1
|
Benton R, Mermet J, Jang A, Endo K, Cruchet S, Menuz K. An integrated anatomical, functional and evolutionary view of the Drosophila olfactory system. EMBO Rep 2025:10.1038/s44319-025-00476-8. [PMID: 40389758 DOI: 10.1038/s44319-025-00476-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/21/2025] Open
Abstract
The Drosophila melanogaster olfactory system is one of the most intensively studied parts of the nervous system in any animal. Composed of ~50 independent olfactory neuron classes, with several associated hygrosensory and thermosensory pathways, it has been subject to diverse types of experimental analyses. However, synthesizing the available information is limited by the incomplete data and inconsistent nomenclature found in the literature. In this work, we first "complete" the peripheral sensory map through the identification of a previously uncharacterized antennal sensory neuron population expressing Or46aB, and the definition of an exceptional "hybrid" olfactory neuron class comprising functional Or and Ir receptors. Second, we survey developmental, anatomical, connectomic, functional, and evolutionary studies to generate an integrated dataset and associated visualizations of these sensory neuron pathways, creating an unprecedented resource. Third, we illustrate the utility of the dataset to reveal relationships between different organizational properties of this sensory system, and the new questions these stimulate. Such examples emphasize the power of this resource to promote further understanding of the construction, function, and evolution of these neural circuits.
Collapse
Affiliation(s)
- Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015, Lausanne, Switzerland.
| | - Jérôme Mermet
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Andre Jang
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA
| | - Keita Endo
- RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Steeve Cruchet
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Karen Menuz
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA.
- Connecticut Institute for Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
2
|
Soffers JH, Beck E, Sytkowski DJ, Maughan ME, Devarakonda D, Zhu Y, Wilson B, David Chen YC, Erclik T, Truman JW, Skeath JB, Lacin H. A library of lineage-specific driver lines connects developing neuronal circuits to behavior in the Drosophila Ventral Nerve Cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.27.625713. [PMID: 39651218 PMCID: PMC11623677 DOI: 10.1101/2024.11.27.625713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Understanding developmental changes in neuronal lineages is crucial to elucidate how they assemble into functional neural networks. Studies investigating nervous system development in model systems have only focused on select regions of the central nervous system due to the limited availability of genetic drivers that target specific neuronal lineages throughout development and adult life. This has hindered our understanding of how distinct neuronal lineages interconnect to form neuronal circuits during development. Here, we present a split-GAL4 library composed of genetic driver lines, which we generated via editing the genomic locus of lineage-specific transcription factors and demonstrate that we can use this library to specifically target most individual neuronal hemilineages in the Drosophila ventral nerve cord (VNC) throughout development and into adulthood. Using these genetic driver lines, we found striking morphological changes in neuronal processes within a lineage during metamorphosis. We also demonstrated how neurochemical features of neuronal classes can be quickly assessed. Lastly, we documented behaviors elicited in response to optogenetic activation of individual neuronal lineages and generated a comprehensive lineage-behavior map of the entire fly VNC. Looking forward, this lineage-specific split-GAL4 driver library will provide the genetic tools needed to address the questions emerging from the analysis of the recent VNC connectome and transcriptome datasets.
Collapse
|
3
|
Fuenzalida-Uribe N, Hidalgo S, Silva B, Gandhi S, Vo D, Zamani P, Holmes TC, Sayin S, Grunwald Kadow IC, Hadjieconomou D, O’Dowd DK, Campusano JM. The innexin 7 gap junction protein contributes to synchronized activity in the Drosophila antennal lobe and regulates olfactory function. Front Neural Circuits 2025; 19:1563401. [PMID: 40352759 PMCID: PMC12062127 DOI: 10.3389/fncir.2025.1563401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
In the mammalian olfactory bulb (OB), gap junctions coordinate synchronous activity among mitral and tufted cells to process olfactory information. In insects, gap junctions are also present in the antennal lobe (AL), a structure homologous to the mammalian OB. The invertebrate gap junction protein ShakB contributes to electrical synapses between AL projection neurons (PNs) in Drosophila. Other gap junction proteins, including innexin 7 (Inx7), are also expressed in the Drosophila AL, but little is known about their contribution to intercellular communication during olfactory information processing. In this study, we report spontaneous calcium transients in PNs grown in cell culture that are highly synchronous when these neurons are physically connected. RNAi-mediated knockdown of Inx7 in cultured PNs blocks calcium transient neuronal synchronization. In vivo, downregulation of Inx7 in the AL impairs both vinegar-induced electrophysiological calcium responses and behavioral responses to this appetitive stimulus. These results demonstrate that Inx7-encoded gap junctions functionally coordinate PN activity and modulate olfactory information processing in the adult Drosophila AL.
Collapse
Affiliation(s)
- Nicolás Fuenzalida-Uribe
- Laboratorio Neurogenética de la Conducta, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Institute of Neurobiology, University of Puerto Rico- Medical Sciences Campus, San Juan, Puerto Rico
| | - Sergio Hidalgo
- Laboratorio Neurogenética de la Conducta, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington, DC, United States
| | - Bryon Silva
- Laboratorio Neurogenética de la Conducta, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Institut du Cerveau-Paris Brain Institute (ICM), Paris, France
| | - Saurin Gandhi
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, United States
| | - David Vo
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, United States
| | - Parham Zamani
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, United States
| | - Todd C. Holmes
- Department of Physiology and Biophysics, and Center for Neural Circuit Mapping, University of California Irvine, Irvine, CA, United States
| | - Sercan Sayin
- Department of Biology, University of Konstanz, Konstanz, Germany
| | | | - Dafni Hadjieconomou
- Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Institut du Cerveau-Paris Brain Institute (ICM), Paris, France
| | - Diane K. O’Dowd
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, United States
| | - Jorge M. Campusano
- Laboratorio Neurogenética de la Conducta, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
4
|
Dürr BR, Bertolini E, Takagi S, Pascual J, Abuin L, Lucarelli G, Benton R, Auer TO. Olfactory projection neuron rewiring in the brain of an ecological specialist. Cell Rep 2025; 44:115615. [PMID: 40287940 DOI: 10.1016/j.celrep.2025.115615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/24/2024] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Animal behaviors can differ greatly between closely related species. These behavioral changes are frequently linked to sensory system modifications, but central brain cell-type alterations might also be involved. Here, we develop advanced genetic tools to compare homologous central neurons in Drosophila sechellia, an ecological specialist, with the generalist Drosophila melanogaster. Through systematic morphological analysis of olfactory projection neurons (PNs), we reveal that the global anatomy of these second-order neurons is conserved. However, high-resolution, quantitative comparisons identify a striking case of convergent rewiring of PNs in two olfactory pathways critical for D. sechellia's host location. Calcium imaging and labeling of pre-synaptic sites in these evolved D. sechellia PNs indicate that species-specific connections with third-order partners are formed. This work demonstrates that peripheral sensory evolution is accompanied by selective wiring changes in the central brain to facilitate ecological specialization and paves the way to compare other cell types throughout the nervous system.
