBs (Pereanu and Hartenstein, 2006; Younossi-Hartenstein et al., 1996). The optic lobes contain >800 NBs (Bertet et al., 2014; Li et al., 2013; Yasugi et al., 2008). These T1NB lineages generate most of the Drosophila central nervous system; thus, it is no surprise that these NB lineages have been the most studied. T2NBs have been recently discovered and offer an exciting division pattern that closely resembles some primate cortical neural stem cell lineages (reviewed in El-Danaf et al., 2023; Holguera and Desplan, 2018) (discussed more in Chapter V). There are 8 T2NB lineages localized to each central brain lobe; each having a unique spatial identity with 6 lineages that are dorsal medial and 2 lineages dorsal lateral (Bello et al., 2008; Boone and Doe, 2008; Bowman et al., 2008) (Fig 1.3, left). Each T2NB produces a unique cell lineage with morphologically distinct progeny cells that populate the adult brain (Andrade et al., 2019; Pereanu and 19 Hartenstein, 2006; Riebli et al., 2013; Yang et al., 2013). Moreover, each progenitor cell type within the Type 2 lineages have been identified to express unique markers (Fig 1.3, right; Table 1.1). Thus, the Type 2 lineage has proven to be an excellent model for precise lineage manipulations for understanding the development of neural stem cells and neurons (Bayraktar and Doe, 2013; Dillon et al., 2024; Hamid et al., 2024; Li et al., 2016; Li et al., 2017b; Munroe et al., 2022; Rives-Quinto et al., 2020; San-Juán and Baonza, 2011; Sullivan et al., 2019; Syed et al., 2017; Zhu et al., 2011; Zhu et al., 2012). Type 2 lineages will be discussed in length below and are the focus in Chapters III and IV. Fig. 1.3 Spatial positioning of Type 2 neuroblast and cell lineage markers. The eight Type 2 neuroblast lineages are bilaterally symmetrical with stereotyped spatial positions in the brain lobes (left). Type 2 neuroblasts generate identifiable lineage clusters of cell types (right). See Table 1.1 for references. A, Anterior; P, Posterior; M, Medial; L, Lateral; D, Dorsal; V, Ventral; DM, Dorsal-medial; DL; Dorsal-lateral; T2NB, Type 2 Neuroblast; INP, Intermediate Neural Progenitor; GMC, Ganglion Mother Cell. PntP1, Pointed; Dpn, Deadpan; Tll, Tailless; Ase, Asense; Ham, Hamlet; Erm, Earmuff; Tap, Target of poxn; Dap, Dacapo; Mir, Miranda; Elav, Rmbryonic lethal abnormal vision; Repo; Reverse polarity; Brp, Bruchpilot; Fne, Found in neurons; Ncad, Neural cadherin. 20 Table 1.1 Markers distinguishing the Type 2 neuroblast lineage progenitor cell types. Cell type Marker References Type 2 neuroblast (T2NB) Deadpan (Dpn) + (Bello et al., 2008; Bier et al., 1992; Boone and Doe, 2008; Bowman et al., 2008; Rives-Quinto et al., 2020; Zhu et al., 2011) Pointed (Pnt) + Tailless (Tll) + Asense (Ase) - Intermediate neural progenitor (INP) Deadpan (Dpn) + (Bello et al., 2008; Boone and Doe, 2008; Bowman et al., 2008; Li et al., 2016; Rives-Quinto et al., 2020; Zhu et al., 2011) Asense (Ase) + Hamlet (Ham) + Earmuff (Erm) + Ganglion mother cell (GMC) Target of Poxn (Tap) + (Ikeshima-Kataoka et al., 1997; Lane et al., 1996; Michki et al., 2021) Dacapo (Dap) + Deadpan (Dpn) - Miranda (Mir) - Glia Reverse polarity (Repo) + (Campbell et al., 1994; Xiong et al., 1994) Neuron Embyonic lethal abnormal vision (Elav) + (Robinow and White, 1991; Samson and Chalvet, 2003; Wagh et al., 2006; Young and Armstrong, 2010) Bruchpilot (Brp) + Found in neurons (Fne) + Neural cadherin (Ncad) + The discovery of temporal transcription factors In addition to the spatial factors determining the unique NB identities, temporal patterning within the lineages have been extensively studied as a mechanism for generating neuronal diversity. Embryonic NBs were first observed to sequentially express a series of transcription factors that were hypothesized to regulate temporal fate decisions (Brody and Odenwald, 2000; Kambadur et al., 1998). Isshiki et al. first discovered the canonical embryonic temporal transcription factor (TTF) cascade where narrow windows of transiently expressed factors, TTFs, in the NBs are required and sufficient to specify individual neuron identities (Isshiki et al., 2001) (Fig 1.4). Subsequent work has shown this TTF cascade to be a widely used mechanism across most embryonic T1NB lineages, persists in larval NBs, and is cross-regulating (Almeida and Bray, 2005; Baumgardt et al., 2014; Benito-Sipos et al., 2011; Cenci and Gould, 2005; Cleary and Doe, 2006; Grosskortenhaus, 2006; Grosskortenhaus et al., 2005; Meng et al., 2019; Meng et al., 2020; Moris-Sanz et al., 2014; Novotny et al., 2002; Pearson and Doe, 2003; Seroka and Doe, 2019; Tran and Doe, 2008) (Fig 1.4). 21 Fig. 1.4 The canonical temporal transcription factor cascade of embryonic Type 1 neuroblasts in the ventral nerve cord. Most embryonic T1NBs of the ventral nerve cord will transiently express the series of Hb, Kr, Pdm, Cas, and Grh. Variations of this temporal cascade is seen across some NB lineages (reviewed in: Doe, 2017; Pearson and Doe, 2004; Pollington et al., 2023). Hemilineages are formed during the GMC division when one daughter cell receives a Non cue and the other cell a Noff signal. Dashed lines indicate cross- regulatory relationship between the TTFs. Hb, Hunchback; Kr, Krüppel; Pdm, POU Domain protein; Cas, Castor; Grh, Grainyhead; NB, Neuroblast; GMC, Ganglion Mother Cell; N, Notch; TTF, Temporal Transcription Factor. The most extensively studied T1NB lineage for understanding TTFs has been NB 7-1 with the earliest TTF Hb studied the most within this lineage (Grosskortenhaus et al., 2005; Hirono et al., 2017; Isshiki et al., 2001; Kanai et al., 2005; Kohwi et al., 2011; Meng et al., 2019; Meng et al., 2020; Pearson and Doe, 2003; Seroka and Doe, 2019; Seroka et al., 2020; Seroka et al., 2022) (reviewed in Doe, 2017; Pollington et al., 2023). Hb was reported to be required and sufficient for specifying the first two neuron cell fates born from NB 7-1, motor neurons U1 and U2 (Isshiki et al., 2001). Recent studies have demonstrated that Hb is required for proper U motor neuron dendritic morphology and targeting to body wall muscles (Meng et al., 2019; Meng et al., 2020; Seroka and Doe, 2019). Interestingly, after more than 20 years since its discovery as a TTF, a mechanism for Hb in specifying the U1 and U2 motor neurons has not been identified (i.e., U1/U2 specific identity genes activated by Hb). Previous work has shown that Hb binds differentially across NB lineages 5-6 and 7-4 due to a difference in lineage-specific chromatin accessibility imparted by spatial factors (Sen et al., 2019). These studies suggest that determining the DNA-binding targets of TTFs across NB lineages will provide insight into the mechanism for specifying diverse cell fates. It remains an open question how any TTF acts within any NB lineage to specify unique identities. 