1
|
Tann JY, Xu F, Kimura M, Wilkes OR, Yoong LF, Skibbe H, Moore AW. Study of Dendrite Differentiation Using Drosophila Dendritic Arborization Neurons. Cold Spring Harb Protoc 2024; 2024:pdb.top108146. [PMID: 38148165 DOI: 10.1101/pdb.top108146] [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: 12/28/2023]
Abstract
Neurons receive, process, and integrate inputs. These operations are organized by dendrite arbor morphology, and the dendritic arborization (da) neurons of the Drosophila peripheral sensory nervous system are an excellent experimental model for examining the differentiation processes that build and shape the dendrite arbor. Studies in da neurons are enabled by a wealth of fly genetic tools that allow targeted neuron manipulation and labeling of the neuron's cytoskeletal or organellar components. Moreover, as da neuron dendrite arbors cover the body wall, they are highly accessible for live imaging analysis of arbor patterning. Here, we outline the structure and function of different da neuron types and give examples of how they are used to elucidate central mechanisms of dendritic arbor formation.
Collapse
Affiliation(s)
- Jason Y Tann
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Fangke Xu
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Minami Kimura
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Oliver R Wilkes
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
- Department of Cellular and Molecular Biology, Institute for Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Li-Foong Yoong
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Henrik Skibbe
- Brain Image Analysis Unit, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Adrian W Moore
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| |
Collapse
|
2
|
Kim MS, Yang Z, Lee JS. In silico identification and characterization of microRNAs from rotifers, cladocerans, and copepods. MARINE POLLUTION BULLETIN 2024; 209:117098. [PMID: 39442355 DOI: 10.1016/j.marpolbul.2024.117098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024]
Abstract
MicroRNAs (miRNAs) are short non-coding RNA molecules that regulate post-transcription and influence various biological processes across species. Despite various studies of miRNAs in vertebrates, plants, and other organisms, miRNA data in aquatic invertebrates are insufficient. In this study, we identified miRNAs from four aquatic invertebrate species that are widely used in aquatic toxicology: the rotifer Brachionus koreanus, the water flea Daphnia magna, the cyclopoid copepod Paracyclopina nana, and the harpacticoid copepod Tigriopus japonicus, using next-generation sequencing and in silico analysis. We identified total 188, 41, 47, and 100 miRNAs from each species, and target genes were predicted based on 3'-untranslated region information. Target prediction and functional annotation results provided the biological processes of these miRNAs in various development-related mechanisms, signaling transduction, and metabolism-related pathways. Moreover, the network between the miRNAs and their targets concerning defense-related and antioxidant genes suggests the suitability of miRNAs as biomarkers in ecotoxicological studies.
Collapse
Affiliation(s)
- Min-Sub Kim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Zhou Yang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, School of Biological Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China
| | - Jae-Seong Lee
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
3
|
Reimels TA, Steinberg M, Yan H, Shahar S, Rosenberg A, Kalafsky K, Luf M, Kelly L, Octaviani S, Pfleger CM. Rabex-5 E3 and Rab5 GEF domains differ in their regulation of Ras, Notch, and PI3K signaling in Drosophila wing development. PLoS One 2024; 19:e0312274. [PMID: 39466792 PMCID: PMC11515992 DOI: 10.1371/journal.pone.0312274] [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: 05/15/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
Rabex-5 (also called RabGEF1), a protein originally characterized for its Rab5 GEF function, also has an A20-like E3 ubiquitin ligase domain. We and others reported that Rabex-5 E3 activity promotes Ras mono- and di-ubiquitination to inhibit Ras signaling in Drosophila and mammals. Subsequently, we reported that Rabex-5 inhibits Notch signaling in the Drosophila hematopoietic system. Here we report genetic interactions using Rabex-5 transgenes encoding domain-specific mutations that show that Rabex-5 requires an intact E3 domain to inhibit Notch signaling in the epithelial tissue of the developing wing. Surprisingly, we discovered that Rabex-5 with an impaired E3 domain but active Rab5 GEF domain suppresses Notch loss-of-function phenotypes and enhances both Notch duplication phenotypes and activated Ras phenotypes consistent with a model that the Rab5 GEF activity of Rabex-5 might positively regulate Ras and Notch. Positive and negative regulation of developmental signaling by its different catalytic domains could allow Rabex-5 to precisely coordinate developmental signaling to fine-tune patterning. Finally, we report that Rabex-5 also inhibits the overgrowth due to loss of PTEN or activation of PI3K but not activation of AKT. Inhibition of Ras, Notch, and PI3K signaling may explain why Rabex-5 is deleted in some cancers. Paradoxically, Rabex-5 is reported to be an oncogene in other cancers. We propose that Rabex-5 acts as a tumor suppressor via its E3 activity to inhibit Ras, Notch, and PI3K signaling and as an oncogene via its Rab5 GEF activity to enhance Ras and Notch signaling.
