1
|
Garbett K, Tosun B, Lopez JM, Smith CM, Honkanen K, Sando RC. Synaptic Gα12/13 signaling establishes hippocampal PV inhibitory circuits. Proc Natl Acad Sci U S A 2024; 121:e2407828121. [PMID: 39693341 DOI: 10.1073/pnas.2407828121] [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: 04/18/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Combinatorial networks of cell adhesion molecules and cell surface receptors drive fundamental aspects of neural circuit establishment and function. However, the intracellular signals orchestrated by these cell surface complexes remain less understood. Here, we report that the Gα12/13 pathway lies downstream of several GPCRs with critical synaptic functions. Impairment of the Gα12/13 pathway in postnatal hippocampal neurons diminishes inhibitory inputs without altering neuronal morphology or excitatory transmission. Gα12/13 signaling in hippocampal CA1 neurons in vivo selectively regulates PV interneuron synaptic connectivity, supporting an inhibitory synapse subtype-specific function of this pathway. Our studies establish Gα12/13 as a signaling node that shapes inhibitory hippocampal circuitry.
Collapse
Affiliation(s)
- Krassimira Garbett
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240
| | - Baris Tosun
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240
| | - Jaybree M Lopez
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240
| | - Cassandra M Smith
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240
| | - Kelly Honkanen
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240
| | - Richard C Sando
- Department of Pharmacology, Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240
| |
Collapse
|
2
|
Xu C, Luo L. The sparse driver system for in vivo single-cell labeling and manipulation in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626507. [PMID: 39677673 PMCID: PMC11642855 DOI: 10.1101/2024.12.02.626507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
In this protocol, we introduce a sparse driver system for cell-type specific single-cell labeling and manipulation in Drosophila, enabling complete and simultaneous expression of multiple transgenes in the same cells. The system precisely controls expression probability and sparsity via mutant FRT sites with reduced recombination efficiency and tunable FLP levels adjusted by heat-shock durations. We demonstrate that this generalizable toolkit enables tunable sparsity, multi-color staining, single-cell trans-synaptic tracing, single-cell manipulation, and in vivo analysis of cell-autonomous gene function. For details on the use and execution of this protocol, please refer to Xu et al. 2024.
Collapse
Affiliation(s)
- Chuanyun Xu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
- Biology Graduate Program, Stanford University, CA 94305, USA
- Lead contact
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
3
|
Xu C, Li Z, Lyu C, Hu Y, McLaughlin CN, Wong KKL, Xie Q, Luginbuhl DJ, Li H, Udeshi ND, Svinkina T, Mani DR, Han S, Li T, Li Y, Guajardo R, Ting AY, Carr SA, Li J, Luo L. Molecular and cellular mechanisms of teneurin signaling in synaptic partner matching. Cell 2024; 187:5081-5101.e19. [PMID: 38996528 DOI: 10.1016/j.cell.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/20/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024]
Abstract
In developing brains, axons exhibit remarkable precision in selecting synaptic partners among many non-partner cells. Evolutionarily conserved teneurins are transmembrane proteins that instruct synaptic partner matching. However, how intracellular signaling pathways execute teneurins' functions is unclear. Here, we use in situ proximity labeling to obtain the intracellular interactome of a teneurin (Ten-m) in the Drosophila brain. Genetic interaction studies using quantitative partner matching assays in both olfactory receptor neurons (ORNs) and projection neurons (PNs) reveal a common pathway: Ten-m binds to and negatively regulates a RhoGAP, thus activating the Rac1 small GTPases to promote synaptic partner matching. Developmental analyses with single-axon resolution identify the cellular mechanism of synaptic partner matching: Ten-m signaling promotes local F-actin levels and stabilizes ORN axon branches that contact partner PN dendrites. Combining spatial proteomics and high-resolution phenotypic analyses, this study advanced our understanding of both cellular and molecular mechanisms of synaptic partner matching.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Cheng Lyu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Yixin Hu
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Colleen N McLaughlin
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Kenneth Kin Lam Wong
- 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
| | - David J Luginbuhl
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Hongjie Li
- 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
| | - Tanya Svinkina
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - D R Mani
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shuo Han
- Departments of Genetics, Biology, and Chemistry, Chan Zuckerberg Biohub, Stanford University, Stanford, CA 94305, USA
| | - Tongchao Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Yang Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Guajardo
- Department of Biology and Howard Hughes Medical Institute, 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
| | - Jiefu Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Biology Graduate Program, Stanford University, Stanford, CA 94305, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
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
|
5
|
Inal MA, Banzai K, Kamiyama R, Kamiyama D. Cell-type-specific Labeling of Endogenous Proteins Using the Split GFP System in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592806. [PMID: 38826199 PMCID: PMC11140830 DOI: 10.1101/2024.05.06.592806] [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: 06/04/2024]
Abstract
i. Accurate identification of the locations of endogenous proteins is crucial for understanding their functions in tissues and cells. However, achieving precise cell-type-specific labeling of proteins has been challenging in vivo . A notable solution to this challenge is the self-complementing split green fluorescent protein (GFP 1-10/11 ) system. In this paper, we present a detailed protocol for labeling endogenous proteins in a cell-type-specific manner using the GFP 1-10/11 system in fruit flies. This approach depends on the reconstitution of the GFP 1-10 and GFP 11 fragments, creating a fluorescence signal. We insert the GFP 11 fragment into a specific genomic locus while expressing its counterpart, GFP 1-10 , through an available Gal4 driver line. The unique aspect of this system is that neither GFP 1-10 nor GFP 11 alone emits fluorescence, enabling the precise detection of protein localization only in Gal4-positive cells expressing the GFP 11 tagged endogenous protein. We illustrate this technique using the adhesion molecule gene teneurin-m ( Ten-m ) as a model, highlighting the generation and validation of GFP 11 protein trap lines via Minos-mediated integration cassette (MiMIC) insertion. Furthermore, we demonstrate the cell-type-specific labeling of Ten-m proteins in the larval brains of fruit flies. This method significantly enhances our ability to image endogenous protein localization patterns in a cell-type-specific manner and is adaptable to various model organisms beyond fruit flies.
Collapse
|
6
|
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
|
7
|
Vien KM, Duan Q, Yeung C, Barish S, Volkan PC. Atypical cadherin, Fat2, regulates axon terminal organization in the developing Drosophila olfactory receptor neurons. iScience 2024; 27:110340. [PMID: 39055932 PMCID: PMC11269957 DOI: 10.1016/j.isci.2024.110340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/08/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
The process of how neuronal identity confers circuit organization is intricately related to the mechanisms underlying neurodegeneration and neuropathologies. Modeling this process, the olfactory circuit builds a functionally organized topographic map, which requires widely dispersed neurons with the same identity to converge their axons into one a class-specific neuropil, a glomerulus. In this article, we identified Fat2 (also known as Kugelei) as a regulator of class-specific axon organization. In fat2 mutants, axons belonging to the highest fat2-expressing classes present with a more severe phenotype compared to axons belonging to low fat2-expressing classes. In extreme cases, mutations lead to neural degeneration. Lastly, we found that Fat2 intracellular domain interactors, APC1/2 (Adenomatous polyposis coli) and dop (Drop out), likely orchestrate the cytoskeletal remodeling required for axon condensation. Altogether, we provide a potential mechanism for how cell surface proteins' regulation of cytoskeletal remodeling necessitates identity specific circuit organization.