22 Larval INPs, derived from T2NBs, display a similar temporal patterning progression as embryonic T1NBs. INPs express a series of cross-regulatory TTFs as INPs age (Bayraktar and Doe, 2013; Tang et al., 2022) (Fig 1.5). Most INP TTFs have been validated for being required to specify neuronal fates based on molecular markers and adult neuron morphology (Bayraktar and Doe, 2013; Sullivan et al., 2019). Neurons derived from INPs express markers tied to their INP temporal window. Young INP derived neurons generally express Dichaete, Runt, or Bsh (Bayraktar and Doe, 2013; Sullivan et al., 2019) (Fig 1.5). Old INP derived glia express Repo and the neurons Toy (Bayraktar and Doe, 2013) (Fig 1.5). It remains unknown if some of these markers are maintained from the larval to adult stages. Interestingly, the glia (Repo+) are restricted to an early T2NB window and Bsh+ neurons to a late T2NB window (Bayraktar and Doe, 2013). These data suggest that the temporal patterning in INPs can act in combination with temporal patterning of T2NBs to generate significant cell type diversity (discussed below). It remains unknown if Type 2 GMCs produce Non and Noff hemilineages, as seen in the embryonic T1NB lineages. If hemilineages exist in the Type 2 lineage, this would provide a fourth mechanism (1 - Spatial, 2 – T2NB temporal, 3 - INP temporal, 4 - hemilineage) for generating the neuronal diversity seen in the adult brain. Fig. 1.5 The temporal transcription factor cascade of larval Type 2 intermediate neural progenitors. Larval INPs express a series of TTFs as they age from young (recently derived from the T2NB) INPs that produce D+, Runt+, and Bsh+ progeny to old (final divisions for the INP) INPs that produce Repo+ and Toy+ progeny (Bayraktar and Doe, 2013; Sullivan et al., 2019; Tang et al., 2022). Dashed lines indicate cross-regulatory relationship between the TTFs. D, Dichaete; Hbn, Homeobrain; Grh, Grainyhead; Ey, Eyeless; Scro, Scarecrow; INP, Intermediate Neural Progenitor; GMC, Ganglion Mother Cell; N, Notch; TTF, Temporal Transcription Factor. 23 Temporal progression in the larval neuroblasts Most late-stage embryonic T1NBs and T2NBs will enter quiescence, cell cycle arrest when the NB does not divide, and reactivate ~12-24h after larval hatching (Munroe et al., 2022; Prokop and Technau, 1991; Truman and Bate, 1988) (Fig 1.6). The reactivation of larval NBs requires insulin signaling with quiescent NBs shown to be primed to respond to Insulin-like peptides to reactive proliferation (Chell and Brand, 2010; Dillon et al., 2022; Otsuki and Brand, 2018; Sousa-Nunes et al., 2011) (Fig 1.6; see Chapter II). Additionally, glia secrete the signaling molecules Ana and Trol to inhibit NB proliferation and promote NB proliferation, respectively, to temporally control neurogenesis in the early larvae (Datta, 1995; Datta, 1999; Ebens et al., 1993) (Fig 1.6). Quiescent NBs have been shown to repress Artichoke (Atk) and reactivate proliferation based on a dorsal-ventral positioning due to a heterogeneity in arrested cell stated of G0 or G2 (Otsuki and Brand, 2018; Otsuki and Brand, 2019). Additionally, Hh has been suggested to play a role in regulating quiescence downstream of the embryonic TTF cascade (Chai et al., 2013). Recent work has shown that the RNA-binding protein IGF-II mRNA-binding protein (Imp) is required for T2NBs to exit from quiescence (Munroe et al., 2022). It remains to be seen if this cue is also required for other NB lineages. Ongoing work aims to understand the mechanisms controlling the reactivation of NBs in the larval stages with the transcription factor Foxo suspected to be an essential regulator (see Dr. Sarah Siegrist’s lab: https://siegristlab.org/research/; personal communication). See Chapter V for further discussion. Fig. 1.6 Neuroblast progression of neurogenesis across fly development. Embryonic NBs proliferate until late stages when most lineages enter quiescence. Larval NBs express Trbls during quiescence with Ana secreted by glia to inhibit proliferation (Datta, 1995; Datta, 1999; Ebens et al., 1993). NBs express InR and are reactivated by signaling molecules Trol and Ilps (Chell and Brand, 2010; Datta, 1995; Datta, 1999; Dillon et al., 2022; Sousa-Nunes et al., 2011). Green indicates proliferating NB, magenta a quiescent NB, and grey a 24 decommissioning NB. Dashed lines indicate regulatory inputs. NB, Neuroblast; Trbl, Tribbles; Ana, Anachronism; InR, Insulin Receptor; Trol, Terribly reduce optic lobes; Ilps, Insulin like peptides. Larval NBs, excluding the optic lobes, have been less studied than embryonic NBs with an open question remaining about how temporal patterning occurs during larval stages. Mushroom body T1NBs have been shown to express opposing temporal gradients of the RNA-binding proteins Imp and Syncrip that are required for proper specification of neuronal fates (Liu et al., 2015). Imp is expressed at high levels in early larval NBs and decreases in expression level as Syncrip increases in expression level during later stages (Liu et al., 2015). The temporal gradient of Imp and Syncrip has been seen in the T2NB lineages (Ren et al., 2017; Syed et al., 2017) (Fig 1.7). This suggests that larval NB lineages may use the broad protein gradients as a mechanism for temporal patterning instead of the narrow windows of TTF expression. This is supported by several studies showing that Imp and Syncrip gradients in the T2NBs and INPs are required for specifying some neuronal fates (Hamid et al., 2024; Munroe and Doe, 2023; Ren et al., 2017; Syed et al., 2017). Alternatively, candidate TTFs have been identified as temporally expressed within T2NBs and may be required to specify neuronal identities, similar to the embryonic T1NB lineages (Bayraktar and Doe, 2013; Ren et al., 2017; Syed et al., 2017). Recent unpublished studies indicate the these candidate TTFs are indeed necessary for specifying neuronal identities (Wani et al., 2023 preprint) (Dillon and Doe, 2024, preprint; see Chapter IV). Regardless of the mechanism, the temporal progression in T2NBs is required for specifying birth order-dependent neuronal identities (Dillon et al., 2024). These data suggest that both protein gradients and TTFs regulate temporal patterning in the larval T2NBs. 25 Fig. 1.7 Temporal patterning of the Type 2 neuroblast in larval stages. T2NBs express opposing gradients of the RNA-binding proteins Imp and Syncrip (Ren et al., 2017; Syed et al., 2017). Seven-up is known to be required for switching the temporal progression from early-to-late (Dillon et al., 2024; Ren et al., 2017; Syed et al., 2017). Candidate TTFs are hypothesized to specify neuronal fates (Bayraktar and Doe, 2013; Dillon et al., 2024; Ren et al., 2017; Syed et al., 2017). Green indicates a relationship to an early temporal fates and magenta a relationship to a late temporal fates (Bayraktar and Doe, 2013; Dillon et al., 2024; Hamid et al., 2024; Ren et al., 2017; Syed et al., 2017) (Dillon and Doe, 2024, preprint). White indicates an unvalidated relationship. T2NB, Type 2 Neuroblast; Imp, IGF-II mRNA- binding protein; EcR-B1, Ecdysone Receptor isoform 1B; E93, Ecdysone induced protein 93; TTF, Temporal Transcription Factor. The mechanism for temporal progression in proliferating larval NBs remains unknown. Recent work has shown that the early factor Imp is required for reactivation of proliferation (Munroe et al., 2022). Additionally, the Imp and Syncrip gradients are cross-regulatory for the early-to-late progression (Liu et al., 2015; Ren et al., 2017). The switching factor Seven-up has been identified as being required for regulating this transition and the T2NB production switch from early-to-late identity neurons (Dillon et al., 2024; Ren et al., 2017; Syed et al., 2017) (Fig 1.7). This role for Seven-up as a switching has also been seen in the embryonic and larval T1NBs (Benito-Sipos et al., 2011; Kanai et al., 2005; Kohwi et al., 2011; Maurange et al., 2008; Mettler et al., 2006). The ecdysone receptor isoform B1 (EcR-B1) is also required for initiating the early- to-late switch and acts downstream of Seven-up in T2NBs (Syed et al., 2017). These data suggest that both intrinsic (Imp, Syncrip, and Seven-up) and extrinsic (EcR-B1 and ecdysone signaling) are required for temporal progression in proliferating T2NBs. It remains unknown what other factors are involved. See Chapter V for further discussion. 