Collapse
Affiliation(s)
- Benedikt R Dürr
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland; Centre for Organismal Studies, Heidelberg University, 69120 Heidelberg, Germany
| | - Enrico Bertolini
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Suguru Takagi
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Justine Pascual
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Liliane Abuin
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Giovanna Lucarelli
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Thomas O Auer
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland.
| |
Collapse
|
5
|
Lyu C, Li Z, Xu C, Kalai J, Luo L. Rewiring an olfactory circuit by altering the combinatorial code of cell-surface proteins. RESEARCH SQUARE 2025:rs.3.rs-6099298. [PMID: 40162206 PMCID: PMC11952648 DOI: 10.21203/rs.3.rs-6099298/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Proper brain function requires the precise assembly of neural circuits during development. Despite the identification of many cell-surface proteins (CSPs) that help guide axons to their targets1,2, it remains largely unknown how multiple CSPs work together to assemble a functional circuit. Here, we used synaptic partner matching in the Drosophila olfactory circuit3,4 to address this question. By systematically altering the combination of differentially expressed CSPs in a single olfactory receptor neuron (ORN) type, which senses a male pheromone that inhibits male-male courtship, we switched its connection from its endogenous postsynaptic projection neuron (PN) type nearly completely to a new PN type that promotes courtship. To achieve this switch, we deduced a combinatorial code including CSPs that mediate both attractive and repulsive interactions between synaptic partners5,6. The anatomical switch changed the odor response of the new PN partner and markedly increased male-male courtship. We generalized three manipulation strategies from this rewiring to successfully rewire a second ORN type to multiple distinct PN types. This work demonstrates that manipulating a small set of CSPs is sufficient to respecify synaptic connections, paving ways to explore how neural systems evolve through changes of circuit connectivity.
Collapse
Affiliation(s)
- Cheng Lyu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Zhuoran Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Jordan Kalai
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
6
|
Li Z, Lyu C, Xu C, Hu Y, Luginbuhl DJ, Lehovic AB, Priest JM, Özkan E, Luo L. Repulsive interactions instruct synaptic partner matching in an olfactory circuit. RESEARCH SQUARE 2025:rs.3.rs-6099208. [PMID: 40162214 PMCID: PMC11952656 DOI: 10.21203/rs.3.rs-6099208/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Neurons exhibit extraordinary precision in selecting synaptic partners. Whereas cell-surface proteins (CSPs) mediating attractive interactions between developing axons and dendrites have been shown to instruct synaptic partner matching1,2, it is less clear the degree to which repulsive interactions play a role. Here, using a genetic screen guided by single cell transcriptomes3,4, we identified three CSP pairs-Toll2-Ptp10D, Fili-Kek1, and Hbs/Sns-Kirre-in mediating repulsive interactions between non-partner olfactory receptor neuron (ORN) axons and projection neuron (PN) dendrites in the developing Drosophila olfactory circuit. Each CSP pair exhibits inverse expression patterns in the select PN-ORN partners. Loss of each CSP in ORNs led to similar synaptic partner matching deficits as the loss of its partner CSP in PNs, and mistargeting phenotypes caused by overexpressing one CSP could be suppressed by loss of its partner CSP. Each CSP pair is also differentially expressed in other brain regions. Together, our data reveal that multiple repulsive CSP pairs work together to ensure precise synaptic partner matching during development by preventing neurons from forming connections with non-cognate partners.
Collapse
Affiliation(s)
- Zhuoran Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Cheng Lyu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ying Hu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - David J. Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Asaf B. Lehovic
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Jessica M. Priest
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
7
|
Churgin MA, Lavrentovich DO, Smith MAY, Gao R, Boyden ES, de Bivort BL. A neural correlate of individual odor preference in Drosophila. eLife 2025; 12:RP90511. [PMID: 40067954 PMCID: PMC11896609 DOI: 10.7554/elife.90511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025] Open
Abstract
Behavior varies even among genetically identical animals raised in the same environment. However, little is known about the circuit or anatomical origins of this individuality. Here, we demonstrate a neural correlate of Drosophila odor preference behavior in the olfactory sensory periphery. Namely, idiosyncratic calcium responses in projection neuron (PN) dendrites and densities of the presynaptic protein Bruchpilot in olfactory receptor neuron (ORN) axon terminals correlate with individual preferences in a choice between two aversive odorants. The ORN-PN synapse appears to be a locus of individuality where microscale variation gives rise to idiosyncratic behavior. Simulating microscale stochasticity in ORN-PN synapses of a 3062 neuron model of the antennal lobe recapitulates patterns of variation in PN calcium responses matching experiments. Conversely, stochasticity in other compartments of this circuit does not recapitulate those patterns. Our results demonstrate how physiological and microscale structural circuit variations can give rise to individual behavior, even when genetics and environment are held constant.
Collapse
Affiliation(s)
- Matthew A Churgin
- Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Center for Brain Science, Harvard University, CambridgeCambridgeUnited States
| | - Danylo O Lavrentovich
- Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Center for Brain Science, Harvard University, CambridgeCambridgeUnited States
| | - Matthew A-Y Smith
- Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Center for Brain Science, Harvard University, CambridgeCambridgeUnited States
| | - Ruixuan Gao
- McGovern Institute, MITCambridgeUnited States
- MIT Media Lab, MITCambridgeUnited States
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Edward S Boyden
- McGovern Institute, MITCambridgeUnited States
- Department of Biological Engineering, MITCambridgeUnited States
- Koch Institute, Department of Biology, MITCambridgeUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Brain and Cognitive Sciences, MITCambridgeUnited States
| | - Benjamin L de Bivort
- Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
- Center for Brain Science, Harvard University, CambridgeCambridgeUnited States
| |
Collapse
|
8
|
Li Z, Lyu C, Xu C, Hu Y, Luginbuhl DJ, Caspi-Lebovic AB, Priest JM, Özkan E, Luo L. Repulsive interactions instruct synaptic partner matching in an olfactory circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.01.640985. [PMID: 40060423 PMCID: PMC11888401 DOI: 10.1101/2025.03.01.640985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Neurons exhibit extraordinary precision in selecting synaptic partners. Whereas cell-surface proteins (CSPs) mediating attractive interactions between developing axons and dendrites have been shown to instruct synaptic partner matching1,2, it is less clear the degree to which repulsive interactions play a role. Here, using a genetic screen guided by single cell transcriptomes3,4, we identified three CSP pairs-Toll2-Ptp10D, Fili-Kek1, and Hbs/Sns-Kirre-in mediating repulsive interactions between non-partner olfactory receptor neuron (ORN) axons and projection neuron (PN) dendrites in the developing Drosophila olfactory circuit. Each CSP pair exhibits inverse expression patterns in the select PN-ORN partners. Loss of each CSP in ORNs led to similar synaptic partner matching deficits as the loss of its partner CSP in PNs, and mistargeting phenotypes caused by overexpressing one CSP could be suppressed by loss of its partner CSP. Each CSP pair is also differentially expressed in other brain regions. Together, our data reveal that multiple repulsive CSP pairs work together to ensure precise synaptic partner matching during development by preventing neurons from forming connections with non-cognate partners.