Collapse
Affiliation(s)
- Khanh M. Vien
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Qichen Duan
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Chun Yeung
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Scott Barish
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Pelin Cayirlioglu Volkan
- Department of Biology, Duke University, Durham, NC 27708, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
8
|
Maraslioglu-Sperber A, Pizzi E, Fisch JO, Kattler K, Ritter T, Friauf E. Molecular and functional profiling of cell diversity and identity in the lateral superior olive, an auditory brainstem center with ascending and descending projections. Front Cell Neurosci 2024; 18:1354520. [PMID: 38846638 PMCID: PMC11153811 DOI: 10.3389/fncel.2024.1354520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/15/2024] [Indexed: 06/09/2024] Open
Abstract
The lateral superior olive (LSO), a prominent integration center in the auditory brainstem, contains a remarkably heterogeneous population of neurons. Ascending neurons, predominantly principal neurons (pLSOs), process interaural level differences for sound localization. Descending neurons (lateral olivocochlear neurons, LOCs) provide feedback into the cochlea and are thought to protect against acoustic overload. The molecular determinants of the neuronal diversity in the LSO are largely unknown. Here, we used patch-seq analysis in mice at postnatal days P10-12 to classify developing LSO neurons according to their functional and molecular profiles. Across the entire sample (n = 86 neurons), genes involved in ATP synthesis were particularly highly expressed, confirming the energy expenditure of auditory neurons. Two clusters were identified, pLSOs and LOCs. They were distinguished by 353 differentially expressed genes (DEGs), most of which were novel for the LSO. Electrophysiological analysis confirmed the transcriptomic clustering. We focused on genes affecting neuronal input-output properties and validated some of them by immunohistochemistry, electrophysiology, and pharmacology. These genes encode proteins such as osteopontin, Kv11.3, and Kvβ3 (pLSO-specific), calcitonin-gene-related peptide (LOC-specific), or Kv7.2 and Kv7.3 (no DEGs). We identified 12 "Super DEGs" and 12 genes showing "Cluster similarity." Collectively, we provide fundamental and comprehensive insights into the molecular composition of individual ascending and descending neurons in the juvenile auditory brainstem and how this may relate to their specific functions, including developmental aspects.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Erika Pizzi
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Jonas O. Fisch
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Kathrin Kattler
- Genetics/Epigenetics Group, Department of Biological Sciences, Saarland University, Saarbrücken, Germany
| | - Tamara Ritter
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
9
|
Liakath-Ali K, Refaee R, Südhof TC. Cartography of teneurin and latrophilin expression reveals spatiotemporal axis heterogeneity in the mouse hippocampus during development. PLoS Biol 2024; 22:e3002599. [PMID: 38713721 PMCID: PMC11101112 DOI: 10.1371/journal.pbio.3002599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 05/17/2024] [Accepted: 03/26/2024] [Indexed: 05/09/2024] Open
Abstract
Synaptic adhesion molecules (SAMs) are evolutionarily conserved proteins that play an important role in the form and function of neuronal synapses. Teneurins (Tenms) and latrophilins (Lphns) are well-known cell adhesion molecules that form a transsynaptic complex. Recent studies suggest that Tenm3 and Lphn2 (gene symbol Adgrl2) are involved in hippocampal circuit assembly via their topographical expression. However, it is not known whether other teneurins and latrophilins display similar topographically restricted expression patterns during embryonic and postnatal development. Here, we reveal the cartography of all teneurin (Tenm1-4) and latrophilin (Lphn1-3 [Adgrl1-3]) paralog expression in the mouse hippocampus across prenatal and postnatal development as monitored by large-scale single-molecule RNA in situ hybridization mapping. Our results identify a striking heterogeneity in teneurin and latrophilin expression along the spatiotemporal axis of the hippocampus. Tenm2 and Tenm4 expression levels peak at the neonatal stage when compared to Tenm1 and Tenm3, while Tenm1 expression is restricted to the postnatal pyramidal cell layer. Tenm4 expression in the dentate gyrus (DG) exhibits an opposing topographical expression pattern in the embryonic and neonatal hippocampus. Our findings were validated by analyses of multiple RNA-seq datasets at bulk, single-cell, and spatial levels. Thus, our study presents a comprehensive spatiotemporal map of Tenm and Lphn expression in the hippocampus, showcasing their diverse expression patterns across developmental stages in distinct spatial axes.
Collapse
Affiliation(s)
- Kif Liakath-Ali
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Rebecca Refaee
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
- Howard Hughes Medical Institute, Stanford University, Stanford, California, United States of America
| |
Collapse
|
10
|
Wolterhoff N, Hiesinger PR. Synaptic promiscuity in brain development. Curr Biol 2024; 34:R102-R116. [PMID: 38320473 PMCID: PMC10849093 DOI: 10.1016/j.cub.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Precise synaptic connectivity is a prerequisite for the function of neural circuits, yet individual neurons, taken out of their developmental context, readily form unspecific synapses. How does the genome encode brain wiring in light of this apparent contradiction? Synaptic specificity is the outcome of a long series of developmental processes and mechanisms before, during and after synapse formation. How much promiscuity is permissible or necessary at the moment of synaptic partner choice depends on the extent to which prior development restricts available partners or subsequent development corrects initially made synapses. Synaptic promiscuity at the moment of choice can thereby play important roles in the development of precise connectivity, but also facilitate developmental flexibility and robustness. In this review, we assess the experimental evidence for the prevalence and roles of promiscuous synapse formation during brain development. Many well-established experimental approaches are based on developmental genetic perturbation and an assessment of synaptic connectivity only in the adult; this can make it difficult to pinpoint when a given defect or mechanism occurred. In many cases, such studies reveal mechanisms that restrict partner availability already prior to synapse formation. Subsequently, at the moment of choice, factors including synaptic competency, interaction dynamics and molecular recognition further restrict synaptic partners. The discussion of the development of synaptic specificity through the lens of synaptic promiscuity suggests an algorithmic process based on neurons capable of promiscuous synapse formation that are continuously prevented from making the wrong choices, with no single mechanism or developmental time point sufficient to explain the outcome.
Collapse
Affiliation(s)
- Neele Wolterhoff
- Division of Neurobiology, Free University Berlin, 14195 Berlin, Germany
| | - P Robin Hiesinger
- Division of Neurobiology, Free University Berlin, 14195 Berlin, Germany.
| |
Collapse
|
11
|
Hunyara JL, Daly KM, Torres K, Yurgel ME, Komal R, Hattar S, Kolodkin AL. Teneurin-3 regulates the generation of non-image-forming visual circuitry and responsiveness to light in the suprachiasmatic nucleus. PLoS Biol 2023; 21:e3002412. [PMID: 38048352 PMCID: PMC10729976 DOI: 10.1371/journal.pbio.3002412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 12/19/2023] [Accepted: 10/31/2023] [Indexed: 12/06/2023] Open
Abstract
Visual system function depends upon the elaboration of precise connections between retinal ganglion cell (RGC) axons and their central targets in the brain. Though some progress has been made in defining the molecules that regulate RGC connectivity required for the assembly and function of image-forming circuitry, surprisingly little is known about factors required for intrinsically photosensitive RGCs (ipRGCs) to target a principal component of the non-image-forming circuitry: the suprachiasmatic nucleus (SCN). Furthermore, the molecules required for forming circuits critical for circadian behaviors within the SCN are not known. We observe here that the adhesion molecule teneurin-3 (Tenm3) is highly expressed in vasoactive intestinal peptide (VIP) neurons located in the core region of the SCN. Since Tenm3 is required for other aspects of mammalian visual system development, we investigate roles for Tenm3 in regulating ipRGC-SCN connectivity and function. Our results show that Tenm3 negatively regulates association between VIP and arginine vasopressin (AVP) neurons within the SCN and is essential for M1 ipRGC axon innervation to the SCN. Specifically, in Tenm3-/- mice, we find a reduction in ventro-medial innervation to the SCN. Despite this reduction, Tenm3-/- mice have higher sensitivity to light and faster re-entrainment to phase advances, probably due to the increased association between VIP and AVP neurons. These data show that Tenm3 plays key roles in elaborating non-image-forming visual system circuitry and that it influences murine responses to phase-advancing light stimuli.
Collapse
Affiliation(s)
- John L. Hunyara
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - K. M. Daly
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Katherine Torres
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Maria E. Yurgel
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ruchi Komal
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Samer Hattar
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alex L. Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Kavli Neuroscience Discovery Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
12
|
Randolph EC, Fieber LA. Improvements in operant memory of Aplysia are correlated with age and specific gene expression. Front Behav Neurosci 2023; 17:1221794. [PMID: 37936650 PMCID: PMC10626442 DOI: 10.3389/fnbeh.2023.1221794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023] Open
Abstract
The transcription factor Aplysia CCAAT/enhancer binding protein (ApC/EBP) is expressed as an immediate early gene in the cAMP responsive element binding protein (CREB) mediated gene cascade, and it has essential functions in the synaptic consolidation of memory following a learning event. Synaptic consolidation primarily involves morphological changes at neuronal synapses, which are facilitated through the reorganization of the actin and microtubular cytoarchitecture of the cell. During early nervous system development, the transmembrane synaptic protein teneurin acts directly upon neuronal presynaptic microtubules and postsynaptic spectrin-based cytoskeletons to facilitate the creation of new synapses. It is reasonable to hypothesize that teneurin may also be linked to learning-induced synaptic changes and is a potential candidate to be a later gene expressed in the CREB-mediated gene cascade downstream of ApC/EBP. To assess the role of ApC/EBP and teneurin in learning and memory in the marine snail Aplysia californica, young (age 7-8 months) and aged (age 13-15 months; aging stage AII) siblings of Aplysia were trained in an operant conditioning paradigm-learning food is inedible (LFI)-over 2 days, during which they learned to modify the feeding reflex. Aged Aplysia had enhanced performance of the LFI task on the second day than younger siblings although far more aged animals were excluded from the analysis because of the initial failure in learning to recognize the inedible probe. After 2 days of training, ApC/EBP isoform X1 mRNA and teneurin mRNA were quantified in selected neurons of the buccal ganglia, the locus of neural circuits in LFI. Teneurin expression was elevated in aged Aplysia compared to young siblings regardless of training. ApC/EBP isoform X1 expression was significantly higher in untrained aged animals than in untrained young siblings but decreased in trained aged animals compared to untrained aged animals. Elevated levels of ApC/EBP isoform X1 and teneurin mRNA before training may have contributed to the enhancement of LFI performance in the aged animals that successfully learned.