26 Larval NBs go through decommissioning, a process where the NB will stop proliferating to either differentiate into a postmitotic cell fate or undergo cell death (Fig 1.6). Most larval NBs decommission in the late larval or early pupal stages (Homem et al., 2014; Ito and Hotta, 1992; Maurange et al., 2008; Narbonne-Reveau et al., 2016; Siegrist et al., 2010; Yang et al., 2017). This process in the T1NBs is initiated by the Imp-to-Syncrip transition with atypically high Imp expression sustaining mushroom body NBs to continue proliferating longer than other T1NB lineages (Yang et al., 2017). Thus, it is no surprise that Seven-up is required for T1NB and T2NB lineages to initiate the timely onset of decommissioning as this switching factor acts upstream of the Imp-to-Syncrip transition (Dillon et al., 2024; Maurange et al., 2008; Narbonne-Reveau et al., 2016). Interestingly, the expression of Seven-up is early in the larval T2NBs at ~24h yet decommissioning occurs in the early pupal stages, days after Seven-up is no longer expressed (Bayraktar and Doe, 2013; Dillon et al., 2024; Homem et al., 2014; Ren et al., 2017; Syed et al., 2017). It remains unknown how Seven-up acts to initiate temporal progression for the onset of decommissioning in T2NB. See Chapter V for further discussion. 27 Establishing complexity within the Drosophila central brain As discussed in the previous section, neurogenesis in the developing Drosophila central nervous system produces diverse neuronal cell types. The integration of spatial patterning of distinct NB lineages and the subsequent temporal patterning provide a robust framework for understanding neurogenesis that has been extensively reviewed (reviewed in Doe, 2017; El-Danaf et al., 2023; Pearson and Doe, 2004; Pollington et al., 2023; Rossi et al., 2021; Skeath and Thor, 2003). The result of these 5 days of neurogenesis leads to the adult fruit fly that contains the complex neural circuitry to allow context-dependent behavior. This section will focus on the adult central brain with brief mentions of some circuits. The cell type diversity and function of the larval nervous system will not be discussed here. The diversity and function of the Central Complex An extensively studied region of the adult Drosophila brain is the Central Complex (CX). The CX is comprised of four neuropils, regions of high synaptic connectivity, known as the Protocerebral Bridge (PB), Fan-shaped Body (FB), Ellipsoid Body (EB), and Noduli (NO) (Hanesch et al., 1989) (Fig 1.8, left). The main two classes of neurons in the CX are the tangential neurons that are transversally oriented and the columnar neurons that are longitudinally oriented (Hanesch et al., 1989). These broad neuron classes have distinct functions in the CX and arise from distinct NB lineages. Fig. 1.8 The adult Central Complex neuropils and neurons. The CX is comprised of four main neuropils: PB, FB, EB, and NO. Other neuropils to note are the GA and BU (left). Most 28 studied neurons of the CX include the tangential Ring neurons and the columnar neurons (right). CX, Central Complex; PB, Protocerebral Bridge; FB, Fan-shaped Body; EB, Ellipsoid Body; NO, Noduli; GA, Gall; BU, Bulb. Figure diagrams inspired and adapted from: Wolff and Rubin, 2018; Seeling and Jayaraman 2013. Ring neurons have been the most studied of the tangential neurons. These neurons bridge the anterior optic tubercle, the dominant visual input region, to columnar neurons to form the anterior visual pathway circuit (Omoto et al., 2017; Seelig and Jayaraman, 2013). Importantly, Ring neurons provide spatially organized visual information to the CX circuitry (Omoto et al., 2017; Timaeus et al., 2020). These Ring neurons are characterized by their unique morphology in targeting the Bulb (BU), their concentric rings within the EB, and laterally located cell bodies (Renn et al., 1999; Young and Armstrong, 2010) (Fig 1.8, right). The Ring neurons are derived from two T1NB lineages with other tangential neurons derived from both T1NB and T2NB lineages (Bridi et al., 2019; Larsen et al., 2009; Omoto et al., 2017; Wong et al., 2013; Yang et al., 2013). One lineage tracing study has demonstrated that Ring neuron subtypes are born in a birth order-dependent manner with unique molecular markers across embryonic, larval, and pupal stages (Bridi et al., 2019). It will be important work investigating the mechanisms behind the specification of tangential neurons. Interest should be placed in the Ring neurons as they provide a system of two T1NB lineages that generate a diverse class of neurons across all of Drosophila neurogenesis. The majority of the CX is generated from T2NBs starting from late-stage embryo and into the pupal stages (Riebli et al., 2013; Walsh and Doe, 2017). The four T2NB lineages DM1-4 (Fig 1.3, left) generate all the adult columnar neurons (Andrade et al., 2019; Ito and Hotta, 1992; Pereanu and Hartenstein, 2006; Riebli et al., 2013; Yang et al., 2013) These columnar neurons are characterized by their apical cell body location with the majority innervating the PB, a subset of other CX neuropils, and occasionally neuropils outside of the CX (reviewed in Pfeiffer and Homberg, 2014) (Fig 1.8, right). Lineage tracing studies have shown that columnar neuron subtypes are born in a birth order-dependent manner (Andrade et al., 2019; Riebli et al., 2013; Walsh and Doe, 2017). Thus, columnar neurons provide an excellent model for understanding temporal patterning in the T2NB that give rise to this unique class of neurons. 29 Due to the diversity in CX neuron morphology, these neurons are named after the neuropils they innervate for a dendrite-axon naming convention (Wolff and Rubin, 2018; Wolff et al., 2015). For example, E-PG neurons are named for their dendrite targeting to the EB and axonal targeting to the PB and GA. Similarly, P-EN neurons are named for their dendritic targeting of the PB and axonal targeting to the EB and NO. Connectomes of the CX show hundreds of morphologically distinct neuron subtypes with complex synaptic connectivity (Franconville et al., 2018; Hulse et al., 2021; Scheffer et al., 2020; Zheng et al., 2018). Studies in other insect specicies has shown conserved and evolutionarily divergent neuroarchitecture with the Drosophila CX (reviewed in Pfeiffer and Homberg, 2014). These studies show that the CX is primed as a model for understanding not only developmental biology but also probe questions about evolution and systems neuroscience. Research on the circuitry of the CX has focused on the integration of visual sensory input and the locomotor output with some recent work aiming to understand the CX’s role in sleep (reviewed in: Fisher, 2022; Helfrich-Förster, 2018; Strauss, 2002; Turner-Evans and Jayaraman, 2016). The most studied role of the CX has been in navigational behavior. The EB can be considered the fly’s compass as the directional heading of the adult fly is encoded in the EB and can be maintained even in the absence of a visual stimulus (Green et al., 2017; Green et al., 2019; Seelig and Jayaraman, 2013; Turner-Evans et al., 2017; Turner-Evans et al., 2020). The two columnar identities of E-PG and P-EN neurons are integrated to maintain and continuously update directional heading from sensory cues (Green et al., 2017; Green et al., 2019; Turner-Evans et al., 2017; Turner-Evans et al., 2020). Surprisingly, the P-EN neurons were found to contain two subtypes, P-ENa and P-ENb neurons, based on behavioral