Collapse
Affiliation(s)
- Theresa A. Reimels
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Mia Steinberg
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
| | - Hua Yan
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Biology, University of Florida, Gainesville, Florida, United States of America
| | - Sivan Shahar
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
| | - Ashley Rosenberg
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
| | - Kristina Kalafsky
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Max Luf
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
| | - Lindsay Kelly
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Stacia Octaviani
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
| | - Cathie M. Pfleger
- Department of Oncological Sciences, Salt Lake City, Utah, United States of America
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
4
|
Sung HH, Li H, Huang YC, Ai CL, Hsieh MY, Jan HM, Peng YJ, Lin HY, Yeh CH, Lin SY, Yeh CY, Cheng YJ, Khoo KH, Lin CH, Chien CT. Galectins induced from hemocytes bridge phosphatidylserine and N-glycosylated Drpr/CED-1 receptor during dendrite pruning. Nat Commun 2024; 15:7402. [PMID: 39191750 DOI: 10.1038/s41467-024-51581-6] [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: 08/29/2023] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
During neuronal pruning, phagocytes engulf shed cellular debris to avoid inflammation and maintain tissue homeostasis. How phagocytic receptors recognize degenerating neurites had been unclear. Here, we identify two glucosyltransferases Alg8 and Alg10 of the N-glycosylation pathway required for dendrite fragmentation and clearance through genetic screen. The scavenger receptor Draper (Drpr) is N-glycosylated with complex- or hybrid-type N-glycans that interact specifically with galectins. We also identify the galectins Crouching tiger (Ctg) and Hidden dragon (Hdg) that interact with N-glycosylated Drpr and function in dendrite pruning via the Drpr pathway. Ctg and Hdg are required in hemocytes for expression and function, and are induced during dendrite injury to localize to injured dendrites through specific interaction with exposed phosphatidylserine (PS) on the surface membrane of injured dendrites. Thus, the galectins Ctg and Hdg bridge the interaction between PS and N-glycosylated Drpr, leading to the activation of phagocytosis.
Collapse
Affiliation(s)
- Hsin-Ho Sung
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hsun Li
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yi-Chun Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chun-Lu Ai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ming-Yen Hsieh
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hau-Ming Jan
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yu-Ju Peng
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hsien-Ya Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chih-Hsuan Yeh
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Shu-Yu Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chun-Yen Yeh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ying-Ju Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Cheng-Ting Chien
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
5
|
Shree S, Howard J. Protocol for Imaging the Same Class IV Neurons at Different Stages of Development. Bio Protoc 2024; 14:e5052. [PMID: 39210953 PMCID: PMC11349491 DOI: 10.21769/bioprotoc.5052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 09/04/2024] Open
Abstract
In this protocol, we focused on analyzing internal branches of Drosophila class IV neurons. These neurons are characterized by their highly branched axons and dendrites and intricately tile the larval body. As Drosophila larvae progress through developmental stages, the dendritic arbors of Class IV neurons undergo notable transformations. As Drosophila larvae develop, their Class IV dendritic arbors grow. In the initial 24 h after egg laying (AEL), the dendrites are smaller than segments. During the subsequent 24 h of the first instar larval stage, dendritic arbors outpace segment growth, achieving tiling. After 48 h, arbors and segments grow concurrently. Epidermal cells near Class IV dendrites expand in proportion to segment growth. This observation suggested that Class IV cells might grow via branch dilation-uniformly elongating branches, akin to Class I cells [1,2]. To understand whether the class IV complex arbor structure is formed by dilation or simply from growing tips, we developed this protocol to introduce a systematic approach for quantitatively assessing the growth dynamics of internal branches. Key features • This protocol employs imaging the same neuron over different development times • Drosophila embryo and larvae genotype is ;;ppkCD4-tdGFP, which explicitly tags class IV neurons • This protocol for the preparation of agar pads to mount and image Drosophila larvae is adapted from Monica Driscoll's method • Neurons are imaged without the use of anesthetics and for a short duration of time • This technique involves the use of a spinning disk confocal microscope.
Collapse
Affiliation(s)
- Sonal Shree
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| |
Collapse
|
6
|
Zhu J, Hou Y, Li W, Wang X, Li F, Li N, Hu Y, Wang X, Ge SN. miR-181a expressed in the dorsal hippocampus regulates the reinstatement of cocaine CPP by targeting PRKAA1. Behav Brain Res 2024; 471:115097. [PMID: 38878971 DOI: 10.1016/j.bbr.2024.115097] [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: 11/09/2023] [Revised: 06/04/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Neuroadaptive changes in the hippocampus underlie addictive-like behaviors in humans or animals chronically exposed to cocaine. miR-181a, which is widely expressed in the hippocampus, acts as a regulator for synaptic plasticity, while its role in drug reinstatement is unclear. In this study, we found that miR-181a regulates the reinstatement of cocaine conditioned place preference(CPP), and altered miR-181a expression changes the complexity of hippocampal neurons and the density and morphology of dendritic spines. By using a luciferase gene reporter, we found that miR-181a targets PRKAA1, an upstream molecule in the mTOR pathway. High miR-181a expression reduced the expression of the PRKAA1 mRNA and promoted mTOR activity and the reinstatement of cocaine CPP. These results indicate that miR-181a is involved in neuronal structural plasticity induced by reinstatement of cocaine CPP, possibly through the activation of the mTOR signaling pathway. This study provides new microRNA targets and a theoretical foundation for the prevention of cocaine-induced reinstatement.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China; Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Yueru Hou