Collapse
Affiliation(s)
| | - Lynne A. Fieber
- Department of Marine Biology and Ecology, University of Miami Rosenstiel School, Miami, FL, United States
| |
Collapse
|
13
|
Tran H, Sawatari A, Leamey CA. Ten-m3 plays a role in the formation of thalamostriatal projections. Dev Neurobiol 2023; 83:255-267. [PMID: 37700636 DOI: 10.1002/dneu.22927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 06/28/2023] [Accepted: 08/25/2023] [Indexed: 09/14/2023]
Abstract
The importance of the thalamostriatal pathway for a myriad of brain functions is becoming increasingly apparent. Little is known about the formation of this pathway in mice. Further, while Ten-m3, a member of the Ten-m/teneurin/Odz family, is implicated in the proper wiring of mature thalamostriatal projections, its developmental time course is unknown. Here, we describe the normal development of thalamostriatal projections arising from the parafascicular nucleus (PFN) and show a role for Ten-m3 in its formation. Ten-m3 is expressed in both the PFN and the striatum by embryonic day 17 (E17). By postnatal day 3 (P3), it had a patchy appearance in the striatum, overlaid on a high dorsal-low ventral expression gradient in both structures. In wild-type mice, axons from the PFN begin to innervate the striatum by E17. By P3, terminals had ramified but were not confined to any striatal subregion. By P7, the axons had begun to avoid striosomes. The first indication of clustering of thalamic terminals within the striatal matrix was also seen at this time point. The compartmental targeting and clustering of PFN projections became more apparent by P10. Analysis of Ten-m3 knockout mice showed that while the early developmental progression of the thalamostriatal pathway is conserved, by P10 differences emerged, with a loss of topographic precision and the absence of terminal clustering. No evidence of the involvement of EphA7 downstream of Ten-m3 was found. Overall, our results suggest that Ten-m3 plays a role in the consolidation and refinement of thalamic axons to a specific subregion of the striatal matrix.
Collapse
Affiliation(s)
- Heidi Tran
- School of Medical Science, FMH, University of Sydney, Sydney, New South Wales, Australia
| | - Atomu Sawatari
- School of Medical Science, FMH, University of Sydney, Sydney, New South Wales, Australia
| | - Catherine A Leamey
- School of Medical Science, FMH, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
14
|
Kuebler CA, Paré AC. Striped Expression of Leucine-Rich Repeat Proteins Coordinates Cell Intercalation and Compartment Boundary Formation in the Early Drosophila Embryo. Symmetry (Basel) 2023; 15:1490. [PMID: 38650964 PMCID: PMC11034934 DOI: 10.3390/sym15081490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Planar polarity is a commonly observed phenomenon in which proteins display a consistent asymmetry in their subcellular localization or activity across the plane of a tissue. During animal development, planar polarity is a fundamental mechanism for coordinating the behaviors of groups of cells to achieve anisotropic tissue remodeling, growth, and organization. Therefore, a primary focus of developmental biology research has been to understand the molecular mechanisms underlying planar polarity in a variety of systems to identify conserved principles of tissue organization. In the early Drosophila embryo, the germband neuroectoderm epithelium rapidly doubles in length along the anterior-posterior axis through a process known as convergent extension (CE); it also becomes subdivided into tandem tissue compartments through the formation of compartment boundaries (CBs). Both processes are dependent on the planar polarity of proteins involved in cellular tension and adhesion. The enrichment of actomyosin-based tension and adherens junction-based adhesion at specific cell-cell contacts is required for coordinated cell intercalation, which drives CE, and the creation of highly stable cell-cell contacts at CBs. Recent studies have revealed a system for rapid cellular polarization triggered by the expression of leucine-rich-repeat (LRR) cell-surface proteins in striped patterns. In particular, the non-uniform expression of Toll-2, Toll-6, Toll-8, and Tartan generates local cellular asymmetries that allow cells to distinguish between cell-cell contacts oriented parallel or perpendicular to the anterior-posterior axis. In this review, we discuss (1) the biomechanical underpinnings of CE and CB formation, (2) how the initial symmetry-breaking events of anterior-posterior patterning culminate in planar polarity, and (3) recent advances in understanding the molecular mechanisms downstream of LRR receptors that lead to planar polarized tension and junctional adhesion.
Collapse
Affiliation(s)
- Chloe A. Kuebler
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Adam C. Paré
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
15
|
Ahmed M, Rajagopalan AE, Pan Y, Li Y, Williams DL, Pedersen EA, Thakral M, Previero A, Close KC, Christoforou CP, Cai D, Turner GC, Clowney EJ. Input density tunes Kenyon cell sensory responses in the Drosophila mushroom body. Curr Biol 2023; 33:2742-2760.e12. [PMID: 37348501 PMCID: PMC10529417 DOI: 10.1016/j.cub.2023.05.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/02/2023] [Accepted: 05/26/2023] [Indexed: 06/24/2023]
Abstract
The ability to discriminate sensory stimuli with overlapping features is thought to arise in brain structures called expansion layers, where neurons carrying information about sensory features make combinatorial connections onto a much larger set of cells. For 50 years, expansion coding has been a prime topic of theoretical neuroscience, which seeks to explain how quantitative parameters of the expansion circuit influence sensory sensitivity, discrimination, and generalization. Here, we investigate the developmental events that produce the quantitative parameters of the arthropod expansion layer, called the mushroom body. Using Drosophila melanogaster as a model, we employ genetic and chemical tools to engineer changes to circuit development. These allow us to produce living animals with hypothesis-driven variations on natural expansion layer wiring parameters. We then test the functional and behavioral consequences. By altering the number of expansion layer neurons (Kenyon cells) and their dendritic complexity, we find that input density, but not cell number, tunes neuronal odor selectivity. Simple odor discrimination behavior is maintained when the Kenyon cell number is reduced and augmented by Kenyon cell number expansion. Animals with increased input density to each Kenyon cell show increased overlap in Kenyon cell odor responses and become worse at odor discrimination tasks.
Collapse
Affiliation(s)
- Maria Ahmed
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Adithya E Rajagopalan
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yijie Pan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ye Li
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Donnell L Williams
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erik A Pedersen
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Manav Thakral
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Angelica Previero
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kari C Close
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | | | - Dawen Cai
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48104, USA; Biophysics LS&A, University of Michigan, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute Affiliate, University of Michigan, Ann Arbor, MI 48109, USA
| | - Glenn C Turner
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | - E Josephine Clowney
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA; Michigan Neuroscience Institute Affiliate, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
16
|
Parisi MJ, Aimino MA, Mosca TJ. A conditional strategy for cell-type-specific labeling of endogenous excitatory synapses in Drosophila. CELL REPORTS METHODS 2023; 3:100477. [PMID: 37323572 PMCID: PMC10261928 DOI: 10.1016/j.crmeth.2023.100477] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/28/2023] [Accepted: 04/19/2023] [Indexed: 06/17/2023]
Abstract
Chemical neurotransmission occurs at specialized contacts where neurotransmitter release machinery apposes neurotransmitter receptors to underlie circuit function. A series of complex events underlies pre- and postsynaptic protein recruitment to neuronal connections. To better study synaptic development in individual neurons, we need cell-type-specific strategies to visualize endogenous synaptic proteins. Although presynaptic strategies exist, postsynaptic proteins remain less studied because of a paucity of cell-type-specific reagents. To study excitatory postsynapses with cell-type specificity, we engineered dlg1[4K], a conditionally labeled marker of Drosophila excitatory postsynaptic densities. With binary expression systems, dlg1[4K] labels central and peripheral postsynapses in larvae and adults. Using dlg1[4K], we find that distinct rules govern postsynaptic organization in adult neurons, multiple binary expression systems can concurrently label pre- and postsynapse in a cell-type-specific manner, and neuronal DLG1 can sometimes localize presynaptically. These results validate our strategy for conditional postsynaptic labeling and demonstrate principles of synaptic organization.
Collapse
Affiliation(s)
- Michael J. Parisi
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Michael A. Aimino
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| | - Timothy J. Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Bluemle Life Sciences Building, Philadelphia, PA 19107, USA
| |
Collapse
|
17
|
Duan Q, Estrella R, Carson A, Chen Y, Volkan PC. The effect of Drosophila attP40 background on the glomerular organization of Or47b olfactory receptor neurons. G3 (BETHESDA, MD.) 2023; 13:jkad022. [PMID: 36695023 PMCID: PMC10085800 DOI: 10.1093/g3journal/jkad022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/26/2023]
Abstract
Bacteriophage integrase-directed insertion of transgenic constructs into specific genomic loci has been widely used by Drosophila community. The attP40 landing site located on the second chromosome gained popularity because of its high inducible transgene expression levels. Here, unexpectedly, we found that homozygous attP40 chromosome disrupts normal glomerular organization of Or47b olfactory receptor neuron (ORN) class in Drosophila. This effect is not likely to be caused by the loss of function of Msp300, where the attP40 docking site is inserted. Moreover, the attP40 background seems to genetically interact with the second chromosome Or47b-GAL4 driver, which results in a similar glomerular defect. Whether the ORN phenotype is caused by the neighbouring genes around Msp300 locus in the presence of attP40-based insertions or a second unknown mutation in the attP40 background remains elusive. Our findings tell a cautionary tale about using this popular transgenic landing site, highlighting the importance of rigorous controls to rule out the attP40 landing site-associated background effects.