differences (Green et al., 2017). Subsequent EM reconstructions have identified morphological differences in synaptic connectivity initially missed by light microscopy (Scheffer et al., 2020; Turner-Evans et al., 2020; Wolff et al., 2015). The visual inputs for the directional heading circuit is encoded by the Ring neurons (discussed above), and other similar neuron classes not discussed here, connecting the visual system to the CX via the EB (Omoto et al., 2017; Seelig and Jayaraman, 2013; Timaeus et al., 2020). It remains an open question how these CX circuits are initially formed and if there is an underlining developmental mechanism that connects these neuron subtypes. For example: Are the CX neurons that are wired together born in the same temporal windows and are 30 their identities specified by shared temporal factors? See Chapters III and IV for studies on the development of E-PG and P-EN neurons and Chapter V for further discussion. Tools for accessing the Central Complex While connectomes of the CX provide unparallel access to the morphology and connectivity of neurons, they fail to provide data on the genetic programs underlining the brain’s development. Recent single-cell/ single-nuclei RNA sequencing (scRNAseq) datasets of the adult Drosophila brain demonstrate that neurons are equally diverse in their transcriptomes as they are in morphology (Abruzzi et al., 2017; Croset et al., 2018; Davie et al., 2018; Epiney et al., 2023, preprint; Janssens et al., 2021; Li et al., 2017a). The Doe lab (https://www.doelab.org/) has ongoing work with a comprehensive dataset on adult neurons and glia derived from the T2NB lineages (Epiney et al., 2023, preprint). This work has identified unique molecular markers that distinguish CX neuron identities and provide support that combinatorial expression patterns define diverse neuronal cell types (Epiney et al., 2023, preprint). Importantly, these data sets provide access to the transcriptomes of individual neuronal subtypes. These data support a model similar to that found in C. elegans where combinations of homeodomain transcription factors delineate all neuronal cell types (Reilly et al., 2020). It remains to be seen if this is consistent across NB lineages in Drosophila much less vertebrate nervous systems, both of which contain significantly more neuronal diversity. These large transcriptomic datasets are valuable tools for understanding neuron diversity and building testable hypotheses prior to running experiments. One of the greatest genetic tools in Drosophila neurobiology has been the development and wide use of the UAS/Gal4 and LexA/LexAop systems (Brand and Perrimon, 1993; Fischer et al., 1988; Lai and Lee, 2006; Szüts and Bienz, 2000). These tools allow selective expression of genetic constructs in cell-specific patterns to provide spatial and temporal control, a geneticist’s dream. Combined with the recent advancements in CRISPR/Cas9 tools to use in flies, tissue- specific genetic knockouts offer a tool to study null mutations with precision of the affected cell types (Ewen-Campen et al., 2017; Port and Bullock, 2016; Port et al., 2020). Most relevant for the CX has been the extensive Gal4 and LexA lines that label hundreds of CX neuron types (Jenett et al., 2012; Wolff and Rubin, 2018; Wolff et al., 2015). These genetic stocks have allowed for the precise labeling of CX neuron molecular identity (i.e., pattern of Gal4 and LexA 31 expression) and labeling of neuron morphology. In addition to these genetic markers, several studies have identified unique transcription factors for CX cell types that can be identified through immunohistochemistry (Bayraktar and Doe, 2013; Dillon et al., 2024; Epiney et al., 2023; Sullivan et al., 2019) (Dillon and Doe, 2024, preprint). The tools described approve have been used extensively throughout the following work to investigate the development of the CX. 32 Bridge This chapter covered the development of the Drosophila central nervous system starting at the delamination of the NB from embryonic neuroectoderm. I describe how NBs are spatially patterned to produce unique NBs identities and how each NB identity generates unique cell lineages. I focused on how temporal patterning by TTFs are required for generating neuronal diversity in embryonic T1NB lineages. It remains an open question whether temporal protein gradients or TTFs act as temporal patterning mechanisms in the larval T1NB and T2NB lineages. Furthermore, it is unknown how the larval NB lineages temporally progress in exiting from quiescence to proliferating and eventually decommissioning. I shifted focus to discuss how the adult CX is comprised of diverse cell types that are apart of important behavioral neural circuits. I finished the chapter by discussing the available tools used to study the CX. In the following chapters, I present my previously published and unpublished work investigating the development of the Drosophila central nervous system. Chapter II will cover my 2022 publication that investigated a large scRNAseq dataset across multiple stages of early larval development. Chapter III will cover my 2024 publication that shows Seven-up is required in T2NB lineages for important temporal progression of a) switching production from early-to-late neuron identities and b) initiating T2NB decommissioning. Chapter IV covers my recent work, under review at Development, on how Castor acts as a TTF in T2NBs to specify early born CX identities. The last chapter, Chapter V, will discuss future directions from my work and others. Additionally, I will cover the similarities and differences between neurogenesis in the fly and mammalian brains. 33 References Abruzzi, K. C., Zadina, A., Luo, W., Wiyanto, E., Rahman, R., Guo, F., Shafer, O. and Rosbash, M. (2017). RNA-seq analysis of Drosophila clock and non-clock neurons reveals neuron-specific cycling and novel candidate neuropeptides. PLOS Genet. 13, e1006613. Almeida, M. S. and Bray, S. J. (2005). Regulation of post-embryonic neuroblasts by Drosophila Grainyhead. Mech. Dev. 122, 1282–1293. Andrade, I. V., Riebli, N., Nguyen, B.-C. M., Omoto, J. J., Cardona, A. and Hartenstein, V. (2019). Developmentally Arrested Precursors of Pontine Neurons Establish an Embryonic Blueprint of the Drosophila Central Complex. Curr. Biol. 29, 412-425.e3. Baumgardt, M., Karlsson, D., Salmani, B. Y., Bivik, C., MacDonald, R. B., Gunnar, E. and Thor, S. (2014). Global Programmed Switch in Neural Daughter Cell Proliferation Mode Triggered by a Temporal Gene Cascade. Dev. Cell 30, 192–208. Bayraktar, O. A. and Doe, C. Q. (2013). Combinatorial temporal patterning in progenitors expands neural diversity. Nature 498, 449–455. Bello, B. C., Izergina, N., Caussinus, E. and Reichert, H. (2008). Amplification of neural stem cell proliferation by intermediate progenitor cells in Drosophila brain development. Neural Develop. 3, 5. Benito-Sipos, J., Ulvklo, C., Gabilondo, H., Baumgardt, M., Angel, A., Torroja, L. and Thor, S. (2011). Seven up acts as a temporal factor during two different stages of neuroblast 5-6 development. Development 138, 5311–5320. Bertet, C., Li, X., Erclik, T., Cavey, M., Wells, B. and Desplan, C. (2014). Temporal patterning of neuroblasts controls Notch-mediated cell survival through regulation of Hid or Reaper. Cell 158, 1173–1186. Bhat, K. M. (1996). The patched signaling pathway mediates repression of gooseberry allowing neuroblast specification by wingless during Drosophila neurogenesis. Dev. Camb. Engl. 122, 2921–2932. Bier, E., Vaessin, H., Younger-Shepherd, S., Jan, L. Y. and Jan, Y. N. (1992). deadpan, an essential pan-neural gene in Drosophila, encodes a helix-loop-helix protein similar to the hairy gene product. Genes Dev. 6, 2137–2151. Boone, J. Q. and Doe, C. Q. (2008). Identification of Drosophila type II neuroblast lineages containing transit amplifying ganglion mother cells. Dev. Neurobiol. 