Collapse
Affiliation(s)
- Zhuoran Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Cheng Lyu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- These authors contributed equally
| | - Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ying Hu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - David J. Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Asaf B. Caspi-Lebovic
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Jessica M. Priest
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, The Neuroscience Institute and Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
9
|
Shuai Y, Sammons M, Sterne GR, Hibbard KL, Yang H, Yang CP, Managan C, Siwanowicz I, Lee T, Rubin GM, Turner GC, Aso Y. Driver lines for studying associative learning in Drosophila. eLife 2025; 13:RP94168. [PMID: 39879130 PMCID: PMC11778931 DOI: 10.7554/elife.94168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025] Open
Abstract
The mushroom body (MB) is the center for associative learning in insects. In Drosophila, intersectional split-GAL4 drivers and electron microscopy (EM) connectomes have laid the foundation for precise interrogation of the MB neural circuits. However, investigation of many cell types upstream and downstream of the MB has been hindered due to lack of specific driver lines. Here we describe a new collection of over 800 split-GAL4 and split-LexA drivers that cover approximately 300 cell types, including sugar sensory neurons, putative nociceptive ascending neurons, olfactory and thermo-/hygro-sensory projection neurons, interneurons connected with the MB-extrinsic neurons, and various other cell types. We characterized activation phenotypes for a subset of these lines and identified a sugar sensory neuron line most suitable for reward substitution. Leveraging the thousands of confocal microscopy images associated with the collection, we analyzed neuronal morphological stereotypy and discovered that one set of mushroom body output neurons, MBON08/MBON09, exhibits striking individuality and asymmetry across animals. In conjunction with the EM connectome maps, the driver lines reported here offer a powerful resource for functional dissection of neural circuits for associative learning in adult Drosophila.
Collapse
Affiliation(s)
- Yichun Shuai
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Megan Sammons
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Gabriella R Sterne
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Karen L Hibbard
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - He Yang
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Ching-Po Yang
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Claire Managan
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Igor Siwanowicz
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Tzumin Lee
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Gerald M Rubin
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Glenn C Turner
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Yoshinori Aso
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| |
Collapse
|
10
|
Benton R, Mermet J, Jang A, Endo K, Cruchet S, Menuz K. An integrated anatomical, functional and evolutionary view of the Drosophila olfactory system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.632927. [PMID: 39868125 PMCID: PMC11760703 DOI: 10.1101/2025.01.16.632927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The Drosophila melanogaster olfactory system is one of the most intensively studied parts of the nervous system in any animal. Composed of ~60 independent olfactory neuron classes, with several associated hygrosensory and thermosensory pathways, it has been subject to diverse types of experimental analyses. However, synthesizing the available data is limited by the incompleteness and inconsistent nomenclature found in the literature. In this work, we first "complete" the peripheral sensory map through the identification of a previously uncharacterized antennal sensory neuron population expressing Or46aB, and the definition of an exceptional "hybrid" olfactory neuron class comprising functional Or and Ir receptors. Second, we survey developmental, anatomical, connectomic, functional and evolutionary studies to generate an integrated dataset of these sensory neuron pathways - and associated visualizations - creating an unprecedented comprehensive resource. Third, we illustrate the utility of the dataset to reveal relationships between different organizational properties of this sensory system, and the new questions these stimulate. These examples emphasize the power of this resource to promote further understanding of the construction, function and evolution of these neural circuits.
Collapse
Affiliation(s)
- Richard Benton
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne CH-1015 Lausanne Switzerland
| | - Jérôme Mermet
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne CH-1015 Lausanne Switzerland
| | - Andre Jang
- Department of Physiology and Neurobiology University of Connecticut Storrs Connecticut 06269 United States
| | - Keita Endo
- RIKEN Center for Brain Science Wako Saitama 351-0198 Japan
| | - Steeve Cruchet
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne CH-1015 Lausanne Switzerland
| | - Karen Menuz
- Department of Physiology and Neurobiology University of Connecticut Storrs Connecticut 06269 United States
- Connecticut Institute for Brain and Cognitive Sciences University of Connecticut Storrs Connecticut 06269 United States
| |
Collapse
|
11
|
Lyu C, Li Z, Xu C, Wong KKL, Luginbuhl DJ, McLaughlin CN, Xie Q, Li T, Li H, Luo L. Dimensionality reduction simplifies synaptic partner matching in an olfactory circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609939. [PMID: 39253519 PMCID: PMC11383009 DOI: 10.1101/2024.08.27.609939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
The distribution of postsynaptic partners in three-dimensional (3D) space presents complex choices for a navigating axon. Here, we discovered a dimensionality reduction principle in establishing the 3D glomerular map in the fly antennal lobe. Olfactory receptor neuron (ORN) axons first contact partner projection neuron (PN) dendrites at the 2D spherical surface of the antennal lobe during development, regardless of whether the adult glomeruli are at the surface or interior of the antennal lobe. Along the antennal lobe surface, axons of each ORN type take a specific 1D arc-shaped trajectory that precisely intersects with their partner PN dendrites. Altering axon trajectories compromises synaptic partner matching. Thus, a 3D search problem is reduced to 1D, which simplifies synaptic partner matching and may generalize to the wiring process of more complex brains.
Collapse
Affiliation(s)
- Cheng Lyu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Zhuoran Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Kenneth Kin Lam Wong
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David J. Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Colleen N. McLaughlin
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Qijing Xie
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Tongchao Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Present address: Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
| | - Hongjie Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Present address: Huffington Center on Aging, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
12
|
Thornton-Kolbe EM, Ahmed M, Gordon FR, Sieriebriennikov B, Williams DL, Kurmangaliyev YZ, Clowney EJ. Spatial constraints and cell surface molecule depletion structure a randomly connected learning circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603956. [PMID: 39071296 PMCID: PMC11275898 DOI: 10.1101/2024.07.17.603956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The brain can represent almost limitless objects to "categorize an unlabeled world" (Edelman, 1989). This feat is supported by expansion layer circuit architectures, in which neurons carrying information about discrete sensory channels make combinatorial connections onto much larger postsynaptic populations. Combinatorial connections in expansion layers are modeled as randomized sets. The extent to which randomized wiring exists in vivo is debated, and how combinatorial connectivity patterns are generated during development is not understood. Non-deterministic wiring algorithms could program such connectivity using minimal genomic information. Here, we investigate anatomic and transcriptional patterns and perturb partner availability to ask how Kenyon cells, the expansion layer neurons of the insect mushroom body, obtain combinatorial input from olfactory projection neurons. Olfactory projection neurons form their presynaptic outputs in an orderly, predictable, and biased fashion. We find that Kenyon cells accept spatially co-located but molecularly heterogeneous inputs from this orderly map, and ask how Kenyon cell surface molecule expression impacts partner choice. Cell surface immunoglobulins are broadly depleted in Kenyon cells, and we propose that this allows them to form connections with molecularly heterogeneous partners. This model can explain how developmentally identical neurons acquire diverse wiring identities.