Collapse
Affiliation(s)
- Qichen Duan
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Rachel Estrella
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Allison Carson
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Yang Chen
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Pelin C Volkan
- Department of Biology, Duke University, Durham, NC 27708, USA
| |
Collapse
|
18
|
Osaka J, Yasuda H, Watanuki Y, Kato Y, Nitta Y, Sugie A, Sato M, Suzuki T. Identification of genes regulating stimulus-dependent synaptic assembly in Drosophila using an automated synapse quantification system. Genes Genet Syst 2023; 97:297-309. [PMID: 36878557 DOI: 10.1266/ggs.22-00114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Neural activity-dependent synaptic plasticity is an important physiological phenomenon underlying environmental adaptation, memory and learning. However, its molecular basis, especially in presynaptic neurons, is not well understood. Previous studies have shown that the number of presynaptic active zones in the Drosophila melanogaster photoreceptor R8 is reversibly changed in an activity-dependent manner. During reversible synaptic changes, both synaptic disassembly and assembly processes were observed. Although we have established a paradigm for screening molecules involved in synaptic stability and several genes have been identified, genes involved in stimulus-dependent synaptic assembly are still elusive. Therefore, the aim of this study was to identify genes regulating stimulus-dependent synaptic assembly in Drosophila using an automated synapse quantification system. To this end, we performed RNAi screening against 300 memory-defective, synapse-related or transmembrane molecules in photoreceptor R8 neurons. Candidate genes were narrowed down to 27 genes in the first screen using presynaptic protein aggregation as a sign of synaptic disassembly. In the second screen, we directly quantified the decreasing synapse number using a GFP-tagged presynaptic protein marker. We utilized custom-made image analysis software, which automatically locates synapses and counts their number along individual R8 axons, and identified cirl as a candidate gene responsible for synaptic assembly. Finally, we present a new model of stimulus-dependent synaptic assembly through the interaction of cirl and its possible ligand, ten-a. This study demonstrates the feasibility of using the automated synapse quantification system to explore activity-dependent synaptic plasticity in Drosophila R8 photoreceptors in order to identify molecules involved in stimulus-dependent synaptic assembly.
Collapse
Affiliation(s)
- Jiro Osaka
- School of Life Science and Technology, Tokyo Institute of Technology
| | - Haruka Yasuda
- School of Life Science and Technology, Tokyo Institute of Technology
| | - Yusuke Watanuki
- School of Life Science and Technology, Tokyo Institute of Technology
| | - Yuya Kato
- School of Life Science and Technology, Tokyo Institute of Technology
| | - Yohei Nitta
- Brain Research Institute, Niigata University
| | | | - Makoto Sato
- Mathematical Neuroscience Unit, Institute for Frontier Science Initiative, Kanazawa University.,Graduate School of Frontier Science Initiative, Kanazawa University
| | - Takashi Suzuki
- School of Life Science and Technology, Tokyo Institute of Technology
| |
Collapse
|
19
|
Ahmed M, Rajagopalan AE, Pan Y, Li Y, Williams DL, Pedersen EA, Thakral M, Previero A, Close KC, Christoforou CP, Cai D, Turner GC, Clowney EJ. Hacking brain development to test models of sensory coding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.25.525425. [PMID: 36747712 PMCID: PMC9900841 DOI: 10.1101/2023.01.25.525425] [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: 01/28/2023]
Abstract
Animals can discriminate myriad sensory stimuli but can also generalize from learned experience. You can probably distinguish the favorite teas of your colleagues while still recognizing that all tea pales in comparison to coffee. Tradeoffs between detection, discrimination, and generalization are inherent at every layer of sensory processing. During development, specific quantitative parameters are wired into perceptual circuits and set the playing field on which plasticity mechanisms play out. A primary goal of systems neuroscience is to understand how material properties of a circuit define the logical operations-computations--that it makes, and what good these computations are for survival. A cardinal method in biology-and the mechanism of evolution--is to change a unit or variable within a system and ask how this affects organismal function. Here, we make use of our knowledge of developmental wiring mechanisms to modify hard-wired circuit parameters in the Drosophila melanogaster mushroom body and assess the functional and behavioral consequences. By altering the number of expansion layer neurons (Kenyon cells) and their dendritic complexity, we find that input number, but not cell number, tunes odor selectivity. Simple odor discrimination performance is maintained when Kenyon cell number is reduced and augmented by Kenyon cell expansion.
Collapse
Affiliation(s)
- Maria Ahmed
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Adithya E. Rajagopalan
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yijie Pan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ye Li
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Donnell L. Williams
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Erik A. Pedersen
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Manav Thakral
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Angelica Previero
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kari C. Close
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | | | - Dawen Cai
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48104, USA
- Biophysics LS&A, University of Michigan, Ann Arbor, MI 48109, United States
- Michigan Neuroscience Institute Affiliate
| | - Glenn C. Turner
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA 20147, USA
| | - E. Josephine Clowney
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Michigan Neuroscience Institute Affiliate
| |
Collapse
|
20
|
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
|
21
|
Hogg DW, Reid AL, Dodsworth TL, Chen Y, Reid RM, Xu M, Husic M, Biga PR, Slee A, Buck LT, Barsyte-Lovejoy D, Locke M, Lovejoy DA. Skeletal muscle metabolism and contraction performance regulation by teneurin C-terminal-associated peptide-1. Front Physiol 2022; 13:1031264. [DOI: 10.3389/fphys.2022.1031264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Skeletal muscle regulation is responsible for voluntary muscular movement in vertebrates. The genes of two essential proteins, teneurins and latrophilins (LPHN), evolving in ancestors of multicellular animals form a ligand-receptor pair, and are now shown to be required for skeletal muscle function. Teneurins possess a bioactive peptide, termed the teneurin C-terminal associated peptide (TCAP) that interacts with the LPHNs to regulate skeletal muscle contractility strength and fatigue by an insulin-independent glucose importation mechanism in rats. CRISPR-based knockouts and siRNA-associated knockdowns of LPHN-1 and-3 in the C2C12 mouse skeletal cell line shows that TCAP stimulates an LPHN-dependent cytosolic Ca2+ signal transduction cascade to increase energy metabolism and enhance skeletal muscle function via increases in type-1 oxidative fiber formation and reduce the fatigue response. Thus, the teneurin/TCAP-LPHN system is presented as a novel mechanism that regulates the energy requirements and performance of skeletal muscle.
Collapse
|
22
|
Cheung A, Schachermayer G, Biehler A, Wallis A, Missaire M, Hindges R. Teneurin paralogues are able to localise synaptic sites driven by the intracellular domain and have the potential to form cis-heterodimers. Front Neurosci 2022; 16:915149. [PMID: 36408396 PMCID: PMC9670113 DOI: 10.3389/fnins.2022.915149] [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: 04/07/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Abstract
Synaptic specificity during neurodevelopment is driven by combinatorial interactions between select cell adhesion molecules expressed at the synaptic membrane. These protein-protein interactions are important for instructing the correct connectivity and functionality of the nervous system. Teneurins are one family of synaptic adhesion molecules, highly conserved and widely expressed across interconnected areas during development. These type-II transmembrane glycoproteins are involved in regulating key neurodevelopmental processes during the establishment of neural connectivity. While four teneurin paralogues are found in vertebrates, their subcellular distribution within neurons and interaction between these different paralogues remains largely unexplored. Here we show, through fluorescently tagging teneurin paralogues, that true to their function as synaptic adhesion molecules, all four paralogues are found in a punctate manner and partially localised to synapses when overexpressed in neurons in vitro. Interestingly, each paralogue is differentially distributed across different pre- and post-synaptic sites. In organotypic cultures, Tenm3 is similarly localised to dendritic spines in CA1 neurons, particularly to spine attachment points. Furthermore, we show that the intracellular domain of teneurin plays an important role for synaptic localisation. Finally, while previous studies have shown that the extracellular domain of teneurins allows for active dimer formation and transsynaptic interactions, we find that all paralogues are able to form the full complement of homodimers and cis-heterodimers. This suggests that the combinatorial power to generate distinct molecular teneurin complexes underlying synaptic specificity is even higher than previously thought. The emerging link between teneurin with cancers and neurological disorders only serves to emphasise the importance of further elucidating the molecular mechanisms of teneurin function and their relation to human health and disease.
Collapse
Affiliation(s)
- Angela Cheung
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
| | - Greta Schachermayer
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
| | - Aude Biehler
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
| | - Amber Wallis
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
| | - Mégane Missaire
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
| | - Robert Hindges
- Centre for Developmental Neurobiology, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| |
Collapse
|
23
|
Roberts RJV, Pop S, Prieto-Godino LL. Evolution of central neural circuits: state of the art and perspectives. Nat Rev Neurosci 2022; 23:725-743. [DOI: 10.1038/s41583-022-00644-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 11/09/2022]
|
24
|
Abstract
Single-pass transmembrane receptors (SPTMRs) represent a diverse group of integral membrane proteins that are involved in many essential cellular processes, including signal transduction, cell adhesion, and transmembrane transport of materials. Dysregulation of the SPTMRs is linked with many human diseases. Despite extensive efforts in past decades, the mechanisms of action of the SPTMRs remain incompletely understood. One major hurdle is the lack of structures of the full-length SPTMRs in different functional states. Such structural information is difficult to obtain by traditional structural biology methods such as X-ray crystallography and nuclear magnetic resonance (NMR). The recent rapid development of single-particle cryo-electron microscopy (cryo-EM) has led to an exponential surge in the number of high-resolution structures of integral membrane proteins, including SPTMRs. Cryo-EM structures of SPTMRs solved in the past few years have tremendously improved our understanding of how SPTMRs function. In this review, we will highlight these progresses in the structural studies of SPTMRs by single-particle cryo-EM, analyze important structural details of each protein involved, and discuss their implications on the underlying mechanisms. Finally, we also briefly discuss remaining challenges and exciting opportunities in the field.