68, 1185–1195. Bossing, T., Udolph, G., Doe, C. Q. and Technau, G. M. (1996). The Embryonic Central Nervous System Lineages ofDrosophila melanogaster. Dev. Biol. 179, 41–64. Bowman, S. K., Rolland, V., Betschinger, J., Kinsey, K. A., Emery, G. and Knoblich, J. A. (2008). The Tumor Suppressors Brat and Numb Regulate Transit-Amplifying Neuroblast Lineages in Drosophila. Dev. Cell 14, 535–546. 34 Brand, A. H. and Perrimon, N. (1993). Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Dev. Camb. Engl. 118, 401–415. Bridi, J. C., Ludlow, Z. N. and Hirth, F. (2019). Lineage-specific determination of ring neuron circuitry in the central complex of Drosophila. Biol. Open 8, bio045062. Broadus, J., Skeath, J. B., Spana, E. P., Bossing, T., Technau, G. and Doe, C. Q. (1995). New neuroblast markers and the origin of the aCC/pCC neurons in the Drosophila central nervous system. Mech. Dev. 53, 393–402. Brody, T. and Odenwald, W. F. (2000). Programmed transformations in neuroblast gene expression during Drosophila CNS lineage development. Dev. Biol. 226, 34–44. Campbell, G., Göring, H., Lin, T., Spana, E., Andersson, S., Doe, C. Q. and Tomlinson, A. (1994). RK2, a glial-specific homeodomain protein required for embryonic nerve cord condensation and viability in Drosophila. Dev. Camb. Engl. 120, 2957–2966. Cenci, C. and Gould, A. P. (2005). Drosophila Grainyhead specifies late programmes of neural proliferation by regulating the mitotic activity and Hox-dependent apoptosis of neuroblasts. Dev. Camb. Engl. 132, 3835–3845. Chai, P. C., Liu, Z., Chia, W. and Cai, Y. (2013). Hedgehog Signaling Acts with the Temporal Cascade to Promote Neuroblast Cell Cycle Exit. PLoS Biol. 11, e1001494. Chell, J. M. and Brand, A. H. (2010). Nutrition-Responsive Glia Control Exit of Neural Stem Cells from Quiescence. Cell 143, 1161–1173. Chu-LaGraff, Q. and Doe, C. Q. (1993). Neuroblast specification and formation regulated by wingless in the Drosophila CNS. Science 261, 1594–1597. Cleary, M. D. and Doe, C. Q. (2006). Regulation of neuroblast competence: multiple temporal identity factors specify distinct neuronal fates within a single early competence window. Genes Dev. 20, 429–434. Croset, V., Treiber, C. D. and Waddell, S. (2018). Cellular diversity in the Drosophila midbrain revealed by single-cell transcriptomics. eLife 7, e34550. Datta, S. (1995). Control of proliferation activation in quiescent neuroblasts of the Drosophila central nervous system. Development 121, 1173–1182. Datta, S. (1999). Activation of neuroblast proliferation in explant culture of the Drosophila larval CNS. Brain Res. 818, 77–83. Davie, K., Janssens, J., Koldere, D., De Waegeneer, M., Pech, U., Kreft, Ł., Aibar, S., Makhzami, S., Christiaens, V., Bravo González-Blas, C., et al. (2018). A Single-Cell Transcriptome Atlas of the Aging Drosophila Brain. Cell 174, 982-998.e20. 35 Deshpande, N., Dittrich, R., Technau, G. M. and Urban, J. (2001). Successive specification of Drosophila neuroblasts NB 6-4 and NB 7-3 depends on interaction of the segment polarity genes wingless, gooseberry and naked cuticle. Dev. Camb. Engl. 128, 3253–3261. Dillon, N. R. and Doe, C. Q. (2024). Castor is a temporal transcription factor that specifies early born central complex neuron identity. 2024.08.22.609207. Dillon, N., Cocanougher, B., Sood, C., Yuan, X., Kohn, A. B., Moroz, L. L., Siegrist, S. E., Zlatic, M. and Doe, C. Q. (2022). Single cell RNA-seq analysis reveals temporally-regulated and quiescence-regulated gene expression in Drosophila larval neuroblasts. Neural Develop. 17, 7. Dillon, N. R., Manning, L., Hirono, K. and Doe, C. Q. (2024). Seven-up acts in neuroblasts to specify adult central complex neuron identity and initiate neuroblast decommissioning. Development 151, dev202504. Doe, C. Q. (1992). Molecular markers for identified neuroblasts and ganglion mother cells in the Drosophila central nervous system. Development 116, 855–863. Doe, C. Q. (2017). Temporal Patterning in the Drosophila CNS. Annu. Rev. Cell Dev. Biol. 33, 219–240. Ebens, A. J., Garren, H., Cheyette, B. N. R. and Zipursky, S. L. (1993). The Drosophila anachronism locus: A glycoprotein secreted by glia inhibits neuroblast proliferation. Cell 74, 15– 27. El-Danaf, R. N., Rajesh, R. and Desplan, C. (2023). Temporal regulation of neural diversity in Drosophila and vertebrates. Semin. Cell Dev. Biol. 142, 13–22. Epiney, D., Chaya, G. M., Dillon, N., Lai, S.-L. and Doe, C. (2023). Transcriptional complexity in the insect central complex: single nuclei RNA sequencing of adult brain neurons derived from type 2 neuroblasts. 2023.12.10.571022. Ewen-Campen, B., Yang-Zhou, D., Fernandes, V. R., González, D. P., Liu, L.-P., Tao, R., Ren, X., Sun, J., Hu, Y., Zirin, J., et al. (2017). Optimized strategy for in vivo Cas9-activation in Drosophila. Proc. Natl. Acad. Sci. 114, 9409–9414. Fischer, J. A., Giniger, E., Maniatis, T. and Ptashne, M. (1988). GAL4 activates transcription in Drosophila. Nature 332, 853–856. Fisher, Y. E. (2022). Flexible navigational computations in the Drosophila central complex. Curr. Opin. Neurobiol. 73, 102514. Franconville, R., Beron, C. and Jayaraman, V. (2018). Building a functional connectome of the Drosophila central complex. eLife 7, e37017. Green, J., Adachi, A., Shah, K. K., Hirokawa, J. D., Magani, P. S. and Maimon, G. (2017). A neural circuit architecture for angular integration in Drosophila. Nature 546, 101–106. 36 Green, J., Vijayan, V., Mussells Pires, P., Adachi, A. and Maimon, G. (2019). A neural heading estimate is compared with an internal goal to guide oriented navigation. Nat. Neurosci. 22, 1460– 1468. Grosskortenhaus, R. (2006). Pdm and Castor specify late-born motor neuron identity in the NB7- 1 lineage. Genes Dev. 20, 2618–2627. Grosskortenhaus, R., Pearson, B. J., Marusich, A. and Doe, C. Q. (2005). Regulation of Temporal Identity Transitions in Drosophila Neuroblasts. Dev. Cell 8, 193–202. Hamid, A., Gattuso, H., Caglar, A. N., Pillai, M., Steele, T., Gonzalez, A., Nagel, K. and Syed, M. H. (2024). The conserved RNA-binding protein Imp is required for the specification and function of olfactory navigation circuitry in Drosophila. Curr. Biol. 34, 473-488.e6. Hanesch, U., Fischbach, K.-F. and Heisenberg, M. (1989). Neuronal architecture of the central complex in Drosophila melanogaster. Cell Tissue Res. 257, 343–366. Hartenstein, V. and Campos-Ortega, J. A. (1984). Early neurogenesis in wild-type Drosophila melanogaster. Wilhelm Rouxs Arch. Dev. Biol. 193, 308–325. Hartenstein, V. and Wodarz, A. (2013). Initial neurogenesis in Drosophila. Wiley Interdiscip. Rev. Dev. Biol. 2, 701–721. Hartenstein, V., Younossi-Hartenstein, A. and Lekven, A. (1994). Delamination and Division in the Drosophila Neurectoderm: Spatiotemporal Pattern, Cytoskeletal Dynamics, and Common Control by Neurogenic and Segment Polarity Genes. Dev. Biol. 165, 480–499. Helfrich-Förster, C. (2018). Sleep in Insects. Annu. Rev. Entomol. 63, 69–86. Hirono, K., Kohwi, M., Clark, M. Q., Heckscher, E. S. and Doe, C. Q. (2017). The Hunchback temporal transcription factor establishes, but is not required to maintain, early-born neuronal identity. Neural Develop. 