Collapse
Affiliation(s)
- Emma M. Thornton-Kolbe
- Neurosciences Graduate Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maria Ahmed
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Finley R. Gordon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | | | - Donnell L. Williams
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | | | - E. Josephine Clowney
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Liu W, Li Q. Single-cell transcriptomics dissecting the development and evolution of nervous system in insects. CURRENT OPINION IN INSECT SCIENCE 2024; 63:101201. [PMID: 38608931 DOI: 10.1016/j.cois.2024.101201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
Insects can display a vast repertoire of complex and adaptive behaviors crucial for survival and reproduction. Yet, how the neural circuits underlying insect behaviors are assembled throughout development and remodeled during evolution remains largely obscure. The advent of single-cell transcriptomics has opened new paths to illuminate these historically intractable questions. Insect behavior is governed by its brain, whose functional complexity is realized through operations across multiple levels, from the molecular and cellular to the circuit and organ. Single-cell transcriptomics enables dissecting brain functions across all these levels and allows tracking regulatory dynamics throughout development and under perturbation. In this review, we mainly focus on the achievements of single-cell transcriptomics in dissecting the molecular and cellular architectures of nervous systems in representative insects, then discuss its applications in tracking the developmental trajectory and functional evolution of insect brains.
Collapse
Affiliation(s)
- Weiwei Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China; Yunnan Key Laboratory of Biodiversity Information, Kunming, China.
| | - Qiye Li
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
14
|
Sun C, Shao Y, Iqbal J. Insect Insights at the Single-Cell Level: Technologies and Applications. Cells 2023; 13:91. [PMID: 38201295 PMCID: PMC10777908 DOI: 10.3390/cells13010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Single-cell techniques are a promising way to unravel the complexity and heterogeneity of transcripts at the cellular level and to reveal the composition of different cell types and functions in a tissue or organ. In recent years, advances in single-cell RNA sequencing (scRNA-seq) have further changed our view of biological systems. The application of scRNA-seq in insects enables the comprehensive characterization of both common and rare cell types and cell states, the discovery of new cell types, and revealing how cell types relate to each other. The recent application of scRNA-seq techniques to insect tissues has led to a number of exciting discoveries. Here we provide an overview of scRNA-seq and its application in insect research, focusing on biological applications, current challenges, and future opportunities to make new discoveries with scRNA-seq in insects.
Collapse
Affiliation(s)
- Chao Sun
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, Hangzhou 310058, China;
| | - Yongqi Shao
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Junaid Iqbal
- Institute of Sericulture and Apiculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
15
|
Ahmed OM, Crocker A, Murthy M. Transcriptional profiling of Drosophila male-specific P1 (pC1) neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566045. [PMID: 37986870 PMCID: PMC10659367 DOI: 10.1101/2023.11.07.566045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
In Drosophila melanogaster, the P1 (pC1) cluster of male-specific neurons both integrates sensory cues and drives or modulates behavioral programs such as courtship, in addition to contributing to a social arousal state. The behavioral function of these neurons is linked to the genes they express, which underpin their capacity for synaptic signaling, neuromodulation, and physiology. Yet, P1 (pC1) neurons have not been fully characterized at the transcriptome level. Moreover, it is unknown how the molecular landscape of P1 (pC1) neurons acutely changes after flies engage in social behaviors, where baseline P1 (pC1) neural activity is expected to increase. To address these two gaps, we use single cell-type RNA sequencing to profile and compare the transcriptomes of P1 (pC1) neurons harvested from socially paired versus solitary male flies. Compared to control transcriptome datasets, we find that P1 (pC1) neurons are enriched in 2,665 genes, including those encoding receptors, neuropeptides, and cell-adhesion molecules (dprs/DIPs). Furthermore, courtship is characterized by changes in ~300 genes, including those previously implicated in regulating behavior (e.g. DopEcR, Octβ3R, Fife, kairos, rad). Finally, we identify a suite of genes that link conspecific courtship with the innate immune system. Together, these data serve as a molecular map for future studies of an important set of higher-order and sexually-dimorphic neurons.
Collapse
Affiliation(s)
- Osama M Ahmed
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540, USA
- Department of Psychology, University of Washington, Seattle, WA 98105, USA
| | - Amanda Crocker
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540, USA
- Program in Neuroscience, Middlebury College, Middlebury, VT 05753, USA
| | - Mala Murthy
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540, USA
| |
Collapse
|
16
|
Lyu C, Li Z, Luo L. Toward building a library of cell type-specific drivers across developmental stages. Proc Natl Acad Sci U S A 2023; 120:e2312196120. [PMID: 37590431 PMCID: PMC10466085 DOI: 10.1073/pnas.2312196120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023] Open
Affiliation(s)
- Cheng Lyu
- HHMI, Stanford University, Stanford, CA94305
- Department of Biology, Stanford University, Stanford, CA94305
| | - Zhuoran Li
- HHMI, Stanford University, Stanford, CA94305
- Department of Biology, Stanford University, Stanford, CA94305
| | - Liqun Luo
- HHMI, Stanford University, Stanford, CA94305
- Department of Biology, Stanford University, Stanford, CA94305
| |
Collapse
|
17
|
Temporal control of neuronal wiring. Semin Cell Dev Biol 2023; 142:81-90. [PMID: 35644877 DOI: 10.1016/j.semcdb.2022.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/22/2022]
Abstract
Wiring an animal brain is a complex process involving a staggering number of cell-types born at different times and locations in the developing brain. Incorporation of these cells into precise circuits with high fidelity is critical for animal survival and behavior. Assembly of neuronal circuits is heavily dependent upon proper timing of wiring programs, requiring neurons to express specific sets of genes (sometimes transiently) at the right time in development. While cell-type specificity of genetic programs regulating wiring has been studied in detail, mechanisms regulating proper timing and coordination of these programs across cell-types are only just beginning to emerge. In this review, we discuss some temporal regulators of wiring programs and how their activity is controlled over time and space. A common feature emerges from these temporal regulators - they are induced by cell-extrinsic cues and control transcription factors capable of regulating a highly cell-type specific set of target genes. Target specificity in these contexts comes from cell-type specific transcription factors. We propose that the spatiotemporal specificity of wiring programs is controlled by the combinatorial activity of temporal programs and cell-type specific transcription factors. Going forward, a better understanding of temporal regulators will be key to understanding the mechanisms underlying brain wiring, and will be critical for the development of in vitro models like brain organoids.
Collapse
|
18
|
Ma D, Herndon N, Le JQ, Abruzzi KC, Zinn K, Rosbash M. Neural connectivity molecules best identify the heterogeneous clock and dopaminergic cell types in the Drosophila adult brain. SCIENCE ADVANCES 2023; 9:eade8500. [PMID: 36812309 PMCID: PMC9946362 DOI: 10.1126/sciadv.ade8500] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/26/2023] [Indexed: 05/25/2023]
Abstract
Our recent single-cell sequencing of most adult Drosophila circadian neurons indicated notable and unexpected heterogeneity. To address whether other populations are similar, we sequenced a large subset of adult brain dopaminergic neurons. Their gene expression heterogeneity is similar to that of clock neurons, i.e., both populations have two to three cells per neuron group. There was also unexpected cell-specific expression of neuron communication molecule messenger RNAs: G protein-coupled receptor or cell surface molecule (CSM) transcripts alone can define adult brain dopaminergic and circadian neuron cell type. Moreover, the adult expression of the CSM DIP-beta in a small group of clock neurons is important for sleep. We suggest that the common features of circadian and dopaminergic neurons are general, essential for neuronal identity and connectivity of the adult brain, and that these features underlie the complex behavioral repertoire of Drosophila.