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China; Shaanxi University of Chinese Medicine, Xian Yang, Shaanxi 712046, China
| | - Wan Li
- Xi'an Technological University, Xi'an 710021, China
| | - Xin Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Fei Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Nan Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Yan Hu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Xuelian Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| | - Shun-Nan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| |
Collapse
|
7
|
Lottes EN, Ciger F, Bhattacharjee S, Timmins EA, Tete B, Tran T, Matta J, Patel AA, Cox DN. CCT and Cullin1 Regulate the TORC1 Pathway to Promote Dendritic Arborization in Health and Disease. Cells 2024; 13:1029. [PMID: 38920658 PMCID: PMC11201622 DOI: 10.3390/cells13121029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 06/03/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024] Open
Abstract
The development of cell-type-specific dendritic arbors is integral to the proper functioning of neurons within their circuit networks. In this study, we examine the regulatory relationship between the cytosolic chaperonin CCT, key insulin pathway genes, and an E3 ubiquitin ligase (Cullin1) in dendritic development. CCT loss of function (LOF) results in dendritic hypotrophy in Drosophila Class IV (CIV) multi-dendritic larval sensory neurons, and CCT has recently been shown to fold components of the TOR (Target of Rapamycin) complex 1 (TORC1) in vitro. Through targeted genetic manipulations, we confirm that an LOF of CCT and the TORC1 pathway reduces dendritic complexity, while overexpression of key TORC1 pathway genes increases the dendritic complexity in CIV neurons. Furthermore, both CCT and TORC1 LOF significantly reduce microtubule (MT) stability. CCT has been previously implicated in regulating proteinopathic aggregation, thus, we examine CIV dendritic development in disease conditions as well. The expression of mutant Huntingtin leads to dendritic hypotrophy in a repeat-length-dependent manner, which can be rescued by Cullin1 LOF. Together, our data suggest that Cullin1 and CCT influence dendritic arborization through the regulation of TORC1 in both health and disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
8
|
Mukherjee A, Andrés Jeske Y, Becam I, Taïeb A, Brooks P, Aouad J, Monguillon C, Conduit PT. γ-TuRCs and the augmin complex are required for the development of highly branched dendritic arbors in Drosophila. J Cell Sci 2024; 137:jcs261534. [PMID: 38606636 PMCID: PMC11128279 DOI: 10.1242/jcs.261534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
Microtubules are nucleated by γ-tubulin ring complexes (γ-TuRCs) and are essential for neuronal development. Nevertheless, γ-TuRC depletion has been reported to perturb only higher-order branching in elaborated Drosophila larval class IV dendritic arborization (da) neurons. This relatively mild phenotype has been attributed to defects in microtubule nucleation from Golgi outposts, yet most Golgi outposts lack associated γ-TuRCs. By analyzing dendritic arbor regrowth in pupae, we show that γ-TuRCs are also required for the growth and branching of primary and secondary dendrites, as well as for higher-order branching. Moreover, we identify the augmin complex (hereafter augmin), which recruits γ-TuRCs to the sides of pre-existing microtubules, as being required predominantly for higher-order branching. Augmin strongly promotes the anterograde growth of microtubules in terminal dendrites and thus terminal dendrite stability. Consistent with a specific role in higher-order branching, we find that augmin is expressed less strongly and is largely dispensable in larval class I da neurons, which exhibit few higher-order dendrites. Thus, γ-TuRCs are essential for various aspects of complex dendritic arbor development, and they appear to function in higher-order branching via the augmin pathway, which promotes the elaboration of dendritic arbors to help define neuronal morphology.
Collapse
Affiliation(s)
- Amrita Mukherjee
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Yaiza Andrés Jeske
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Isabelle Becam
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Anaelle Taïeb
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Paul Brooks
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
| | - Joanna Aouad
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | | | - Paul T. Conduit
- Department of Zoology, University of Cambridge, Downing Street, Cambridge CB2 3EJ, UK
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France
| |
Collapse
|
9
|
Luedke KP, Yoshino J, Yin C, Jiang N, Huang JM, Huynh K, Parrish JZ. Dendrite intercalation between epidermal cells tunes nociceptor sensitivity to mechanical stimuli in Drosophila larvae. PLoS Genet 2024; 20:e1011237. [PMID: 38662763 DOI: 10.1371/journal.pgen.1011237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 05/07/2024] [Accepted: 03/29/2024] [Indexed: 05/07/2024] Open
Abstract
An animal's skin provides a first point of contact with the sensory environment, including noxious cues that elicit protective behavioral responses. Nociceptive somatosensory neurons densely innervate and intimately interact with epidermal cells to receive these cues, however the mechanisms by which epidermal interactions shape processing of noxious inputs is still poorly understood. Here, we identify a role for dendrite intercalation between epidermal cells in tuning sensitivity of Drosophila larvae to noxious mechanical stimuli. In wild-type larvae, dendrites of nociceptive class IV da neurons intercalate between epidermal cells at apodemes, which function as body wall muscle attachment sites, but not at other sites in the epidermis. From a genetic screen we identified miR-14 as a regulator of dendrite positioning in the epidermis: miR-14 is expressed broadly in the epidermis but not in apodemes, and miR-14 inactivation leads to excessive apical dendrite intercalation between epidermal cells. We found that miR-14 regulates expression and distribution of the epidermal Innexins ogre and Inx2 and that these epidermal gap junction proteins restrict epidermal dendrite intercalation. Finally, we found that altering the extent of epidermal dendrite intercalation had corresponding effects on nociception: increasing epidermal intercalation sensitized larvae to noxious mechanical inputs and increased mechanically evoked calcium responses in nociceptive neurons, whereas reducing epidermal dendrite intercalation had the opposite effects. Altogether, these studies identify epidermal dendrite intercalation as a mechanism for mechanical coupling of nociceptive neurons to the epidermis, with nociceptive sensitivity tuned by the extent of intercalation.