12, 1. Holguera, I. and Desplan, C. (2018). Neuronal specification in space and time. Science. Homem, C. C. F., Steinmann, V., Burkard, T. R., Jais, A., Esterbauer, H. and Knoblich, J. A. (2014). Ecdysone and Mediator Change Energy Metabolism to Terminate Proliferation in Drosophila Neural Stem Cells. Cell 158, 874–888. Hulse, B. K., Haberkern, H., Franconville, R., Turner-Evans, D. B., Takemura, S., Wolff, T., Noorman, M., Dreher, M., Dan, C., Parekh, R., et al. (2021). A connectome of the Drosophila central complex reveals network motifs suitable for flexible navigation and context-dependent action selection. eLife 10, e66039. Ikeshima-Kataoka, H., Skeath, J. B., Nabeshima, Y., Doe, C. Q. and Matsuzaki, F. (1997). Miranda directs Prospero to a daughter cell during Drosophila asymmetric divisions. Nature 390, 625–629. 37 Isshiki, T., Takeichi, M. and Nose, A. (1997). The role of the msh homeobox gene during Drosophila neurogenesis: implication for the dorsoventral specification of the neuroectoderm. Development 124, 3099–3109. Isshiki, T., Pearson, B., Holbrook, S. and Doe, C. Q. (2001). Drosophila Neuroblasts Sequentially Express Transcription Factors which Specify the Temporal Identity of Their Neuronal Progeny. Cell 106, 511–521. Ito, K. and Hotta, Y. (1992). Proliferation pattern of postembryonic neuroblasts in the brain of Drosophila melanogaster. Dev. Biol. 149, 134–148. Janssens, J., Aibar, S., Taskiran, I. I., Ismail, J. N., Spanier, K. I., González-Blas, C. B., Quan, X. J., Papasokrati, D., Hulselmans, G., Makhzami, S., et al. (2021). Decoding gene regulation in the fly brain. Jenett, A., Rubin, G. M., Ngo, T.-T. B., Shepherd, D., Murphy, C., Dionne, H., Pfeiffer, B. D., Cavallaro, A., Hall, D., Jeter, J., et al. (2012). A GAL4-Driver Line Resource for Drosophila Neurobiology. Cell Rep. 2, 991–1001. Kambadur, R., Koizumi, K., Stivers, C., Nagle, J., Poole, S. J. and Odenwald, W. F. (1998). Regulation of POU genes by castor and hunchback establishes layered compartments in the Drosophila CNS. Genes Dev. 12, 246. Kanai, M. I., Okabe, M. and Hiromi, Y. (2005). seven-up Controls Switching of Transcription Factors that Specify Temporal Identities of Drosophila Neuroblasts. Dev. Cell 8, 203–213. Kohwi, M., Hiebert, L. S. and Doe, C. Q. (2011). The pipsqueak-domain proteins Distal antenna and Distal antenna-related restrict Hunchback neuroblast expression and early-born neuronal identity. Development 138, 1727–1735. Lai, S.-L. and Lee, T. (2006). Genetic mosaic with dual binary transcriptional systems in Drosophila. Nat. Neurosci. 9, 703–709. Lane, M. E., Sauer, K., Wallace, K., Jan, Y. N., Lehner, C. F. and Vaessin, H. (1996). Dacapo, a cyclin-dependent kinase inhibitor, stops cell proliferation during Drosophila development. Cell 87, 1225–1235. Larsen, C., Shy, D., Spindler, S. R., Fung, S., Pereanu, W., Younossi-Hartenstein, A. and Hartenstein, V. (2009). Patterns of growth, axonal extension and axonal arborization of neuronal lineages in the developing Drosophila brain. Dev. Biol. 335, 289–304. Li, X., Erclik, T., Bertet, C., Chen, Z., Voutev, R., Venkatesh, S., Morante, J., Celik, A. and Desplan, C. (2013). Temporal patterning of Drosophila medulla neuroblasts controls neural fates. Nature 498, 456–462. Li, X., Xie, Y. and Zhu, S. (2016). Notch maintains Drosophila type II neuroblasts by suppressing expression of the Fez transcription factor Earmuff. Development 143, 2511–2521. 38 Li, H., Horns, F., Wu, B., Xie, Q., Li, J., Li, T., Luginbuhl, D. J., Quake, S. R. and Luo, L. (2017a). Classifying Drosophila Olfactory Projection Neuron Subtypes by Single-Cell RNA Sequencing. Cell 171, 1206-1220.e22. Li, X., Chen, R. and Zhu, S. (2017b). bHLH-O proteins balance the self-renewal and differentiation of Drosophila neural stem cells by regulating Earmuff expression. Dev. Biol. 431, 239–251. Liu, Z., Yang, C.-P., Sugino, K., Fu, C.-C., Liu, L.-Y., Yao, X., Lee, L. P. and Lee, T. (2015). Opposing intrinsic temporal gradients guide neural stem cell production of varied neuronal fates. Science 350, 317–320. Matsuzaki, M. and Saigo, K. (1996). hedgehog signaling independent of engrailed and wingless required for post-S1 neuroblast formation in Drosophila CNS. Dev. Camb. Engl. 122, 3567– 3575. Maurange, C., Cheng, L. and Gould, A. P. (2008). Temporal Transcription Factors and Their Targets Schedule the End of Neural Proliferation in Drosophila. Cell 133, 891–902. McDonald, J. A. and Doe, C. Q. (1997). Establishing neuroblast-specific gene expression in the Drosophila CNS: huckebein is activated by Wingless and Hedgehog and repressed by Engrailed and Gooseberry. Development 124, 1079–1087. McDonald, J. A., Holbrook, S., Isshiki, T., Weiss, J., Doe, C. Q. and Mellerick, D. M. (1998). Dorsoventral patterning in the Drosophila central nervous system: the vnd homeobox gene specifies ventral column identity. Genes Dev. 12, 3603–3612. Meng, J. L., Marshall, Z. D., Lobb-Rabe, M. and Heckscher, E. S. (2019). How prolonged expression of Hunchback, a temporal transcription factor, re-wires locomotor circuits. eLife 8, e46089. Meng, J. L., Wang, Y., Carrillo, R. A. and Heckscher, E. S. (2020). Temporal transcription factors determine circuit membership by permanently altering motor neuron-to-muscle synaptic partnerships. eLife 9, e56898. Mettler, U., Vogler, G. and Urban, J. (2006). Timing of identity: spatiotemporal regulation of hunchback in neuroblast lineages of Drosophila by Seven-up and Prospero. Development 133, 429–437. Michki, N. S., Li, Y., Sanjasaz, K., Zhao, Y., Shen, F. Y., Walker, L. A., Cao, W., Lee, C.-Y. and Cai, D. (2021). The molecular landscape of neural differentiation in the developing Drosophila brain revealed by targeted scRNA-seq and multi-informatic analysis. Cell Rep. 35, 109039. Moris-Sanz, M., Estacio-Gómez, A., Alvarez-Rivero, J. and Díaz-Benjumea, F. J. (2014). Specification of neuronal subtypes by different levels of Hunchback. Dev. Camb. Engl. 141, 4366–4374. 39 Munroe, J. A. and Doe, C. Q. (2023). Imp is expressed in INPs and newborn neurons where it regulates neuropil targeting in the central complex. Neural Develop. 18, 9. Munroe, J. A., Syed, M. H. and Doe, C. Q. (2022). Imp is required for timely exit from quiescence in Drosophila type II neuroblasts. PLOS ONE 17, e0272177. Narbonne-Reveau, K., Lanet, E., Dillard, C., Foppolo, S., Chen, C.-H., Parrinello, H., Rialle, S., Sokol, N. S. and Maurange, C. (2016). Neural stem cell-encoded temporal patterning delineates an early window of malignant susceptibility in Drosophila. eLife 5, e13463. Novotny, T., Eiselt, R. and Urban, J. (2002). Hunchback is required for the specification of the early sublineage of neuroblast 7-3 in the Drosophila central nervous system. Dev. Camb. Engl. 129, 1027–1036. Omoto, J. J., Keleş, M. F., Nguyen, B.-C. M., Bolanos, C., Lovick, J. K., Frye, M. A. and Hartenstein, V. (2017). Visual Input to the Drosophila Central Complex by Developmentally and Functionally Distinct Neuronal Populations. Curr. Biol. 27, 1098–1110. Otsuki, L. and Brand, A. H. (2018). Cell cycle heterogeneity directs the timing of neural stem cell activation from quiescence. Science 360, 99–102. Otsuki, L. and Brand, A. H. (2019). Dorsal-Ventral Differences in Neural Stem Cell Quiescence Are Induced by p57KIP2/Dacapo. Dev. Cell 49, 293-300.e3. Pearson, B. J. and Doe, C. Q. (2003). Regulation of neuroblast competence in Drosophila. Nature 425, 624–628. Pearson, B. J. and Doe, C. Q. (2004). SPECIFICATION OF TEMPORAL IDENTITY IN THE DEVELOPING NERVOUS SYSTEM. Annu. Rev. Cell Dev. Biol. 20, 619–647. Pereanu, W. and Hartenstein, V. (2006). Neural Lineages of the Drosophila Brain: A Three- Dimensional Digital Atlas of the Pattern of Lineage Location and Projection at the Late Larval Stage. J. Neurosci. 