Collapse
Affiliation(s)
- Kai Cai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
| | - Xuewu Zhang
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xuewu Zhang, Department of pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Xiao-chen Bai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xiao-chen Bai, Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| |
Collapse
|
25
|
Duhart JC, Mosca TJ. Genetic regulation of central synapse formation and organization in Drosophila melanogaster. Genetics 2022; 221:6597078. [PMID: 35652253 DOI: 10.1093/genetics/iyac078] [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: 03/14/2022] [Accepted: 04/29/2022] [Indexed: 01/04/2023] Open
Abstract
A goal of modern neuroscience involves understanding how connections in the brain form and function. Such a knowledge is essential to inform how defects in the exquisite complexity of nervous system growth influence neurological disease. Studies of the nervous system in the fruit fly Drosophila melanogaster enabled the discovery of a wealth of molecular and genetic mechanisms underlying development of synapses-the specialized cell-to-cell connections that comprise the essential substrate for information flow and processing in the nervous system. For years, the major driver of knowledge was the neuromuscular junction due to its ease of examination. Analogous studies in the central nervous system lagged due to a lack of genetic accessibility of specific neuron classes, synaptic labels compatible with cell-type-specific access, and high resolution, quantitative imaging strategies. However, understanding how central synapses form remains a prerequisite to understanding brain development. In the last decade, a host of new tools and techniques extended genetic studies of synapse organization into central circuits to enhance our understanding of synapse formation, organization, and maturation. In this review, we consider the current state-of-the-field. We first discuss the tools, technologies, and strategies developed to visualize and quantify synapses in vivo in genetically identifiable neurons of the Drosophila central nervous system. Second, we explore how these tools enabled a clearer understanding of synaptic development and organization in the fly brain and the underlying molecular mechanisms of synapse formation. These studies establish the fly as a powerful in vivo genetic model that offers novel insights into neural development.
Collapse
Affiliation(s)
- Juan Carlos Duhart
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Timothy J Mosca
- Department of Neuroscience, Vickie and Jack Farber Institute of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
26
|
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: 8] [Impact Index Per Article: 2.7] [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
|
27
|
Moreland T, Poulain FE. To Stick or Not to Stick: The Multiple Roles of Cell Adhesion Molecules in Neural Circuit Assembly. Front Neurosci 2022; 16:889155. [PMID: 35573298 PMCID: PMC9096351 DOI: 10.3389/fnins.2022.889155] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/28/2022] [Indexed: 01/02/2023] Open
Abstract
Precise wiring of neural circuits is essential for brain connectivity and function. During development, axons respond to diverse cues present in the extracellular matrix or at the surface of other cells to navigate to specific targets, where they establish precise connections with post-synaptic partners. Cell adhesion molecules (CAMs) represent a large group of structurally diverse proteins well known to mediate adhesion for neural circuit assembly. Through their adhesive properties, CAMs act as major regulators of axon navigation, fasciculation, and synapse formation. While the adhesive functions of CAMs have been known for decades, more recent studies have unraveled essential, non-adhesive functions as well. CAMs notably act as guidance cues and modulate guidance signaling pathways for axon pathfinding, initiate contact-mediated repulsion for spatial organization of axonal arbors, and refine neuronal projections during circuit maturation. In this review, we summarize the classical adhesive functions of CAMs in axonal development and further discuss the increasing number of other non-adhesive functions CAMs play in neural circuit assembly.
Collapse
|
28
|
Zhang X, Lin PY, Liakath-Ali K, Südhof TC. Teneurins assemble into presynaptic nanoclusters that promote synapse formation via postsynaptic non-teneurin ligands. Nat Commun 2022; 13:2297. [PMID: 35484136 PMCID: PMC9050732 DOI: 10.1038/s41467-022-29751-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
Extensive studies concluded that homophilic interactions between pre- and postsynaptic teneurins, evolutionarily conserved cell-adhesion molecules, encode the specificity of synaptic connections. However, no direct evidence is available to demonstrate that teneurins are actually required on both pre- and postsynaptic neurons for establishing synaptic connections, nor is it known whether teneurins are localized to synapses. Using super-resolution microscopy, we demonstrate that Teneurin-3 assembles into presynaptic nanoclusters of approximately 80 nm in most excitatory synapses of the hippocampus. Presynaptic deletions of Teneurin-3 and Teneurin-4 in the medial entorhinal cortex revealed that they are required for assembly of entorhinal cortex-CA1, entorhinal cortex-subiculum, and entorhinal cortex-dentate gyrus synapses. Postsynaptic deletions of teneurins in the CA1 region, however, had no effect on synaptic connections from any presynaptic input. Our data suggest that different from the current prevailing view, teneurins promote the establishment of synaptic connections exclusively as presynaptic cell-adhesion molecules, most likely via their nanomolar-affinity binding to postsynaptic latrophilins.
Collapse
Affiliation(s)
- Xuchen Zhang
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA. .,Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA.
| | - Pei-Yi Lin
- grid.168010.e0000000419368956Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA USA
| | - Kif Liakath-Ali
- grid.168010.e0000000419368956Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA USA
| | - Thomas C. Südhof
- grid.168010.e0000000419368956Howard Hughes Medical Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA USA
| |
Collapse
|
29
|
Kamemura K, Moriya H, Ukita Y, Okumura M, Miura M, Chihara T. Endoplasmic reticulum proteins Meigo and Gp93 govern dendrite targeting by regulating Toll-6 localization. Dev Biol 2022; 484:30-39. [DOI: 10.1016/j.ydbio.2022.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/29/2021] [Accepted: 02/02/2022] [Indexed: 12/15/2022]
|
30
|
Meijer DH, Frias CP, Beugelink JW, Deurloo YN, Janssen BJC. Teneurin4 dimer structures reveal a calcium‐stabilized compact conformation supporting homomeric trans‐interactions. EMBO J 2022; 41:e107505. [PMID: 35099835 PMCID: PMC9058538 DOI: 10.15252/embj.2020107505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/18/2022] Open
Abstract
Establishment of correct synaptic connections is a crucial step during neural circuitry formation. The Teneurin family of neuronal transmembrane proteins promotes cell–cell adhesion via homophilic and heterophilic interactions, and is required for synaptic partner matching in the visual and hippocampal systems in vertebrates. It remains unclear how individual Teneurins form macromolecular cis‐ and trans‐synaptic protein complexes. Here, we present a 2.7 Å cryo‐EM structure of the dimeric ectodomain of human Teneurin4. The structure reveals a compact conformation of the dimer, stabilized by interactions mediated by the C‐rich, YD‐shell, and ABD domains. A 1.5 Å crystal structure of the C‐rich domain shows three conserved calcium binding sites, and thermal unfolding assays and SAXS‐based rigid‐body modeling demonstrate that the compactness and stability of Teneurin4 dimers are calcium‐dependent. Teneurin4 dimers form a more extended conformation in conditions that lack calcium. Cellular assays reveal that the compact cis‐dimer is compatible with homomeric trans‐interactions. Together, these findings support a role for teneurins as a scaffold for macromolecular complex assembly and the establishment of cis‐ and trans‐synaptic interactions to construct functional neuronal circuits.
Collapse
Affiliation(s)
- Dimphna H Meijer
- Department of Bionanoscience Kavli Institute of Nanoscience Delft University of Technology Delft The Netherlands
- Department of Chemistry Faculty of Science Structural Biochemistry Bijvoet Center for Biomolecular Research Utrecht University Utrecht The Netherlands
| | - Cátia P Frias
- Department of Bionanoscience Kavli Institute of Nanoscience Delft University of Technology Delft The Netherlands
| | - J Wouter Beugelink
- Department of Chemistry Faculty of Science Structural Biochemistry Bijvoet Center for Biomolecular Research Utrecht University Utrecht The Netherlands
| | - Yanthi N Deurloo
- Department of Bionanoscience Kavli Institute of Nanoscience Delft University of Technology Delft The Netherlands
| | - Bert J C Janssen
- Department of Chemistry Faculty of Science Structural Biochemistry Bijvoet Center for Biomolecular Research Utrecht University Utrecht The Netherlands
| |
Collapse
|
31
|
Derdeyn P, Hui M, Macchia D, Beier KT. Uncovering the Connectivity Logic of the Ventral Tegmental Area. Front Neural Circuits 2022; 15:799688. [PMID: 35153681 PMCID: PMC8832514 DOI: 10.3389/fncir.2021.799688] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Decades of research have revealed the remarkable complexity of the midbrain dopamine (DA) system, which comprises cells principally located in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc). Neither homogenous nor serving a singular function, the midbrain DA system is instead composed of distinct cell populations that (1) receive different sets of inputs, (2) project to separate forebrain sites, and (3) are characterized by unique transcriptional and physiological signatures. To appreciate how these differences relate to circuit function, we first need to understand the anatomical connectivity of unique DA pathways and how this connectivity relates to DA-dependent motivated behavior. We and others have provided detailed maps of the input-output relationships of several subpopulations of midbrain DA cells and explored the roles of these different cell populations in directing behavioral output. In this study, we analyze VTA inputs and outputs as a high dimensional dataset (10 outputs, 22 inputs), deploying computational techniques well-suited to finding interpretable patterns in such data. In addition to reinforcing our previous conclusion that the connectivity in the VTA is dependent on spatial organization, our analysis also uncovered a set of inputs elevated onto each projection-defined VTADA cell type. For example, VTADA→NAcLat cells receive preferential innervation from inputs in the basal ganglia, while VTADA→Amygdala cells preferentially receive inputs from populations sending a distributed input across the VTA, which happen to be regions associated with the brain's stress circuitry. In addition, VTADA→NAcMed cells receive ventromedially biased inputs including from the preoptic area, ventral pallidum, and laterodorsal tegmentum, while VTADA→mPFC cells are defined by dominant inputs from the habenula and dorsal raphe. We also go on to show that the biased input logic to the VTADA cells can be recapitulated using projection architecture in the ventral midbrain, reinforcing our finding that most input differences identified using rabies-based (RABV) circuit mapping reflect projection archetypes within the VTA.