Collapse
Affiliation(s)
- Kory P Luedke
- Department of Biology, University of Washington, Seattle, Washington State, United States of America
| | - Jiro Yoshino
- Department of Biology, University of Washington, Seattle, Washington State, United States of America
| | - Chang Yin
- Department of Biology, University of Washington, Seattle, Washington State, United States of America
| | - Nan Jiang
- Department of Biology, University of Washington, Seattle, Washington State, United States of America
| | - Jessica M Huang
- Department of Biology, University of Washington, Seattle, Washington State, United States of America
| | - Kevin Huynh
- Department of Biology, University of Washington, Seattle, Washington State, United States of America
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, Washington State, United States of America
| |
Collapse
|
10
|
Mitchell JW, Midillioglu I, Schauer E, Wang B, Han C, Wildonger J. Coordination of Pickpocket ion channel delivery and dendrite growth in Drosophila sensory neurons. PLoS Genet 2023; 19:e1011025. [PMID: 37943859 PMCID: PMC10662761 DOI: 10.1371/journal.pgen.1011025] [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: 07/31/2023] [Revised: 11/21/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Sensory neurons enable an organism to perceive external stimuli, which is essential for survival. The sensory capacity of a neuron depends on the elaboration of its dendritic arbor and the localization of sensory ion channels to the dendritic membrane. However, it is not well understood when and how ion channels localize to growing sensory dendrites and whether their delivery is coordinated with growth of the dendritic arbor. We investigated the localization of the DEG/ENaC/ASIC ion channel Pickpocket (Ppk) in the peripheral sensory neurons of developing fruit flies. We used CRISPR-Cas9 genome engineering approaches to tag endogenous Ppk1 and visualize it live, including monitoring Ppk1 membrane localization via a novel secreted split-GFP approach. Fluorescently tagged endogenous Ppk1 localizes to dendrites, as previously reported, and, unexpectedly, to axons and axon terminals. In dendrites, Ppk1 is present throughout actively growing dendrite branches and is stably integrated into the neuronal cell membrane during the expansive growth of the arbor. Although Ppk channels are dispensable for dendrite growth, we found that an over-active channel mutant severely reduces dendrite growth, likely by acting at an internal membrane and not the dendritic membrane. Our data reveal that the molecular motor dynein and recycling endosome GTPase Rab11 are needed for the proper trafficking of Ppk1 to dendrites. Based on our data, we propose that Ppk channel transport is coordinated with dendrite morphogenesis, which ensures proper ion channel density and distribution in sensory dendrites.
Collapse
Affiliation(s)
- Josephine W. Mitchell
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Biochemistry Department, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Chemistry and Biochemistry, Kalamazoo College, Kalamazoo, Michigan, United States of America
| | - Ipek Midillioglu
- Pediatrics, University of California, San Diego, La Jolla, California, United States of America
| | - Ethan Schauer
- Pediatrics, University of California, San Diego, La Jolla, California, United States of America
| | - Bei Wang
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Chun Han
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York, United States of America
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Jill Wildonger
- Biochemistry Department, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Pediatrics, University of California, San Diego, La Jolla, California, United States of America
- Cell & Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
11
|
Luedke KP, Yoshino J, Yin C, Jiang N, Huang JM, Huynh K, Parrish JZ. Dendrite intercalation between epidermal cells tunes nociceptor sensitivity to mechanical stimuli in Drosophila larvae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557275. [PMID: 37745567 PMCID: PMC10515945 DOI: 10.1101/2023.09.14.557275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
An animal's skin provides a first point of contact with the sensory environment, including noxious cues that elicit protective behavioral responses. Nociceptive somatosensory neurons densely innervate and intimately interact with epidermal cells to receive these cues, however the mechanisms by which epidermal interactions shape processing of noxious inputs is still poorly understood. Here, we identify a role for dendrite intercalation between epidermal cells in tuning sensitivity of Drosophila larvae to noxious mechanical stimuli. In wild-type larvae, dendrites of nociceptive class IV da neurons intercalate between epidermal cells at apodemes, which function as body wall muscle attachment sites, but not at other sites in the epidermis. From a genetic screen we identified miR-14 as a regulator of dendrite positioning in the epidermis: miR-14 is expressed broadly in the epidermis but not in apodemes, and miR-14 inactivation leads to excessive apical dendrite intercalation between epidermal cells. We found that miR-14 regulates expression and distribution of the epidermal Innexins ogre and Inx2 and that these epidermal gap junction proteins restrict epidermal dendrite intercalation. Finally, we found that altering the extent of epidermal dendrite intercalation had corresponding effects on nociception: increasing epidermal intercalation sensitized larvae to noxious mechanical inputs and increased mechanically evoked calcium responses in nociceptive neurons, whereas reducing epidermal dendrite intercalation had the opposite effects. Altogether, these studies identify epidermal dendrite intercalation as a mechanism for mechanical coupling of nociceptive neurons to the epidermis, with nociceptive sensitivity tuned by the extent of intercalation.
Collapse
Affiliation(s)
- Kory P. Luedke
- Department of Biology, University of Washington, Campus Box 351800, Seattle, WA 98195, USA
| | - Jiro Yoshino
- Department of Biology, University of Washington, Campus Box 351800, Seattle, WA 98195, USA
| | - Chang Yin
- Department of Biology, University of Washington, Campus Box 351800, Seattle, WA 98195, USA
| | - Nan Jiang
- Department of Biology, University of Washington, Campus Box 351800, Seattle, WA 98195, USA
| | - Jessica M. Huang
- Department of Biology, University of Washington, Campus Box 351800, Seattle, WA 98195, USA
| | - Kevin Huynh
- Department of Biology, University of Washington, Campus Box 351800, Seattle, WA 98195, USA
| | - Jay Z. Parrish
- Department of Biology, University of Washington, Campus Box 351800, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Lottes EN, Ciger FH, Bhattacharjee S, Timmins-Wilde EA, Tete B, Tran T, Matta J, Patel AA, Cox DN. CCT and Cullin1 regulate the TORC1 pathway to promote dendritic arborization in health and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.31.551324. [PMID: 37577581 PMCID: PMC10418059 DOI: 10.1101/2023.07.31.551324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The development of cell-type-specific dendritic arbors is integral to the proper functioning of neurons within their circuit networks. In this study, we examine the regulatory relationship between the cytosolic chaperonin CCT, key insulin pathway genes, and an E3 ubiquitin ligase (Cullin1) in homeostatic dendritic development. CCT loss of function (LOF) results in dendritic hypotrophy in Drosophila Class IV (CIV) multidendritic larval sensory neurons, and CCT has recently been shown to fold components of the TOR (Target of Rapamycin) complex 1 (TORC1), in vitro. Through targeted genetic manipulations, we have confirmed that LOF of CCT and the TORC1 pathway reduces dendritic complexity, while overexpression of key TORC1 pathway genes increases dendritic complexity in CIV neurons. Both CCT and TORC1 LOF significantly reduce microtubule (MT) stability. CCT has been previously implicated in regulating proteinopathic aggregation, thus we examined CIV dendritic development in disease conditions as well. Expression of mutant Huntingtin leads to dendritic hypotrophy in a repeat-length-dependent manner, which can be rescued by TORC1 disinhibition via Cullin1 LOF. Together, our data suggest that Cullin1 and CCT influence dendritic arborization through regulation of TORC1 in both health and disease.