26, 5534–5553. Pfeiffer, K. and Homberg, U. (2014). Organization and Functional Roles of the Central Complex in the Insect Brain. Annu. Rev. Entomol. 59, 165–184. Pollington, H. Q., Seroka, A. Q. and Doe, C. Q. (2023). From temporal patterning to neuronal connectivity in Drosophila type I neuroblast lineages. Semin. Cell Dev. Biol. 142, 4–12. Port, F. and Bullock, S. L. (2016). Augmenting CRISPR applications in Drosophila with tRNA- flanked sgRNAs. Nat. Methods 13, 852–854. Port, F., Strein, C., Stricker, M., Rauscher, B., Heigwer, F., Zhou, J., Beyersdörffer, C., Frei, J., Hess, A., Kern, K., et al. (2020). A large-scale resource for tissue-specific CRISPR mutagenesis in Drosophila. eLife 9, e53865. 40 Prokop, A. and Technau, G. M. (1991). The origin of postembryonic neuroblasts in the ventral nerve cord of Drosophila melanogaster. Development 111, 79–88. Reilly, M. B., Cros, C., Varol, E., Yemini, E. and Hobert, O. (2020). Unique homeobox codes delineate all the neuron classes of C. elegans. Nature 584, 595–601. Ren, Q., Yang, C.-P., Liu, Z., Sugino, K., Mok, K., He, Y., Ito, M., Nern, A., Otsuna, H. and Lee, T. (2017). Stem Cell-Intrinsic, Seven-up-Triggered Temporal Factor Gradients Diversify Intermediate Neural Progenitors. Curr. Biol. 27, 1303–1313. Renn, S. C., Armstrong, J. D., Yang, M., Wang, Z., An, X., Kaiser, K. and Taghert, P. H. (1999). Genetic analysis of the Drosophila ellipsoid body neuropil: organization and development of the central complex. J. Neurobiol. 41, 189–207. Riebli, N., Viktorin, G. and Reichert, H. (2013). Early-born neurons in type II neuroblast lineages establish a larval primordium and integrate into adult circuitry during central complex development in Drosophila. Neural Develop. 8, 6. Rives-Quinto, N., Komori, H., Ostgaard, C. M., Janssens, D. H., Kondo, S., Dai, Q., Moore, A. W. and Lee, C.-Y. (2020). Sequential activation of transcriptional repressors promotes progenitor commitment by silencing stem cell identity genes. eLife 9, e56187. Robinow, S. and White, K. (1991). Characterization and spatial distribution of the ELAV protein during Drosophila melanogaster development. J. Neurobiol. 22, 443–461. Rossi, A. M., Jafari, S. and Desplan, C. (2021). Integrated Patterning Programs During Drosophila Development Generate the Diversity of Neurons and Control Their Mature Properties. Annu. Rev. Neurosci. 44, annurev-neuro-102120-014813. Samson, M.-L. and Chalvet, F. (2003). found in neurons, a third member of the Drosophila elav gene family, encodes a neuronal protein and interacts with elav. Mech. Dev. 120, 373–383. San-Juán, B. P. and Baonza, A. (2011). The bHLH factor deadpan is a direct target of Notch signaling and regulates neuroblast self-renewal in Drosophila. Dev. Biol. 352, 70–82. Scheffer, L. K., Xu, C. S., Januszewski, M., Lu, Z., Takemura, S., Hayworth, K. J., Huang, G. B., Shinomiya, K., Maitlin-Shepard, J., Berg, S., et al. (2020). A connectome and analysis of the adult Drosophila central brain. eLife 9, e57443. Schmid, A., Chiba, A. and Doe, C. Q. (1999). Clonal analysis of Drosophila embryonic neuroblasts: neural cell types, axon projections and muscle targets. Development 126, 4653– 4689. Schmidt, H., Rickert, C., Bossing, T., Vef, O., Urban, J. and Technau, G. M. (1997). The Embryonic Central Nervous System Lineages ofDrosophila melanogaster. Dev. Biol. 189, 186– 204. 41 Seelig, J. D. and Jayaraman, V. (2013). Feature detection and orientation tuning in the Drosophila central complex. Nature 503, 262–266. Sen, S. Q., Chanchani, S., Southall, T. D. and Doe, C. Q. (2019). Neuroblast-specific open chromatin allows the temporal transcription factor, Hunchback, to bind neuroblast-specific loci. eLife 8, e44036. Seroka, A. Q. and Doe, C. Q. (2019). The Hunchback temporal transcription factor determines motor neuron axon and dendrite targeting in Drosophila. Development 146, dev175570. Seroka, A., Yazejian, R. M., Lai, S.-L. and Doe, C. Q. (2020). A novel temporal identity window generates alternating Eve+/Nkx6+ motor neuron subtypes in a single progenitor lineage. Neural Develop. 15, 9. Seroka, A., Lai, S.-L. and Doe, C. Q. (2022). Transcriptional profiling from whole embryos to single neuroblast lineages in Drosophila. Dev. Biol. 489, 21–33. Siegrist, S. E., Haque, N. S., Chen, C.-H., Hay, B. A. and Hariharan, I. K. (2010). Inactivation of Both foxo and reaper Promotes Long-Term Adult Neurogenesis in Drosophila. Curr. Biol. 20, 643–648. Skeath, J. B. and Thor, S. (2003). Genetic control of Drosophila nerve cord development. Curr. Opin. Neurobiol. 13, 8–15. Skeath, J. B., Zhang, Y., Holmgren, R., Carroll, S. B. and Doe, C. Q. (1995). Specification of neuroblast identity in the Drosophila embryonic central nervous system by gooseberry-distal. Nature 376, 427–430. Sousa-Nunes, R., Yee, L. L. and Gould, A. P. (2011). Fat cells reactivate quiescent neuroblasts via TOR and glial insulin relays in Drosophila. Nature 471, 508–512. Strauss, R. (2002). The central complex and the genetic dissection of locomotor behaviour. Curr. Opin. Neurobiol. 12, 633–638. Sullivan, L. F., Warren, T. L. and Doe, C. Q. (2019). Temporal identity establishes columnar neuron morphology, connectivity, and function in a Drosophila navigation circuit. eLife 8, e43482. Syed, M. H., Mark, B. and Doe, C. Q. (2017). Steroid hormone induction of temporal gene expression in Drosophila brain neuroblasts generates neuronal and glial diversity. eLife 6, e26287. Szüts, D. and Bienz, M. (2000). LexA chimeras reveal the function of Drosophila Fos as a context-dependent transcriptional activator. Proc. Natl. Acad. Sci. 97, 5351–5356. Tang, J. L. Y., Hakes, A. E., Krautz, R., Suzuki, T., Contreras, E. G., Fox, P. M. and Brand, A. H. (2022). NanoDam identifies Homeobrain (ARX) and Scarecrow (NKX2.1) as conserved temporal factors in the Drosophila central brain and visual system. Dev. Cell 0,. 42 Timaeus, L., Geid, L., Sancer, G., Wernet, M. F. and Hummel, T. (2020). Parallel Visual Pathways with Topographic versus Nontopographic Organization Connect the Drosophila Eyes to the Central Brain. iScience 23, 101590. Tran, K. D. and Doe, C. Q. (2008). Pdm and Castor close successive temporal identity windows in the NB3-1 lineage. Dev. Camb. Engl. 135, 3491–3499. Truman, J. W. and Bate, M. (1988). Spatial and temporal patterns of neurogenesis in the central nervous system of Drosophila melanogaster. Dev. Biol. 125, 145–157. Turner-Evans, D. B. and Jayaraman, V. (2016). The insect central complex. Curr. Biol. 26, R453– R457. Turner-Evans, D., Wegener, S., Rouault, H., Franconville, R., Wolff, T., Seelig, J. D., Druckmann, S. and Jayaraman, V. (2017). Angular velocity integration in a fly heading circuit. eLife. Turner-Evans, D. B., Jensen, K. T., Ali, S., Paterson, T., Sheridan, A., Ray, R. P., Wolff, T., Lauritzen, J. S., Rubin, G. M., Bock, D. D., et al. (2020). The Neuroanatomical Ultrastructure and Function of a Biological Ring Attractor. Neuron 108, 145-163.e10. von Ohlen, T. and Doe, C. Q. (2000). Convergence of dorsal, dpp, and egfr signaling pathways subdivides the drosophila neuroectoderm into three dorsal-ventral columns. Dev. Biol. 224, 362– 372. Wagh, D. A., Rasse, T. M., Asan, E., Hofbauer, A., Schwenkert, I., Dürrbeck, H., Buchner, S., Dabauvalle, M.