Collapse
Affiliation(s)
- Pieter Derdeyn
- Program in Mathematical, Computational, and Systems Biology, University of California, Irvine, Irvine, CA, United States
| | - May Hui
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Desiree Macchia
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Kevin T. Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
32
|
Günther P, Quentin D, Ahmad S, Sachar K, Gatsogiannis C, Whitney JC, Raunser S. Structure of a bacterial Rhs effector exported by the type VI secretion system. PLoS Pathog 2022; 18:e1010182. [PMID: 34986192 PMCID: PMC8765631 DOI: 10.1371/journal.ppat.1010182] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/18/2022] [Accepted: 12/08/2021] [Indexed: 11/20/2022] Open
Abstract
The type VI secretion system (T6SS) is a widespread protein export apparatus found in Gram-negative bacteria. The majority of T6SSs deliver toxic effector proteins into competitor bacteria. Yet, the structure, function, and activation of many of these effectors remains poorly understood. Here, we present the structures of the T6SS effector RhsA from Pseudomonas protegens and its cognate T6SS spike protein, VgrG1, at 3.3 Å resolution. The structures reveal that the rearrangement hotspot (Rhs) repeats of RhsA assemble into a closed anticlockwise β-barrel spiral similar to that found in bacterial insecticidal Tc toxins and in metazoan teneurin proteins. We find that the C-terminal toxin domain of RhsA is autoproteolytically cleaved but remains inside the Rhs ‘cocoon’ where, with the exception of three ordered structural elements, most of the toxin is disordered. The N-terminal ‘plug’ domain is unique to T6SS Rhs proteins and resembles a champagne cork that seals the Rhs cocoon at one end while also mediating interactions with VgrG1. Interestingly, this domain is also autoproteolytically cleaved inside the cocoon but remains associated with it. We propose that mechanical force is required to remove the cleaved part of the plug, resulting in the release of the toxin domain as it is delivered into a susceptible bacterial cell by the T6SS. Bacteria have developed a variety of strategies to compete for nutrients and limited resources. One system widely used by Gram-negative bacteria is the T6 secretion system which delivers a plethora of effectors into competing bacterial cells. Known functions of effectors are degradation of the cell wall, the depletion of essential metabolites such as NAD+ or the cleavage of DNA. RhsA is an effector from the widespread plant-protecting bacteria Pseudomonas protegens. We found that RhsA forms a closed cocoon similar to that found in bacterial Tc toxins and metazoan teneurin proteins. The effector cleaves its polypeptide chain by itself in three pieces, namely the N-terminal domain including a seal, the cocoon and the actual toxic component which potentially cleaves DNA. The toxic component is encapsulated in the large cocoon, so that the effector producing bacterium is protected from the toxin. In order for the toxin to exit the cocoon, we propose that the seal, which closes the cocoon at one end, is removed by mechanical forces during injection of the effector by the T6 secretion system. We further hypothesize about different scenarios for the delivery of the toxin into the cytoplasm of the host cell. Together, our findings expand the knowledge of the mechanism of action of the T6 secretion system and its essential role in interbacterial competition.
Collapse
Affiliation(s)
- Patrick Günther
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Dennis Quentin
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Shehryar Ahmad
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Kartik Sachar
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Christos Gatsogiannis
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - John C. Whitney
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Canada
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, Canada
- * E-mail: (J.C.W.); (S.R.)
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
- * E-mail: (J.C.W.); (S.R.)
| |
Collapse
|
33
|
Spatiotemporal expression of IgLON family members in the developing mouse nervous system. Sci Rep 2021; 11:19536. [PMID: 34599206 PMCID: PMC8486791 DOI: 10.1038/s41598-021-97768-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023] Open
Abstract
Differential expression of cell adhesion molecules in neuronal populations is one of the many mechanisms promoting the formation of functional neural circuits in the developing nervous system. The IgLON family consists of five cell surface immunoglobulin proteins that have been associated with various developmental disorders, such as autism spectrum disorder, schizophrenia, and major depressive disorder. However, there is still limited and fragmented information about their patterns of expression in certain regions of the developing nervous system and how their expression contributes to their function. Utilizing an in situ hybridization approach, we have analyzed the spatiotemporal expression of all IgLON family members in the developing mouse brain, spinal cord, eye, olfactory epithelium, and vomeronasal organ. At one prenatal (E16) and two postnatal (P0 and P15) ages, we show that each IgLON displays distinct expression patterns in the olfactory system, cerebral cortex, midbrain, cerebellum, spinal cord, and eye, indicating that they likely contribute to the wiring of specific neuronal circuitry. These analyses will inform future functional studies aimed at identifying additional roles for these proteins in nervous system development.
Collapse
|
34
|
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
|
35
|
Yi X, Li M, He G, Du H, Li X, Cao D, Wang L, Wu X, Yang F, Chen X, He L, Ping Y, Zhou D. Genetic and functional analysis reveals TENM4 contributes to schizophrenia. iScience 2021; 24:103063. [PMID: 34568788 PMCID: PMC8449235 DOI: 10.1016/j.isci.2021.103063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/23/2021] [Accepted: 08/26/2021] [Indexed: 12/09/2022] Open
Abstract
TENM4, encoding a member of the teneurin protein family, is a risk gene shared by many types of mental diseases and is implicated in neuronal plasticity and signaling. However, the role and the mechanisms of TENM4 in schizophrenia (SCZ) remain unclear. We identified possible pathogenic mutations in the TENM4 gene through target sequencing of TENM4 in 68 SCZ families. We further demonstrated that aberrant expression of Ten-m leads to lower learning ability, sleep reduction, and increased aggressiveness in animal models. RNA sequencing showed that aberrant expression of Ten-m was related to stimulus perception and metabolic process, and Gene Ontology enrichment terms were neurogenesis and ATPase activity. This study provides strong evidence that TENM4 contributes to SCZ, and its functional mutations might be responsible for the impaired neural circuits and behaviors observed in SCZ. Possible pathogenic rare missense mutations in TENM4 gene contribute to SCZ Aberrant expression of Ten-m leads to behavioral disturbances related to SCZ symptoms Ten-m affects stimulation, metabolic process, neurogenesis, and ATPase activity
Collapse
Affiliation(s)
- Xin Yi
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Minzhe Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Guang He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huihui Du
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Xingwang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongmei Cao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Lu Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Xi Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Fengping Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| | - Xu Chen
- Department of Neurology, Shanghai Eighth People's Hospital, Shanghai Sixth People's Hospital Xuhui Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Ping
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Daizhan Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, 1954 Huashan Rd., Shanghai 200030, PR China
| |
Collapse
|
36
|
Abstract
Daniel Pederick and Liqun Luo introduce teneurins, evolutionarily conserved type II transmembrane proteins with functions in the nervous system.
Collapse
Affiliation(s)
- Daniel T Pederick
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | - Liqun Luo
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
37
|
Zhang S, Saunders T. Mechanical processes underlying precise and robust cell matching. Semin Cell Dev Biol 2021; 120:75-84. [PMID: 34130903 DOI: 10.1016/j.semcdb.2021.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/27/2021] [Accepted: 06/04/2021] [Indexed: 11/26/2022]
Abstract
During the development of complicated multicellular organisms, the robust formation of specific cell-cell connections (cell matching) is required for the generation of precise tissue structures. Mismatches or misconnections can lead to various diseases. Diverse mechanical cues, including differential adhesion and temporally varying cell contractility, are involved in regulating the process of cell-cell recognition and contact formation. Cells often start the process of cell matching through contact via filopodia protrusions, mediated by specific adhesion interactions at the cell surface. These adhesion interactions give rise to differential mechanical signals that can be further perceived by the cells. In conjunction with contractions generated by the actomyosin networks within the cells, this differentially coded adhesion information can be translated to reposition and sort cells. Here, we review the role of these different cell matching components and suggest how these mechanical factors cooperate with each other to facilitate specificity in cell-cell contact formation.