Collapse
Affiliation(s)
- Dingbang Ma
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Nicholas Herndon
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Jasmine Quynh Le
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Katharine C. Abruzzi
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| | - Kai Zinn
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael Rosbash
- Howard Hughes Medical Institute and Department of Biology, Brandeis University, Waltham, MA 02454, USA
| |
Collapse
|
19
|
Palmateer CM, Artikis C, Brovero SG, Friedman B, Gresham A, Arbeitman MN. Single-cell transcriptome profiles of Drosophila fruitless-expressing neurons from both sexes. eLife 2023; 12:e78511. [PMID: 36724009 PMCID: PMC9891730 DOI: 10.7554/elife.78511] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 01/08/2023] [Indexed: 02/02/2023] Open
Abstract
Drosophila melanogaster reproductive behaviors are orchestrated by fruitless neurons. We performed single-cell RNA-sequencing on pupal neurons that produce sex-specifically spliced fru transcripts, the fru P1-expressing neurons. Uniform Manifold Approximation and Projection (UMAP) with clustering generates an atlas containing 113 clusters. While the male and female neurons overlap in UMAP space, more than half the clusters have sex differences in neuron number, and nearly all clusters display sex-differential expression. Based on an examination of enriched marker genes, we annotate clusters as circadian clock neurons, mushroom body Kenyon cell neurons, neurotransmitter- and/or neuropeptide-producing, and those that express doublesex. Marker gene analyses also show that genes that encode members of the immunoglobulin superfamily of cell adhesion molecules, transcription factors, neuropeptides, neuropeptide receptors, and Wnts have unique patterns of enriched expression across the clusters. In vivo spatial gene expression links to the clusters are examined. A functional analysis of fru P1 circadian neurons shows they have dimorphic roles in activity and period length. Given that most clusters are comprised of male and female neurons indicates that the sexes have fru P1 neurons with common gene expression programs. Sex-specific expression is overlaid on this program, to build the potential for vastly different sex-specific behaviors.
Collapse
Affiliation(s)
- Colleen M Palmateer
- Department of Biomedical Sciences, Florida State University, College of MedicineTallahasseeUnited States
| | - Catherina Artikis
- Department of Biomedical Sciences, Florida State University, College of MedicineTallahasseeUnited States
| | - Savannah G Brovero
- Department of Biomedical Sciences, Florida State University, College of MedicineTallahasseeUnited States
| | - Benjamin Friedman
- Department of Biomedical Sciences, Florida State University, College of MedicineTallahasseeUnited States
| | - Alexis Gresham
- Department of Biomedical Sciences, Florida State University, College of MedicineTallahasseeUnited States
| | - Michelle N Arbeitman
- Department of Biomedical Sciences, Florida State University, College of MedicineTallahasseeUnited States
- Program of Neuroscience, Florida State UniversityTallahasseeUnited States
| |
Collapse
|
20
|
Kozlov EN, Tokmatcheva EV, Khrustaleva AM, Grebenshchikov ES, Deev RV, Gilmutdinov RA, Lebedeva LA, Zhukova M, Savvateeva-Popova EV, Schedl P, Shidlovskii YV. Long-Term Memory Formation in Drosophila Depends on the 3'UTR of CPEB Gene orb2. Cells 2023; 12:cells12020318. [PMID: 36672258 PMCID: PMC9856895 DOI: 10.3390/cells12020318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/30/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Activation of local translation in neurites in response to stimulation is an important step in the formation of long-term memory (LTM). CPEB proteins are a family of translation factors involved in LTM formation. The Drosophila CPEB protein Orb2 plays an important role in the development and function of the nervous system. Mutations of the coding region of the orb2 gene have previously been shown to impair LTM formation. We found that a deletion of the 3'UTR of the orb2 gene similarly results in loss of LTM in Drosophila. As a result of the deletion, the content of the Orb2 protein remained the same in the neuron soma, but significantly decreased in synapses. Using RNA immunoprecipitation followed by high-throughput sequencing, we detected more than 6000 potential Orb2 mRNA targets expressed in the Drosophila brain. Importantly, deletion of the 3'UTR of orb2 mRNA also affected the localization of the Csp, Pyd, and Eya proteins, which are encoded by putative mRNA targets of Orb2. Therefore, the 3'UTR of the orb2 mRNA is important for the proper localization of Orb2 and other proteins in synapses of neurons and the brain as a whole, providing a molecular basis for LTM formation.
Collapse
Affiliation(s)
- Eugene N. Kozlov
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Elena V. Tokmatcheva
- Institute of Physiology, Russian Academy of Sciences, 188680 St. Petersburg, Russia
| | - Anastasia M. Khrustaleva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Eugene S. Grebenshchikov
- Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
| | - Roman V. Deev
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Rudolf A. Gilmutdinov
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Lyubov A. Lebedeva
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Mariya Zhukova
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | | | - Paul Schedl
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Molecular Biology, Princeton University, Princeton University, Princeton, NJ 08544-1014, USA
| | - Yulii V. Shidlovskii
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
- Correspondence:
| |
Collapse
|
21
|
Cortés E, Pak JS, Özkan E. Structure and evolution of neuronal wiring receptors and ligands. Dev Dyn 2023; 252:27-60. [PMID: 35727136 PMCID: PMC10084454 DOI: 10.1002/dvdy.512] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 01/04/2023] Open
Abstract
One of the fundamental properties of a neuronal circuit is the map of its connections. The cellular and developmental processes that allow for the growth of axons and dendrites, selection of synaptic targets, and formation of functional synapses use neuronal surface receptors and their interactions with other surface receptors, secreted ligands, and matrix molecules. Spatiotemporal regulation of the expression of these receptors and cues allows for specificity in the developmental pathways that wire stereotyped circuits. The families of molecules controlling axon guidance and synapse formation are generally conserved across animals, with some important exceptions, which have consequences for neuronal connectivity. Here, we summarize the distribution of such molecules across multiple taxa, with a focus on model organisms, evolutionary processes that led to the multitude of such molecules, and functional consequences for the diversification or loss of these receptors.
Collapse
Affiliation(s)
- Elena Cortés
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Joseph S Pak
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
22
|
Abstract
Among the many wonders of nature, the sense of smell of the fly Drosophila melanogaster might seem, at first glance, of esoteric interest. Nevertheless, for over a century, the 'nose' of this insect has been an extraordinary system to explore questions in animal behaviour, ecology and evolution, neuroscience, physiology and molecular genetics. The insights gained are relevant for our understanding of the sensory biology of vertebrates, including humans, and other insect species, encompassing those detrimental to human health. Here, I present an overview of our current knowledge of D. melanogaster olfaction, from molecules to behaviours, with an emphasis on the historical motivations of studies and illustration of how technical innovations have enabled advances. I also highlight some of the pressing and long-term questions.