-C., Schmidt, M., Qin, G., et al. (2006). Bruchpilot, a Protein with Homology to ELKS/CAST, Is Required for Structural Integrity and Function of Synaptic Active Zones in Drosophila. Neuron 49, 833–844. Walsh, K. T. and Doe, C. Q. (2017). Drosophila embryonic type II neuroblasts: origin, temporal patterning, and contribution to the adult central complex. Development 144, 4552–4562. Wani, A. R., Chowdhury, B., Luong, J., Chaya, G. M., Patel, K., Isaacman-Beck, J., Shafer, O., Kayser, M. S. and Syed, M. H. (2023). Stem cell-specific ecdysone signaling regulates the development and function of a Drosophila sleep homeostat. 2023.09.29.560022. Weiss, J. B., Von Ohlen, T., Mellerick, D. M., Dressler, G., Doe, C. Q. and Scott, M. P. (1998). Dorsoventral patterning in the Drosophila central nervous system: the intermediate neuroblasts defective homeobox gene specifies intermediate column identity. Genes Dev. 12, 3591–3602. Wheeler, W. M. (1891). Neuroblasts in the Arthropod Embryo. Wheeler, W. M. (1893). A contribution to insect embryology. J. Morphol. 8, 1–161. Wolff, T. and Rubin, G. M. (2018). Neuroarchitecture of the Drosophila central complex: A catalog of nodulus and asymmetrical body neurons and a revision of the protocerebral bridge catalog. J. Comp. Neurol. 526, 2585–2611. 43 Wolff, T., Iyer, N. A. and Rubin, G. M. (2015). Neuroarchitecture and neuroanatomy of the Drosophila central complex: A GAL4-based dissection of protocerebral bridge neurons and circuits. J. Comp. Neurol. 523, 997–1037. Wong, D. C., Lovick, J. K., Ngo, K. T., Borisuthirattana, W., Omoto, J. J. and Hartenstein, V. (2013). Postembryonic lineages of the Drosophila brain: II. Identification of lineage projection patterns based on MARCM clones. Dev. Biol. 384, 258–289. Xiong, W. C., Okano, H., Patel, N. H., Blendy, J. A. and Montell, C. (1994). repo encodes a glial- specific homeo domain protein required in the Drosophila nervous system. Genes Dev. 8, 981– 994. Yang, J. S., Awasaki, T., Yu, H.-H., He, Y., Ding, P., Kao, J.-C. and Lee, T. (2013). Diverse neuronal lineages make stereotyped contributions to the Drosophila locomotor control center, the central complex: Lineage analysis of central complex neurons. J. Comp. Neurol. 521, 2645– 2662. Yang, C.-P., Samuels, T. J., Huang, Y., Yang, L., Ish-Horowicz, D., Davis, I. and Lee, T. (2017). Imp and Syp RNA-binding proteins govern decommissioning of Drosophila neural stem cells. Development 144, 3454–3464. Yasugi, T., Umetsu, D., Murakami, S., Sato, M. and Tabata, T. (2008). Drosophila optic lobe neuroblasts triggered by a wave of proneural gene expression that is negatively regulated by JAK/STAT. Dev. Camb. Engl. 135, 1471–1480. Young, J. M. and Armstrong, J. D. (2010). Building the central complex in Drosophila: the generation and development of distinct neural subsets. J. Comp. Neurol. 518, 1525–1541. Younossi-Hartenstein, A., Nassif, C., Green, P. and Hartenstein, V. (1996). Early neurogenesis of the Drosophila brain. J. Comp. Neurol. 370, 313–329. Zheng, Z., Lauritzen, J. S., Perlman, E., Robinson, C. G., Nichols, M., Milkie, D., Torrens, O., Price, J., Fisher, C. B., Sharifi, N., et al. (2018). A Complete Electron Microscopy Volume of the Brain of Adult Drosophila melanogaster. Cell 174, 730-743.e22. Zhu, S., Barshow, S., Wildonger, J., Jan, L. Y. and Jan, Y.-N. (2011). Ets transcription factor Pointed promotes the generation of intermediate neural progenitors in Drosophila larval brains. Proc. Natl. Acad. Sci. 108, 20615–20620. Zhu, S., Wildonger, J., Barshow, S., Younger, S., Huang, Y. and Lee, T. (2012). The bHLH Repressor Deadpan Regulates the Self-renewal and Specification of Drosophila Larval Neural Stem Cells Independently of Notch. PLOS ONE 7, e46724. 44 Chapter II A single-cell RNA-seq analysis reveals temporally-regulated and quiescence-regulated gene expression in larval neuroblasts Dillon, N., Cocanougher, B., Sood, C., Yuan, X., Kohn, A. B., Moroz, L. L., Siegrist, S. E., Zlatic, M. and Doe, C. Q. (2022). Single cell RNA-seq analysis reveals temporally-regulated and quiescence-regulated gene expression in Drosophila larval neuroblasts. Neural Development 17, 7. Author contributions ND performed all scRNAseq analysis, generated all figures, and wrote the manuscript. BC, LLM, ABK, and MZ generated the scRNAseq data; CS, XY, and SES generated Fig. 2.2H and provided comments on the manuscript. CQD supervised the project and edited figures and text. The authors read and approved the final manuscript. Introduction A major question in neuroscience is how neural diversity is generated, which underlies complex neural circuits and behavioral output of the central nervous system (CNS). In the past, neuronal diversity was commonly defined by morphological features (axon/dendrite processes), biochemical features (neurotransmitter choice), and physiological features (distinct ion channels and membrane properties) [1]. In addition, “low throughput” assays for molecular differences among neurons, typically for transcription factor (TF) expression, have been crucial for finding insights into the generation of neural diversity for decades [2, 3]. Taken together, these approaches resulted in the definition various classes or subtypes of motor neurons, interneurons, sensory neurons and peptidergic neurons, but they are ill-suited to address the question of how many unique types of neurons exist within the CNS, and the subsequent question of how each cell type contributes to the function of the CNS. The advent of single cell RNA sequencing (scRNA-seq) allowed a more complete inventory of gene expression profiles within individual neurons, with the expression of “validated cell type 45 genes” used as a framework to identify transcriptionally related neurons [4–8]. Further analysis has revealed novel cell types based on common gene expression, but also that trajectories between cell types to be more gradual and less saltatory than previously appreciated, in part due to transcriptional priming [9–11]. In Drosophila, neuronal scRNA-seq has been done on adult brain [12–16], pupae [17–22], larvae [23–25], and blastoderm-stage embryos [26]. These experiments have provided valuable insight into the number of distinct neuronal types and identified gene candidates for regulating neural subtype function or connectivity. However, no studies to date have focused on identifying and characterizing the transcriptional diversity of neural progenitors, nor has any study mapped progenitor transcriptional profile at multiple larval stages. In this study, we identify multiple progenitor subtypes across several larval stages with differential gene expression to provide candidate genes as cell type specific markers and functional roles during development. Results Larval atlas shows distinct cell identities and differentiating neural progenitor axis To identify single cell gene expression profiles throughout larval development, we used scRNA- seq data collected by [27] from dissociated brain and ventral nerve cord (VNC) – together termed the CNS – from larvae at 1h, 24h and 48h (all times in hours after larval hatching; ALH). We used the 10X Genomics pipeline for scRNA-seq analysis and used Cell Ranger Aggregation to aggregate multiple samples from the same timepoint. We used the standard Seurat integration pipeline to filter out low quality cells and clustered 97,845 cells from all larval stages (see methods; Fig. 2.1a). Within our atlas we identified clusters enriched for cell types in the CNS: neural progenitors, immature and mature neurons, glia, trachea, hemocytes and insulin-producing cells (IPCs; Fig. 2.1a; Supp. Table 2. 1). Representative examples of a progenitor marker (Deadpan; dpn), a new-born neuron marker (Hey), a maturing neuron marker (nSyb), and a glia