Collapse
Affiliation(s)
- Shaobo Zhang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Timothy Saunders
- Mechanobiology Institute, National University of Singapore, Singapore; Department of Biological Sciences, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Proteos, Singapore; Warwick Medical School, University of Warwick, Coventry, United Kingdom.
| |
Collapse
|
38
|
Cell-type-specific, multicolor labeling of endogenous proteins with split fluorescent protein tags in Drosophila. Proc Natl Acad Sci U S A 2021; 118:2024690118. [PMID: 34074768 DOI: 10.1073/pnas.2024690118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The impact of the Drosophila experimental system on studies of modern biology cannot be understated. The ability to tag endogenously expressed proteins is essential to maximize the use of this model organism. Here, we describe a method for labeling endogenous proteins with self-complementing split fluorescent proteins (split FPs) in a cell-type-specific manner in Drosophila A short fragment of an FP coding sequence is inserted into a specific genomic locus while the remainder of the FP is expressed using an available GAL4 driver line. In consequence, complementation fluorescence allows examination of protein localization in particular cells. Besides, when inserting tandem repeats of the short FP fragment at the same genomic locus, we can substantially enhance the fluorescence signal. The enhanced signal is of great value in live-cell imaging at the subcellular level. We can also accomplish a multicolor labeling system with orthogonal split FPs. However, other orthogonal split FPs do not function for in vivo imaging besides split GFP. Through protein engineering and in vivo functional studies, we report a red split FP that we can use for duplexed visualization of endogenous proteins in intricate Drosophila tissues. Using the two orthogonal split FP systems, we have simultaneously imaged proteins that reside in distinct subsynaptic compartments. Our approach allows us to study the proximity between and localization of multiple proteins endogenously expressed in essentially any cell type in Drosophila.
Collapse
|
39
|
Regan SL, Williams MT, Vorhees CV. Latrophilin-3 disruption: Effects on brain and behavior. Neurosci Biobehav Rev 2021; 127:619-629. [PMID: 34022279 DOI: 10.1016/j.neubiorev.2021.04.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/22/2022]
Abstract
Latrophilin-3 (LPHN3), a G-protein-coupled receptor belonging to the adhesion subfamily, is a regulator of synaptic function and maintenance in brain regions that mediate locomotor activity, attention, and memory for location and path. Variants of LPHN3 are associated with increased risk for attention deficit hyperactivity disorder (ADHD) in some patients. Here we review the role of LPHN3 in the central nervous system (CNS). We describe synaptic localization of LPHN3, its trans-synaptic binding partners, links to neurodevelopmental disorders, animal models of Lphn3 disruption in different species, and evidence that LPHN3 is involved in cognition as well as activity and attention. The evidence shows that LPHN3 plays a more significant role in neuroplasticity than previously appreciated.
Collapse
Affiliation(s)
- Samantha L Regan
- Neuroscience Graduate Program, University of Cincinnati, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Michael T Williams
- Neuroscience Graduate Program, University of Cincinnati, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Charles V Vorhees
- Neuroscience Graduate Program, University of Cincinnati, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA.
| |
Collapse
|
40
|
Douthit J, Hairston A, Lee G, Morrison CA, Holguera I, Treisman JE. R7 photoreceptor axon targeting depends on the relative levels of lost and found expression in R7 and its synaptic partners. eLife 2021; 10:65895. [PMID: 34003117 PMCID: PMC8205486 DOI: 10.7554/elife.65895] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/17/2021] [Indexed: 01/17/2023] Open
Abstract
As neural circuits form, growing processes select the correct synaptic partners through interactions between cell surface proteins. The presence of such proteins on two neuronal processes may lead to either adhesion or repulsion; however, the consequences of mismatched expression have rarely been explored. Here, we show that the Drosophila CUB-LDL protein Lost and found (Loaf) is required in the UV-sensitive R7 photoreceptor for normal axon targeting only when Loaf is also present in its synaptic partners. Although targeting occurs normally in loaf mutant animals, removing loaf from photoreceptors or expressing it in their postsynaptic neurons Tm5a/b or Dm9 in a loaf mutant causes mistargeting of R7 axons. Loaf localizes primarily to intracellular vesicles including endosomes. We propose that Loaf regulates the trafficking or function of one or more cell surface proteins, and an excess of these proteins on the synaptic partners of R7 prevents the formation of stable connections.
Collapse
Affiliation(s)
- Jessica Douthit
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, United States
| | - Ariel Hairston
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, United States
| | - Gina Lee
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, United States
| | - Carolyn A Morrison
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, United States
| | - Isabel Holguera
- Department of Biology, New York University, New York, United States
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, United States
| |
Collapse
|
41
|
Everetts NJ, Worley MI, Yasutomi R, Yosef N, Hariharan IK. Single-cell transcriptomics of the Drosophila wing disc reveals instructive epithelium-to-myoblast interactions. eLife 2021; 10:61276. [PMID: 33749594 PMCID: PMC8021398 DOI: 10.7554/elife.61276] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/21/2021] [Indexed: 12/20/2022] Open
Abstract
In both vertebrates and invertebrates, generating a functional appendage requires interactions between ectoderm-derived epithelia and mesoderm-derived cells. To investigate such interactions, we used single-cell transcriptomics to generate a temporal cell atlas of the Drosophila wing disc from two developmental time points. Using these data, we visualized gene expression using a multilayered model of the wing disc and cataloged ligand–receptor pairs that could mediate signaling between epithelial cells and adult muscle precursors (AMPs). We found that localized expression of the fibroblast growth factor ligands, Thisbe and Pyramus, in the disc epithelium regulates the number and location of the AMPs. In addition, Hedgehog ligand from the epithelium activates a specific transcriptional program within adjacent AMP cells, defined by AMP-specific targets Neurotactin and midline, that is critical for proper formation of direct flight muscles. More generally, our annotated temporal cell atlas provides an organ-wide view of potential cell–cell interactions between epithelial and myogenic cells.
Collapse
Affiliation(s)
- Nicholas J Everetts
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Department of Electrical Engineering & Computer Science, Center for Computational Biology, UC Berkeley, University of California, Berkeley, Berkeley, United States
| | - Melanie I Worley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Riku Yasutomi
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Nir Yosef
- Department of Electrical Engineering & Computer Science, Center for Computational Biology, UC Berkeley, University of California, Berkeley, Berkeley, United States
| | - Iswar K Hariharan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
42
|
Teneurins: Role in Cancer and Potential Role as Diagnostic Biomarkers and Targets for Therapy. Int J Mol Sci 2021; 22:ijms22052321. [PMID: 33652578 PMCID: PMC7956758 DOI: 10.3390/ijms22052321] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Teneurins have been identified in vertebrates as four different genes (TENM1-4), coding for membrane proteins that are mainly involved in embryonic and neuronal development. Genetic studies have correlated them with various diseases, including developmental problems, neurological disorders and congenital general anosmia. There is some evidence to suggest their possible involvement in cancer initiation and progression, and drug resistance. Indeed, mutations, chromosomal alterations and the deregulation of teneurins expression have been associated with several tumor types and patient survival. However, the role of teneurins in cancer-related regulatory networks is not fully understood, as both a tumor-suppressor role and pro-tumoral functions have been proposed, depending on tumor histotype. Here, we summarize and discuss the literature data on teneurins expression and their potential role in different tumor types, while highlighting the possibility of using teneurins as novel molecular diagnostic and prognostic biomarkers and as targets for cancer treatments, such as immunotherapy, in some tumors.
Collapse
|
43
|
Heckman EL, Doe CQ. Establishment and Maintenance of Neural Circuit Architecture. J Neurosci 2021; 41:1119-1129. [PMID: 33568445 PMCID: PMC7888231 DOI: 10.1523/jneurosci.1143-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/29/2020] [Accepted: 12/09/2020] [Indexed: 02/03/2023] Open
Abstract
The ability to sense the world, process information, and navigate the environment depends on the assembly and continuous function of neural circuits in the brain. Within the past two decades, new technologies have rapidly advanced our understanding of how neural circuits are wired during development and how they are stably maintained, often for years. Electron microscopy reconstructions of model organism connectomes have provided a map of the stereotyped (and variable) connections in the brain; advanced light microscopy techniques have enabled direct observation of the cellular dynamics that underlie circuit construction and maintenance; transcriptomic and proteomic surveys of both developing and mature neurons have provided insights into the molecular and genetic programs governing circuit establishment and maintenance; and advanced genetic techniques have allowed for high-throughput discovery of wiring regulators. These tools have empowered scientists to rapidly generate and test hypotheses about how circuits establish and maintain connectivity. Thus, the set of principles governing circuit formation and maintenance have been expanded. These principles are discussed in this review.
Collapse
Affiliation(s)
- Emily L Heckman
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, Oregon 97403
| | - Chris Q Doe
- Institute of Neuroscience, Institute of Molecular Biology, Howard Hughes Medical Institute, University of Oregon, Eugene, Oregon 97403
| |
Collapse
|
44
|
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: 48] [Impact Index Per Article: 12.0] [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
|
45
|
Lin TY, Chen PJ, Yu HH, Hsu CP, Lee CH. Extrinsic Factors Regulating Dendritic Patterning. Front Cell Neurosci 2021; 14:622808. [PMID: 33519386 PMCID: PMC7838386 DOI: 10.3389/fncel.2020.622808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Stereotypic dendrite arborizations are key morphological features of neuronal identity, as the size, shape and location of dendritic trees determine the synaptic input fields and how information is integrated within developed neural circuits. In this review, we focus on the actions of extrinsic intercellular communication factors and their effects on intrinsic developmental processes that lead to dendrite patterning. Surrounding neurons or supporting cells express adhesion receptors and secreted proteins that respectively, act via direct contact or over short distances to shape, size, and localize dendrites during specific developmental stages. The different ligand-receptor interactions and downstream signaling events appear to direct dendrite morphogenesis by converging on two categorical mechanisms: local cytoskeletal and adhesion modulation and global transcriptional regulation of key dendritic growth components, such as lipid synthesis enzymes. Recent work has begun to uncover how the coordinated signaling of multiple extrinsic factors promotes complexity in dendritic trees and ensures robust dendritic patterning.