Collapse
Affiliation(s)
- Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, CH-1015 Lausanne, Switzerland
| |
Collapse
|
23
|
Shuster SA, Li J, Chon UR, Sinantha-Hu MC, Luginbuhl DJ, Udeshi ND, Carey DK, Takeo YH, Xie Q, Xu C, Mani DR, Han S, Ting AY, Carr SA, Luo L. In situ cell-type-specific cell-surface proteomic profiling in mice. Neuron 2022; 110:3882-3896.e9. [PMID: 36220098 PMCID: PMC9742329 DOI: 10.1016/j.neuron.2022.09.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Cell-surface proteins (CSPs) mediate intercellular communication throughout the lives of multicellular organisms. However, there are no generalizable methods for quantitative CSP profiling in specific cell types in vertebrate tissues. Here, we present in situ cell-surface proteome extraction by extracellular labeling (iPEEL), a proximity labeling method in mice that enables spatiotemporally precise labeling of cell-surface proteomes in a cell-type-specific environment in native tissues for discovery proteomics. Applying iPEEL to developing and mature cerebellar Purkinje cells revealed differential enrichment in CSPs with post-translational protein processing and synaptic functions in the developing and mature cell-surface proteomes, respectively. A proteome-instructed in vivo loss-of-function screen identified a critical, multifaceted role for Armh4 in Purkinje cell dendrite morphogenesis. Armh4 overexpression also disrupts dendrite morphogenesis; this effect requires its conserved cytoplasmic domain and is augmented by disrupting its endocytosis. Our results highlight the utility of CSP profiling in native mammalian tissues for identifying regulators of cell-surface signaling.
Collapse
Affiliation(s)
- S Andrew Shuster
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Neurosciences Program, Stanford University, CA 94305, USA
| | - Jiefu Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - URee Chon
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Neurosciences Program, Stanford University, CA 94305, USA
| | - Miley C Sinantha-Hu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David J Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Namrata D Udeshi
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Yukari H Takeo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Qijing Xie
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Neurosciences Program, Stanford University, CA 94305, USA
| | - Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - D R Mani
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuo Han
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Alice Y Ting
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Steven A Carr
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
24
|
Transcriptional profiling from whole embryos to single neuroblast lineages in Drosophila. Dev Biol 2022; 489:21-33. [PMID: 35660371 PMCID: PMC9805786 DOI: 10.1016/j.ydbio.2022.05.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 01/03/2023]
Abstract
Embryonic development results in the production of distinct tissue types, and different cell types within each tissue. A major goal of developmental biology is to uncover the "parts list" of cell types that comprise each organ. Here we perform single cell RNA sequencing (scRNA-seq) of the Drosophila embryo to identify the genes that characterize different cell and tissue types during development. We assay three different timepoints, revealing a coordinated change in gene expression within each tissue. Interestingly, we find that the elav and Mhc genes, whose protein products are widely used as markers for neurons and muscles, respectively, show broad pan-embryonic expression, indicating the importance of post-transcriptional regulation. We next focus on the central nervous system (CNS), where we identify genes whose expression is enriched at each stage of neuronal differentiation: from neural progenitors, called neuroblasts, to their immediate progeny ganglion mother cells (GMCs), followed by new-born neurons, young neurons, and the most mature neurons. Finally, we ask whether the clonal progeny of a single neuroblast (NB7-1) share a similar transcriptional identity. Surprisingly, we find that clonal identity does not lead to transcriptional clustering, showing that neurons within a lineage are diverse, and that neurons with a similar transcriptional profile (e.g. motor neurons, glia) are distributed among multiple neuroblast lineages. Although each lineage consists of diverse progeny, we were able to identify a previously uncharacterized gene, Fer3, as an excellent marker for the NB7-1 lineage. Within the NB7-1 lineage, neurons which share a temporal identity (e.g. Hunchback, Kruppel, Pdm, and Castor temporal transcription factors in the NB7-1 lineage) have shared transcriptional features, allowing for the identification of candidate novel temporal factors or targets of the temporal transcription factors. In conclusion, we have characterized the embryonic transcriptome for all major tissue types and for three stages of development, as well as the first transcriptomic analysis of a single, identified neuroblast lineage, finding a lineage-enriched transcription factor.
Collapse
|
25
|
Dillon N, Cocanougher B, Sood C, Yuan X, Kohn AB, Moroz LL, Siegrist SE, Zlatic M, Doe CQ. Single cell RNA-seq analysis reveals temporally-regulated and quiescence-regulated gene expression in Drosophila larval neuroblasts. Neural Dev 2022; 17:7. [PMID: 36002894 PMCID: PMC9404614 DOI: 10.1186/s13064-022-00163-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
The mechanisms that generate neural diversity during development remains largely unknown. Here, we use scRNA-seq methodology to discover new features of the Drosophila larval CNS across several key developmental timepoints. We identify multiple progenitor subtypes - both stem cell-like neuroblasts and intermediate progenitors - that change gene expression across larval development, and report on new candidate markers for each class of progenitors. We identify a pool of quiescent neuroblasts in newly hatched larvae and show that they are transcriptionally primed to respond to the insulin signaling pathway to exit from quiescence, including relevant pathway components in the adjacent glial signaling cell type. We identify candidate "temporal transcription factors" (TTFs) that are expressed at different times in progenitor lineages. Our work identifies many cell type specific genes that are candidates for functional roles, and generates new insight into the differentiation trajectory of larval neurons.
Collapse
Affiliation(s)
- Noah Dillon
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, OR, 97403, Eugene, USA
| | - Ben Cocanougher
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Chhavi Sood
- Department of Biology, University of Virginia, VA, 22904, Charlottesville, USA
| | - Xin Yuan
- Department of Biology, University of Virginia, VA, 22904, Charlottesville, USA
| | - Andrea B Kohn
- Whitney Laboratory for Marine Biosciences, University of Florida, FL, 32080, St. Augustine, USA
| | - Leonid L Moroz
- Whitney Laboratory for Marine Biosciences, University of Florida, FL, 32080, St. Augustine, USA
| | - Sarah E Siegrist
- Department of Biology, University of Virginia, VA, 22904, Charlottesville, USA
| | - Marta Zlatic
- MRC Laboratory of Molecular Biology, Dept of Zoology, University of Cambridge, Cambridge, UK
- Janelia Research Campus, VA, Ashburn, USA
| | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, OR, 97403, Eugene, USA.
| |
Collapse
|
26
|
Abstract
Many insect cells are encapsulated within the exoskeleton and cannot be dissociated intact, making them inaccessible to single-cell transcriptomic profiling. We have used single-nucleus RNA sequencing to extract transcriptomic information from multiple Drosophila tissues. Here, we describe procedures for the (1) dissociation of single nuclei, (2) isolation of single nuclei using two popular cell sorters, and (3) preparation of libraries for Smart-seq2 and 10× Genomics. This protocol enables generation of high-quality transcriptomes from single nuclei and can be applied to other species. For complete details on the use and execution of this protocol, please refer to McLaughlin et al. (2021) and Li et al. (2022).