Collapse
Affiliation(s)
- Erin N. Lottes
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Feyza H. Ciger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | | | | | - Benoit Tete
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Tommy Tran
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Jais Matta
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Atit A. Patel
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, United States
| |
Collapse
|
13
|
Bhattacharjee S, Iyer EPR, Iyer SC, Nanda S, Rubaharan M, Ascoli GA, Cox DN. The Zinc-BED Transcription Factor Bedwarfed Promotes Proportional Dendritic Growth and Branching through Transcriptional and Translational Regulation in Drosophila. Int J Mol Sci 2023; 24:6344. [PMID: 37047316 PMCID: PMC10094446 DOI: 10.3390/ijms24076344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Dendrites are the primary points of sensory or synaptic input to a neuron and play an essential role in synaptic integration and neural function. Despite the functional importance of dendrites, relatively less is known about the underlying mechanisms regulating cell type-specific dendritic patterning. Herein, we have dissected the functional roles of a previously uncharacterized gene, CG3995, in cell type-specific dendritic development in Drosophila melanogaster. CG3995, which we have named bedwarfed (bdwf), encodes a zinc-finger BED-type protein that is required for proportional growth and branching of dendritic arbors. It also exhibits nucleocytoplasmic expression and functions in both transcriptional and translational cellular pathways. At the transcriptional level, we demonstrate a reciprocal regulatory relationship between Bdwf and the homeodomain transcription factor (TF) Cut. We show that Cut positively regulates Bdwf expression and that Bdwf acts as a downstream effector of Cut-mediated dendritic development, whereas overexpression of Bdwf negatively regulates Cut expression in multidendritic sensory neurons. Proteomic analyses revealed that Bdwf interacts with ribosomal proteins and disruption of these proteins resulted in phenotypically similar dendritic hypotrophy defects as observed in bdwf mutant neurons. We further demonstrate that Bdwf and its ribosomal protein interactors are required for normal microtubule and F-actin cytoskeletal architecture. Finally, our findings reveal that Bdwf is required to promote protein translation and ribosome trafficking along the dendritic arbor. These findings shed light on the complex, combinatorial, and multi-functional roles of transcription factors (TFs) in directing the diversification of cell type-specific dendritic development.
Collapse
Affiliation(s)
| | | | | | - Sumit Nanda
- Center for Neural Informatics, Structures, and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA
| | - Myurajan Rubaharan
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - Giorgio A. Ascoli
- Center for Neural Informatics, Structures, and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| |
Collapse
|
14
|
The MicroRNA Ame-Bantam-3p Controls Larval Pupal Development by Targeting the Multiple Epidermal Growth Factor-like Domains 8 Gene (megf8) in the Honeybee, Apis mellifera. Int J Mol Sci 2023; 24:ijms24065726. [PMID: 36982800 PMCID: PMC10054489 DOI: 10.3390/ijms24065726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/19/2023] Open
Abstract
20-Hydroxyecdysone (20E) plays an essential role in coordinating developmental transitions in insects through responsive protein-coding genes and microRNAs (miRNAs). However, the interplay between 20E and miRNAs during insect metamorphosis is unknown. In this study, using small RNA sequencing, a comparative miRNA transcriptomic analysis in different development stages, and 20E treatment, we identified ame-bantam-3p as a key candidate miRNA involved in honeybee metamorphosis. Target prediction and in vitro dual-luciferase assays confirmed that ame-bantam-3p interacts with the coding region of the megf8 gene and promotes its expression. Meanwhile, temporal expression analysis revealed that the expression of ame-bantam-3p is higher in the larval stage than in prepupal and pupal stages, and that this expression pattern is similar to that of megf8. In vivo, we found that the mRNA level of megf8 was significantly increased after the injection of ame-bantam-3p agomir. A 20E feeding assay showed that 20E downregulated the expression of both ame-bantam-3p and its target gene megf8 on larval days five, six, and seven. Meanwhile, the injection of ame-bantam-3p agomir also reduced the 20E titer, as well as the transcript levels of essential ecdysteroid synthesis genes, including Dib, Phm, Sad, and Nvd. The transcript levels of 20E cascade genes, including EcRA, ECRB1, USP, E75, E93, and Br-c, were also significantly decreased after ame-bantam-3p agomir injection. However, ame-bantam-3p antagomir injection and dsmegf8 injection showed the opposite effect to ame-bantam-3p agomir injection. Ame-bantam-3p agomir treatment ultimately led to mortality and the failure of larval pupation by inhibiting ecdysteroid synthesis and the 20E signaling pathway. However, the expression of 20E signaling-related genes was significantly increased after megf8 knockdown, and larvae injected with dsmegf8 showed early pupation. Combined, our results indicate that ame-bantam-3p is involved in the 20E signaling pathway through positively regulating its target gene megf8 and is indispensable for larval–pupal development in the honeybee. These findings may enhance our understanding of the relationship between 20E signaling and small RNAs during honeybee development.