Collapse
Affiliation(s)
- Tzu-Yang Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Pei-Ju Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Hsiang Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chao-Ping Hsu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chi-Hon Lee
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
46
|
Xie Q, Brbic M, Horns F, Kolluru SS, Jones RC, Li J, Reddy AR, Xie A, Kohani S, Li Z, McLaughlin CN, Li T, Xu C, Vacek D, Luginbuhl DJ, Leskovec J, Quake SR, Luo L, Li H. Temporal evolution of single-cell transcriptomes of Drosophila olfactory projection neurons. eLife 2021; 10:e63450. [PMID: 33427646 PMCID: PMC7870145 DOI: 10.7554/elife.63450] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/05/2021] [Indexed: 12/18/2022] Open
Abstract
Neurons undergo substantial morphological and functional changes during development to form precise synaptic connections and acquire specific physiological properties. What are the underlying transcriptomic bases? Here, we obtained the single-cell transcriptomes of Drosophila olfactory projection neurons (PNs) at four developmental stages. We decoded the identity of 21 transcriptomic clusters corresponding to 20 PN types and developed methods to match transcriptomic clusters representing the same PN type across development. We discovered that PN transcriptomes reflect unique biological processes unfolding at each stage-neurite growth and pruning during metamorphosis at an early pupal stage; peaked transcriptomic diversity during olfactory circuit assembly at mid-pupal stages; and neuronal signaling in adults. At early developmental stages, PN types with adjacent birth order share similar transcriptomes. Together, our work reveals principles of cellular diversity during brain development and provides a resource for future studies of neural development in PNs and other neuronal types.
Collapse
Affiliation(s)
- Qijing Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
- Neurosciences Graduate Program, Stanford UniversityStanfordUnited States
| | - Maria Brbic
- Department of Computer Science, Stanford UniversityStanfordUnited States
| | - Felix Horns
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Biophysics Graduate Program, Stanford UniversityStanfordUnited States
| | | | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Jiefu Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Anay R Reddy
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Anthony Xie
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Sayeh Kohani
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Zhuoran Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Colleen N McLaughlin
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Tongchao Li
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Chuanyun Xu
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - David Vacek
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - David J Luginbuhl
- Department of Biology, Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Jure Leskovec
- Department of Computer Science, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubStanfordUnited 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
|
47
|
Hiesinger PR. Brain wiring with composite instructions. Bioessays 2020; 43:e2000166. [PMID: 33145823 DOI: 10.1002/bies.202000166] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 11/12/2022]
Abstract
The quest for molecular mechanisms that guide axons or specify synaptic contacts has largely focused on molecules that intuitively relate to the idea of an "instruction." By contrast, "permissive" factors are traditionally considered background machinery without contribution to the information content of a molecularly executed instruction. In this essay, I recast this dichotomy as a continuum from permissive to instructive actions of single factors that provide relative contributions to a necessarily collaborative effort. Individual molecules or other factors do not constitute absolute instructions by themselves; they provide necessary context for each other, thereby creating a composite that defines the overall instruction. The idea of composite instructions leads to two main conclusions: first, a composite of many seemingly permissive factors can define a specific instruction even in the absence of a single dominant contributor; second, individual factors are not necessarily related intuitively to the overall instruction or phenotypic outcome.
Collapse
Affiliation(s)
- P Robin Hiesinger
- Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
48
|
Valdes-Aleman J, Fetter RD, Sales EC, Heckman EL, Venkatasubramanian L, Doe CQ, Landgraf M, Cardona A, Zlatic M. Comparative Connectomics Reveals How Partner Identity, Location, and Activity Specify Synaptic Connectivity in Drosophila. Neuron 2020; 109:105-122.e7. [PMID: 33120017 PMCID: PMC7837116 DOI: 10.1016/j.neuron.2020.10.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 08/12/2020] [Accepted: 10/05/2020] [Indexed: 01/30/2023]
Abstract
The mechanisms by which synaptic partners recognize each other and establish appropriate numbers of connections during embryonic development to form functional neural circuits are poorly understood. We combined electron microscopy reconstruction, functional imaging of neural activity, and behavioral experiments to elucidate the roles of (1) partner identity, (2) location, and (3) activity in circuit assembly in the embryonic nerve cord of Drosophila. We found that postsynaptic partners are able to find and connect to their presynaptic partners even when these have been shifted to ectopic locations or silenced. However, orderly positioning of axon terminals by positional cues and synaptic activity is required for appropriate numbers of connections between specific partners, for appropriate balance between excitatory and inhibitory connections, and for appropriate functional connectivity and behavior. Our study reveals with unprecedented resolution the fine connectivity effects of multiple factors that work together to control the assembly of neural circuits.
Collapse
Affiliation(s)
- Javier Valdes-Aleman
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA; Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Richard D Fetter
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Emily C Sales
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Emily L Heckman
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | | | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Matthias Landgraf
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Albert Cardona
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA; Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK; MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Marta Zlatic
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA; Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK; MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
49
|
Timaeus L, Geid L, Sancer G, Wernet MF, Hummel T. Parallel Visual Pathways with Topographic versus Nontopographic Organization Connect the Drosophila Eyes to the Central Brain. iScience 2020; 23:101590. [PMID: 33205011 PMCID: PMC7648135 DOI: 10.1016/j.isci.2020.101590] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/21/2020] [Accepted: 09/16/2020] [Indexed: 11/12/2022] Open
Abstract
One hallmark of the visual system is a strict retinotopic organization from the periphery toward the central brain, where functional imaging in Drosophila revealed a spatially accurate representation of visual cues in the central complex. This raised the question how, on a circuit level, the topographic features are implemented, as the majority of visual neurons enter the central brain converge in optic glomeruli. We discovered a spatial segregation of topographic versus nontopographic projections of distinct classes of medullo-tubercular (MeTu) neurons into a specific visual glomerulus, the anterior optic tubercle (AOTU). These parallel channels synapse onto different tubercular-bulbar (TuBu) neurons, which in turn relay visual information onto specific central complex ring neurons in the bulb neuropil. Hence, our results provide the circuit basis for spatially accurate representation of visual information and highlight the AOTU's role as a prominent relay station for spatial information from the retina to the central brain. A Drosophila visual circuit conveys input from the periphery to the central brain Several synaptic pathways form parallel channels using the anterior optic tubercle Some pathways maintain topographic relationships across several synaptic steps Different target neurons in the central brain are identified
Collapse
Affiliation(s)
- Lorin Timaeus
- Department of Neurobiology, University of Vienna, Vienna, Austria
| | - Laura Geid
- Department of Neurobiology, University of Vienna, Vienna, Austria.,Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Gizem Sancer
- Department of Biology, Freie Universität Berlin, Berlin, Germany
| | - Mathias F Wernet
- Department of Biology, Freie Universität Berlin, Berlin, Germany
| | - Thomas Hummel
- Department of Neurobiology, University of Vienna, Vienna, Austria
| |
Collapse
|
50
|
Ryba AR, McKenzie SK, Olivos-Cisneros L, Clowney EJ, Pires PM, Kronauer DJC. Comparative Development of the Ant Chemosensory System. Curr Biol 2020; 30:3223-3230.e4. [PMID: 32559450 DOI: 10.1016/j.cub.2020.05.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/24/2020] [Accepted: 05/21/2020] [Indexed: 12/15/2022]
Abstract
The insect antennal lobe (AL) contains the first synapses of the olfactory system, where olfactory sensory neurons (OSNs) contact second-order projection neurons (PNs). In Drosophila melanogaster, OSNs expressing specific receptor genes send stereotyped projections to one or two of about 50 morphologically defined glomeruli [1-3]. The mechanisms for this precise matching between OSNs and PNs have been studied extensively in D. melanogaster, where development is deterministic and independent of neural activity [4-6]. However, a number of insect lineages, most notably the ants, have receptor gene repertoires many times larger than D. melanogaster and exhibit more structurally complex antennal lobes [7-12]. Moreover, perturbation of OSN function via knockout of the odorant receptor (OR) co-receptor, Orco, results in drastic AL reductions in ants [13, 14], but not in Drosophila [15]. Here, we characterize AL development in the clonal raider ant, Ooceraea biroi. We find that, unlike in Drosophila, ORs and Orco are expressed before the onset of glomerulus formation, and Orco protein is trafficked to developing axon terminals, raising the possibility that ORs play a role during ant AL development. Additionally, ablating ant antennae at the onset of pupation results in AL defects that recapitulate the Orco mutant phenotype. Thus, early loss of functional OSN innervation reveals latent structure in the AL that develops independently of peripheral input, suggesting that the AL is initially pre-patterned and later refined in an OSN-dependent manner. This two-step process might increase developmental flexibility and thereby facilitate the rapid evolution and expansion of the ant chemosensory system.
Collapse
Affiliation(s)
- Anna R Ryba
- Laboratory of Social Evolution and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Sean K McKenzie
- Laboratory of Social Evolution and Behavior, The Rockefeller University, New York, NY 10065, USA; Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Leonora Olivos-Cisneros
- Laboratory of Social Evolution and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - E Josephine Clowney
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter Mussells Pires
- Laboratory of Social Evolution and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Daniel J C Kronauer
- Laboratory of Social Evolution and Behavior, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|