Collapse
|
27
|
Wang Y, Lobb-Rabe M, Ashley J, Chatterjee P, Anand V, Bellen HJ, Kanca O, Carrillo RA. Systematic expression profiling of Dpr and DIP genes reveals cell surface codes in Drosophila larval motor and sensory neurons. Development 2022; 149:dev200355. [PMID: 35502740 PMCID: PMC9188756 DOI: 10.1242/dev.200355] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 04/20/2022] [Indexed: 07/26/2023]
Abstract
In complex nervous systems, neurons must identify their correct partners to form synaptic connections. The prevailing model to ensure correct recognition posits that cell-surface proteins (CSPs) in individual neurons act as identification tags. Thus, knowing what cells express which CSPs would provide insights into neural development, synaptic connectivity, and nervous system evolution. Here, we investigated expression of Dpr and DIP genes, two CSP subfamilies belonging to the immunoglobulin superfamily, in Drosophila larval motor neurons (MNs), muscles, glia and sensory neurons (SNs) using a collection of GAL4 driver lines. We found that Dpr genes are more broadly expressed than DIP genes in MNs and SNs, and each examined neuron expresses a unique combination of Dpr and DIP genes. Interestingly, many Dpr and DIP genes are not robustly expressed, but are found instead in gradient and temporal expression patterns. In addition, the unique expression patterns of Dpr and DIP genes revealed three uncharacterized MNs. This study sets the stage for exploring the functions of Dpr and DIP genes in Drosophila MNs and SNs and provides genetic access to subsets of neurons.
Collapse
Affiliation(s)
- Yupu Wang
- Department of Molecular Genetics & Cellular Biology, University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, University of Chicago, Chicago, IL 60637, USA
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Meike Lobb-Rabe
- Department of Molecular Genetics & Cellular Biology, University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, University of Chicago, Chicago, IL 60637, USA
- Program in Cell and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - James Ashley
- Department of Molecular Genetics & Cellular Biology, University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, University of Chicago, Chicago, IL 60637, USA
| | - Purujit Chatterjee
- Department of Molecular Genetics & Cellular Biology, University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, University of Chicago, Chicago, IL 60637, USA
| | - Veera Anand
- Department of Molecular Genetics & Cellular Biology, University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, University of Chicago, Chicago, IL 60637, USA
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics and Jan and Dan Duncan Neurobiological Research Institute, Baylor College of Medicine (BCM), Houston, TX 77030, USA
- Department of Neuroscience and Howard Hughes Medical Institute, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics and Jan and Dan Duncan Neurobiological Research Institute, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Robert A. Carrillo
- Department of Molecular Genetics & Cellular Biology, University of Chicago, Chicago, IL 60637, USA
- Neuroscience Institute, University of Chicago, Chicago, IL 60637, USA
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL 60637, USA
- Program in Cell and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
28
|
Meltzer H, Schuldiner O. Spatiotemporal Control of Neuronal Remodeling by Cell Adhesion Molecules: Insights From Drosophila. Front Neurosci 2022; 16:897706. [PMID: 35645712 PMCID: PMC9135462 DOI: 10.3389/fnins.2022.897706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/22/2022] [Indexed: 01/26/2023] Open
Abstract
Developmental neuronal remodeling is required for shaping the precise connectivity of the mature nervous system. Remodeling involves pruning of exuberant neural connections, often followed by regrowth of adult-specific ones, as a strategy to refine neural circuits. Errors in remodeling are associated with neurodevelopmental disorders such as schizophrenia and autism. Despite its fundamental nature, our understanding of the mechanisms governing neuronal remodeling is far from complete. Specifically, how precise spatiotemporal control of remodeling and rewiring is achieved is largely unknown. In recent years, cell adhesion molecules (CAMs), and other cell surface and secreted proteins of various families, have been implicated in processes of neurite pruning and wiring specificity during circuit reassembly. Here, we review some of the known as well as speculated roles of CAMs in these processes, highlighting recent advances in uncovering spatiotemporal aspects of regulation. Our focus is on the fruit fly Drosophila, which is emerging as a powerful model in the field, due to the extensive, well-characterized and stereotypic remodeling events occurring throughout its nervous system during metamorphosis, combined with the wide and constantly growing toolkit to identify CAM binding and resulting cellular interactions in vivo. We believe that its many advantages pose Drosophila as a leading candidate for future breakthroughs in the field of neuronal remodeling in general, and spatiotemporal control by CAMs specifically.
Collapse
Affiliation(s)
- Hagar Meltzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
- *Correspondence: Hagar Meltzer,
| | - Oren Schuldiner
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
- Oren Schuldiner,
| |
Collapse
|
29
|
The branching code: A model of actin-driven dendrite arborization. Cell Rep 2022; 39:110746. [PMID: 35476974 DOI: 10.1016/j.celrep.2022.110746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/24/2021] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
The cytoskeleton is crucial for defining neuronal-type-specific dendrite morphologies. To explore how the complex interplay of actin-modulatory proteins (AMPs) can define neuronal types in vivo, we focused on the class III dendritic arborization (c3da) neuron of Drosophila larvae. Using computational modeling, we reveal that the main branches (MBs) of c3da neurons follow general models based on optimal wiring principles, while the actin-enriched short terminal branches (STBs) require an additional growth program. To clarify the cellular mechanisms that define this second step, we thus concentrated on STBs for an in-depth quantitative description of dendrite morphology and dynamics. Applying these methods systematically to mutants of six known and novel AMPs, we revealed the complementary roles of these individual AMPs in defining STB properties. Our data suggest that diverse dendrite arbors result from a combination of optimal-wiring-related growth and individualized growth programs that are neuron-type specific.