Collapse
|
15
|
Truman JW, Riddiford LM. Drosophila postembryonic nervous system development: a model for the endocrine control of development. Genetics 2023; 223:iyac184. [PMID: 36645270 PMCID: PMC9991519 DOI: 10.1093/genetics/iyac184] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/13/2022] [Indexed: 01/17/2023] Open
Abstract
During postembryonic life, hormones, including ecdysteroids, juvenile hormones, insulin-like peptides, and activin/TGFβ ligands act to transform the larval nervous system into an adult version, which is a fine-grained mosaic of recycled larval neurons and adult-specific neurons. Hormones provide both instructional signals that make cells competent to undergo developmental change and timing cues to evoke these changes across the nervous system. While touching on all the above hormones, our emphasis is on the ecdysteroids, ecdysone and 20-hydroxyecdysone (20E). These are the prime movers of insect molting and metamorphosis and are involved in all phases of nervous system development, including neurogenesis, pruning, arbor outgrowth, and cell death. Ecdysteroids appear as a series of steroid peaks that coordinate the larval molts and the different phases of metamorphosis. Each peak directs a stereotyped cascade of transcription factor expression. The cascade components then direct temporal programs of effector gene expression, but the latter vary markedly according to tissue and life stage. The neurons read the ecdysteroid titer through various isoforms of the ecdysone receptor, a nuclear hormone receptor. For example, at metamorphosis the pruning of larval neurons is mediated through the B isoforms, which have strong activation functions, whereas subsequent outgrowth is mediated through the A isoform through which ecdysteroids play a permissive role to allow local tissue interactions to direct outgrowth. The major circulating ecdysteroid can also change through development. During adult development ecdysone promotes early adult patterning and differentiation while its metabolite, 20E, later evokes terminal adult differentiation.
Collapse
Affiliation(s)
- James W Truman
- Friday Harbor Laboratories, University of Washington, Friday Harbor, WA 98250, USA
- Department of Biology, University of Washington, Box 351800, Seattle, WA 98195, USA
| | - Lynn M Riddiford
- Friday Harbor Laboratories, University of Washington, Friday Harbor, WA 98250, USA
- Department of Biology, University of Washington, Box 351800, Seattle, WA 98195, USA
| |
Collapse
|
16
|
Bhattacharjee S, Iyer EPR, Iyer SC, Nanda S, Rubaharan M, Ascoli GA, Cox DN. The Zinc-BED transcription factor Bedwarfed promotes proportional dendritic growth and branching through transcriptional and translational regulation in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528686. [PMID: 36824896 PMCID: PMC9948997 DOI: 10.1101/2023.02.15.528686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Dendrites are the primary points of sensory or synaptic inputs to a neuron and play an essential role in synaptic integration and neural function. Despite the functional importance of dendrites, relatively less is known about the underlying mechanisms regulating cell-type specific dendritic patterning. Herein, we have dissected functional roles of a previously uncharacterized gene, CG3995 , in cell-type specific dendritic development in Drosophila melanogaster . CG3995 , which we have named bedwarfed ( bdwf ), encodes a zinc-finger BED-type protein which is required for proportional growth and branching of dendritic arbors, exhibits nucleocytoplasmic expression, and functions in both transcriptional and translational cellular pathways. At the transcriptional level, we demonstrate a reciprocal regulatory relationship between Bdwf and the homeodomain transcription factor (TF) Cut. We show that Cut positively regulates Bdwf expression and that Bdwf acts as a downstream effector of Cut-mediated dendritic development, whereas overexpression of Bdwf negatively regulates Cut expression in multidendritic sensory neurons. Proteomic analyses revealed that Bdwf interacts with ribosomal proteins and disruption of these proteins produced phenotypically similar dendritic hypotrophy defects as observed in bdwf mutant neurons. We further demonstrate that Bdwf and its ribosomal protein interactors are required for normal microtubule and F-actin cytoskeletal architecture. Finally, our findings reveal that Bdwf is required to promote protein translation and ribosome trafficking along the dendritic arbor. Taken together, these results provide new insights into the complex, combinatorial and multi-functional roles of transcription factors (TFs) in directing diversification of cell-type specific dendritic development.
Collapse
Affiliation(s)
| | | | | | - Sumit Nanda
- Center for Neural Informatics, Structures, & Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, 22030, USA
| | | | - Giorgio A. Ascoli
- Center for Neural Informatics, Structures, & Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, 22030, USA
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
17
|
Noncoding RNA Regulation of Hormonal and Metabolic Systems in the Fruit Fly Drosophila. Metabolites 2023; 13:metabo13020152. [PMID: 36837772 PMCID: PMC9967906 DOI: 10.3390/metabo13020152] [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: 12/16/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The importance of RNAs is commonly recognised thanks to protein-coding RNAs, whereas non-coding RNAs (ncRNAs) were conventionally regarded as 'junk'. In the last decade, ncRNAs' significance and roles are becoming noticeable in various biological activities, including those in hormonal and metabolic regulation. Among the ncRNAs: microRNA (miRNA) is a small RNA transcript with ~20 nucleotides in length; long non-coding RNA (lncRNA) is an RNA transcript with >200 nucleotides; and circular RNA (circRNA) is derived from back-splicing of pre-mRNA. These ncRNAs can regulate gene expression levels at epigenetic, transcriptional, and post-transcriptional levels through various mechanisms in insects. A better understanding of these crucial regulators is essential to both basic and applied entomology. In this review, we intend to summarise and discuss the current understanding and knowledge of miRNA, lncRNA, and circRNA in the best-studied insect model, the fruit fly Drosophila.