Collapse
|
30
|
Yang K, Liu T, Wang Z, Liu J, Shen Y, Pan X, Wen R, Xie H, Ruan Z, Tan Z, Chen Y, Guo A, Liu H, Han H, Di Z, Zhang K. Classifying Drosophila Olfactory Projection Neuron Boutons by Quantitative Analysis of Electron Microscopic Reconstruction. iScience 2022; 25:104180. [PMID: 35494235 PMCID: PMC9038572 DOI: 10.1016/j.isci.2022.104180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/25/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Kai Yang
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
- BNU-BUCM Hengqin Innovation Institute of Science and Technology, Zhuhai, Guangdong 518057, China
| | - Tong Liu
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Ze Wang
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Jing Liu
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yuxinyao Shen
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Xinyi Pan
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Ruyi Wen
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Haotian Xie
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Zhaoxuan Ruan
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Zixiao Tan
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Yingying Chen
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Aike Guo
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
- Huitong College, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - He Liu
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Hua Han
- Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Zengru Di
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
| | - Ke Zhang
- International Academic Center of Complex Systems, Advanced Institute of Natural Sciences, Beijing Normal University at Zhuhai, Zhuhai, Guangdong 519087, China
- Corresponding author
| |
Collapse
|
31
|
Cellular diversity and gene expression profiles in the male and female brain of Aedes aegypti. BMC Genomics 2022; 23:119. [PMID: 35144549 PMCID: PMC8832747 DOI: 10.1186/s12864-022-08327-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/18/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Aedes aegypti is a medically-important mosquito vector that transmits arboviruses including yellow fever, dengue, chikungunya, and Zika viruses to humans. The mosquito exhibits typical sexually dimorphic behaviors such as courtship, mating, host seeking, bloodfeeding, and oviposition. All these behaviors are mainly regulated by the brain; however, little is known about the function and neuron composition of the mosquito brain. In this study, we generated an initial atlas of the adult male and female brain of Ae. aegypti using 10xGenomics based single-nucleus RNA sequencing. RESULTS We identified 35 brain cell clusters in male and female brains, and 15 of those clusters were assigned to known cell types. Identified cell types include glia (astrocytes), Kenyon cells, (ventral) projection neurons, monoaminergic neurons, medulla neurons, and proximal medulla neurons. In addition, the cell type compositions of male and female brains were compared to each other showing that they were quantitatively distinct, as 17 out of 35 cell clusters varied significantly in their cell type proportions. Overall, the transcriptomes from each cell cluster looked very similar between the male and female brain as only up to 25 genes were differentially expressed in these clusters. The sex determination factor Nix was highly expressed in neurons and glia of the male brain, whereas doublesex (dsx) was expressed in all neuron and glia cell clusters of the male and female brain. CONCLUSIONS An initial cell atlas of the brain of the mosquito Ae. aegypti has been generated showing that the cellular compositions of the male and female brains of this hematophagous insect differ significantly from each other. Although some of the rare brain cell types have not been detected in our single biological replicate, this study provides an important basis for the further development of a complete brain cell atlas as well as a better understanding of the neurobiology of the brains of male and female mosquitoes and their sexually dimorphic behaviors.
Collapse
|
32
|
Li T, Fu TM, Wong KKL, Li H, Xie Q, Luginbuhl DJ, Wagner MJ, Betzig E, Luo L. Cellular bases of olfactory circuit assembly revealed by systematic time-lapse imaging. Cell 2021; 184:5107-5121.e14. [PMID: 34551316 DOI: 10.1016/j.cell.2021.08.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/21/2021] [Accepted: 08/24/2021] [Indexed: 10/20/2022]
Abstract
Neural circuit assembly features simultaneous targeting of numerous neuronal processes from constituent neuron types, yet the dynamics is poorly understood. Here, we use the Drosophila olfactory circuit to investigate dynamic cellular processes by which olfactory receptor neurons (ORNs) target axons precisely to specific glomeruli in the ipsi- and contralateral antennal lobes. Time-lapse imaging of individual axons from 30 ORN types revealed a rich diversity in extension speed, innervation timing, and ipsilateral branch locations and identified that ipsilateral targeting occurs via stabilization of transient interstitial branches. Fast imaging using adaptive optics-corrected lattice light-sheet microscopy showed that upon approaching target, many ORN types exhibiting "exploring branches" consisted of parallel microtubule-based terminal branches emanating from an F-actin-rich hub. Antennal nerve ablations uncovered essential roles for bilateral axons in contralateral target selection and for ORN axons to facilitate dendritic refinement of postsynaptic partner neurons. Altogether, these observations provide cellular bases for wiring specificity establishment.
Collapse
Affiliation(s)
- Tongchao Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| | - Tian-Ming Fu
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20417, USA
| | - Kenneth Kin Lam Wong
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Hongjie Li
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Qijing Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - David J Luginbuhl
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Mark J Wagner
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Eric Betzig
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20417, USA; Departments of Molecular and Cell Biology and Physics, Howard Hughes Medical Institute, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
33
|
Mohr SE, Tattikota SG, Xu J, Zirin J, Hu Y, Perrimon N. Methods and tools for spatial mapping of single-cell RNAseq clusters in Drosophila. Genetics 2021; 217:6156631. [PMID: 33713129 DOI: 10.1093/genetics/iyab019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/02/2021] [Indexed: 01/26/2023] Open
Abstract
Single-cell RNA sequencing (scRNAseq) experiments provide a powerful means to identify clusters of cells that share common gene expression signatures. A major challenge in scRNAseq studies is to map the clusters to specific anatomical regions along the body and within tissues. Existing data, such as information obtained from large-scale in situ RNA hybridization studies, cell type specific transcriptomics, gene expression reporters, antibody stainings, and fluorescent tagged proteins, can help to map clusters to anatomy. However, in many cases, additional validation is needed to precisely map the spatial location of cells in clusters. Several approaches are available for spatial resolution in Drosophila, including mining of existing datasets, and use of existing or new tools for direct or indirect detection of RNA, or direct detection of proteins. Here, we review available resources and emerging technologies that will facilitate spatial mapping of scRNAseq clusters at high resolution in Drosophila. Importantly, we discuss the need, available approaches, and reagents for multiplexing gene expression detection in situ, as in most cases scRNAseq clusters are defined by the unique coexpression of sets of genes.
Collapse
Affiliation(s)
- Stephanie E Mohr
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sudhir Gopal Tattikota
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Xu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan Zirin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.,Howard Hughes Medical Institute, Boston, MA 02115, USA
| |
Collapse
|
34
|
McLaughlin CN, Brbić M, Xie Q, Li T, Horns F, Kolluru SS, Kebschull JM, Vacek D, Xie A, Li J, Jones RC, Leskovec J, Quake SR, Luo L, Li H. Single-cell transcriptomes of developing and adult olfactory receptor neurons in Drosophila. eLife 2021; 10:e63856. [PMID: 33555999 PMCID: PMC7870146 DOI: 10.7554/elife.63856] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Recognition of environmental cues is essential for the survival of all organisms. Transcriptional changes occur to enable the generation and function of the neural circuits underlying sensory perception. To gain insight into these changes, we generated single-cell transcriptomes of Drosophila olfactory- (ORNs), thermo-, and hygro-sensory neurons at an early developmental and adult stage using single-cell and single-nucleus RNA sequencing. We discovered that ORNs maintain expression of the same olfactory receptors across development. Using receptor expression and computational approaches, we matched transcriptomic clusters corresponding to anatomically and physiologically defined neuron types across multiple developmental stages. We found that cell-type-specific transcriptomes partly reflected axon trajectory choices in development and sensory modality in adults. We uncovered stage-specific genes that could regulate the wiring and sensory responses of distinct ORN types. Collectively, our data reveal transcriptomic features of sensory neuron biology and provide a resource for future studies of their development and physiology.
Collapse
Affiliation(s)
- Colleen N McLaughlin
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Maria Brbić
- Department of Computer Science, Stanford UniversityStanfordUnited States
| | - Qijing Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
- Neurosciences Graduate Program, Stanford UniversityStanfordUnited States
| | - Tongchao Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Felix Horns
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Biophysics Graduate Program, Stanford UniversityStanfordUnited States
| | - Sai Saroja Kolluru
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubStanfordUnited States
| | - Justus M Kebschull
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - David Vacek
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Anthony Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Jiefu Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
- Biology Graduate Program, Stanford UniversityStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Jure Leskovec
- Department of Computer Science, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubStanfordUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - Liqun Luo
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Hongjie Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|