Collapse
|
18
|
Kanaoka Y, Onodera K, Watanabe K, Hayashi Y, Usui T, Uemura T, Hattori Y. Inter-organ Wingless/Ror/Akt signaling regulates nutrient-dependent hyperarborization of somatosensory neurons. eLife 2023; 12:79461. [PMID: 36647607 PMCID: PMC9844989 DOI: 10.7554/elife.79461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 12/11/2022] [Indexed: 01/18/2023] Open
Abstract
Nutrition in early life has profound effects on an organism, altering processes such as organogenesis. However, little is known about how specific nutrients affect neuronal development. Dendrites of class IV dendritic arborization neurons in Drosophila larvae become more complex when the larvae are reared on a low-yeast diet compared to a high-yeast diet. Our systematic search for key nutrients revealed that the neurons increase their dendritic terminal densities in response to a combined deficiency in vitamins, metal ions, and cholesterol. The deficiency of these nutrients upregulates Wingless in a closely located tissue, body wall muscle. Muscle-derived Wingless activates Akt in the neurons through the receptor tyrosine kinase Ror, which promotes the dendrite branching. In larval muscles, the expression of wingless is regulated not only in this key nutrient-dependent manner, but also by the JAK/STAT signaling pathway. Additionally, the low-yeast diet blunts neuronal light responsiveness and light avoidance behavior, which may help larvae optimize their survival strategies under low-nutritional conditions. Together, our studies illustrate how the availability of specific nutrients affects neuronal development through inter-organ signaling.
Collapse
Affiliation(s)
| | - Koun Onodera
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Kaori Watanabe
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Yusaku Hayashi
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Tadao Usui
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
| | - Tadashi Uemura
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
- Research Center for Dynamic Living Systems, Kyoto UniversityKyotoJapan
- AMED-CRESTTokyoJapan
| | - Yukako Hattori
- Graduate School of Biostudies, Kyoto UniversityKyotoJapan
- JST FORESTTokyoJapan
| |
Collapse
|
19
|
O’Brien CE, Younger SH, Jan LY, Jan YN. The GARP complex prevents sterol accumulation at the trans-Golgi network during dendrite remodeling. J Biophys Biochem Cytol 2022; 222:213548. [PMID: 36239632 PMCID: PMC9577387 DOI: 10.1083/jcb.202112108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 08/11/2022] [Accepted: 09/20/2022] [Indexed: 11/29/2022] Open
Abstract
Membrane trafficking is essential for sculpting neuronal morphology. The GARP and EARP complexes are conserved tethers that regulate vesicle trafficking in the secretory and endolysosomal pathways, respectively. Both complexes contain the Vps51, Vps52, and Vps53 proteins, and a complex-specific protein: Vps54 in GARP and Vps50 in EARP. In Drosophila, we find that both complexes are required for dendrite morphogenesis during developmental remodeling of multidendritic class IV da (c4da) neurons. Having found that sterol accumulates at the trans-Golgi network (TGN) in Vps54KO/KO neurons, we investigated genes that regulate sterols and related lipids at the TGN. Overexpression of oxysterol binding protein (Osbp) or knockdown of the PI4K four wheel drive (fwd) exacerbates the Vps54KO/KO phenotype, whereas eliminating one allele of Osbp rescues it, suggesting that excess sterol accumulation at the TGN is, in part, responsible for inhibiting dendrite regrowth. These findings distinguish the GARP and EARP complexes in neurodevelopment and implicate vesicle trafficking and lipid transfer pathways in dendrite morphogenesis.
Collapse
Affiliation(s)
- Caitlin E. O’Brien
- Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA,Department of Physiology, University of California at San Francisco, San Francisco, CA
| | - Susan H. Younger
- Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA,Department of Physiology, University of California at San Francisco, San Francisco, CA
| | - Lily Yeh Jan
- Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA,Department of Physiology, University of California at San Francisco, San Francisco, CA,Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA
| | - Yuh Nung Jan
- Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA,Department of Physiology, University of California at San Francisco, San Francisco, CA,Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA
| |
Collapse
|
20
|
Luo J, Zhao S, Ren Q, Wang Q, Chen Z, Cui J, Jing Y, Liu P, Yan R, Song X, Liu G, Li X. Dynamic Analysis of microRNAs from Different Life Stages of Rhipicephalus microplus (Acari: Ixodidae) by High-Throughput Sequencing. Pathogens 2022; 11:pathogens11101148. [PMID: 36297205 PMCID: PMC9611014 DOI: 10.3390/pathogens11101148] [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: 09/20/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022] Open
Abstract
MicroRNAs (miRNAs), which are small, noncoding RNA molecules, play an important regulatory role in gene expression at the posttranscriptional level. Relatively limited knowledge exists on miRNAs in Rhipicephalus microplus ticks in China; however, understanding the physiology of miRNA functions and expression at different developmental stages is important. In this study, three small RNA libraries were constructed for R. microplus eggs, larvae, and female adults; miRNAs were detected during these developmental stages by high-throughput sequencing, with 18,162,337, 8,090,736, and 11,807,326 clean reads, respectively. A total of 5132 known miRNAs and 31 novel miRNAs were identified. A total of 1736 differentially expressed miRNAs were significantly different at a p-value of <0.01; in female adults, 467 microRNAs were upregulated and 376 miRNAs downregulated compared to larval tick controls. Using larvae as controls, 218 upregulated and 203 downregulated miRNAs were detected in eggs; in eggs, 108 miRNAs were upregulated and 364 downregulated compared to female adults controls. To verify the reliability of the sequencing data, RT−qPCR was applied to compare expression levels of novel miRNAs. Some differentially expressed miRNAs are involved in developmental physiology, signal transduction, and cell-extracellular communications based on GO annotation and KEGG pathway analyses. Here, we provide a dynamic analysis of miRNAs in R. microplus and their potential targets, which has significance for understanding the biology of ticks and lays the foundation for improved understanding of miRNA functioning in the regulation of R. microplus development. These results can assist future miRNA studies in other tick species that have great significance for human and animal health.
Collapse
Affiliation(s)
- Jin Luo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Shuaiyang Zhao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Qiaoyun Ren
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Qilin Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Zeyu Chen
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Jingjing Cui
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Yujiao Jing
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Peiwen Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Guangyuan Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou 730046, China
- Correspondence: (G.L.); (X.L.)
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: (G.L.); (X.L.)
| |
Collapse
|
21
|
Li H, Sung HH, Huang YC, Cheng YJ, Yeh HF, Pi H, Giniger E, Chien CT. Fringe-positive Golgi outposts unite temporal Furin 2 convertase activity and spatial Delta signal to promote dendritic branch retraction. Cell Rep 2022; 40:111372. [PMID: 36130510 PMCID: PMC11463699 DOI: 10.1016/j.celrep.2022.111372] [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: 03/08/2022] [Revised: 07/07/2022] [Accepted: 08/25/2022] [Indexed: 11/19/2022] Open
Abstract
Golgi outposts (GOPs) in dendrites are known for their role in promoting branch extension, but whether GOPs have other functions is unclear. We found that terminal branches of Drosophila class IV dendritic arborization (C4da) neurons actively grow during the early third-instar (E3) larval stage but retract in the late third (L3) stage. Interestingly, the Fringe (Fng) glycosyltransferase localizes increasingly at GOPs in distal dendritic regions through the E3 to the L3 stage. Expression of the endopeptidase Furin 2 (Fur2), which proteolyzes and inactivates Fng, decreases from E3 to L3 in C4da neurons, thereby increasing Fng-positive GOPs in dendrites. The epidermal Delta ligand and neuronal Notch receptor, the substrate for Fng-mediated O-glycosylation, also negatively regulate dendrite growth. Fng inhibits actin dynamics in dendrites, linking dendritic branch retraction to suppression of the C4da-mediated thermal nociception response in late larval stages. Thus, Fng-positive GOPs function in dendrite retraction, which would add another function to the repertoire of GOPs in dendrite arborization.
Collapse
Affiliation(s)
- Hsun Li
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan; Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 11529, Taiwan
| | - Hsin-Ho Sung
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chun Huang
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Ying-Ju Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Hsiao-Fong Yeh
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan
| | - Haiwei Pi
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cheng-Ting Chien
- Institute of Molecular Biology, Academia Sinica, Taipei 11529, Taiwan; Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 11529, Taiwan; Neuroscience Program of Academia Sinica, Academia Sinica, Taipei 11529, Taiwan.
| |
Collapse
|
22
|
Hobin M, Dorfman K, Adel M, Rivera-Rodriguez EJ, Kuklin EA, Ma D, Griffith LC. The Drosophila microRNA bantam regulates excitability in adult mushroom body output neurons to promote early night sleep. iScience 2022; 25:104874. [PMID: 36034229 PMCID: PMC9400086 DOI: 10.1016/j.isci.2022.104874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 07/07/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Sleep circuitry evolved to have both dedicated and context-dependent modulatory elements. Identifying modulatory subcircuits and understanding their molecular machinery is a major challenge for the sleep field. Previously, we identified 25 sleep-regulating microRNAs in Drosophila melanogaster, including the developmentally important microRNA bantam. Here we show that bantam acts in the adult to promote early nighttime sleep through a population of glutamatergic neurons that is intimately involved in applying contextual information to behaviors, the γ5β'2a/β'2mp/β'2mp_bilateral Mushroom Body Output Neurons (MBONs). Calcium imaging revealed that bantam inhibits the activity of these cells during the early night, but not the day. Blocking synaptic transmission in these MBONs rescued the effect of bantam knockdown. This suggests bantam promotes early night sleep via inhibition of the γ5β'2a/β'2mp/β'2mp_bilateral MBONs. RNAseq identifies Kelch and CCHamide-2 receptor as possible mediators, establishing a new role for bantam as an active regulator of sleep and neural activity in the adult fly.
Collapse
Affiliation(s)
- Michael Hobin
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Katherine Dorfman
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Mohamed Adel
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Emmanuel J. Rivera-Rodriguez
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Elena A. Kuklin
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| | - Dingbang Ma
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
- Howard Hughes Medical Institute, Brandeis University, Waltham, MA 02454-9110, USA
| | - Leslie C. Griffith
- Department of Biology, Volen National Center for Complex Systems, Brandeis University, Waltham, MA 02454-9110, USA
| |
Collapse
|
23
|
A photo-switchable assay system for dendrite degeneration and repair in Drosophila melanogaster. Proc Natl Acad Sci U S A 2022; 119:e2204577119. [PMID: 35969739 PMCID: PMC9407391 DOI: 10.1073/pnas.2204577119] [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] [Indexed: 11/18/2022] Open
Abstract
Neurodegeneration arising from aging, injury, or diseases has devastating health consequences. Whereas neuronal survival and axon degeneration have been studied extensively, much less is known about how neurodegeneration affects dendrites, in part due to the limited assay systems available. To develop an assay for dendrite degeneration and repair, we used photo-switchable caspase-3 (caspase-Light-Oxygen-Voltage-sensing [caspase-LOV]) in peripheral class 4 dendrite arborization (c4da) neurons to induce graded neurodegeneration by adjusting illumination duration during development and adulthood in Drosophila melanogaster. We found that both developing and mature c4da neurons were able to survive while sustaining mild neurodegeneration induced by moderate caspase-LOV activation. Further, we observed active dendrite addition and dendrite regeneration in developing and mature c4da neurons, respectively. Using this assay, we found that the mouse Wallerian degeneration slow (WldS) protein can protect c4da neurons from caspase-LOV-induced dendrite degeneration and cell death. Furthermore, our data show that WldS can reduce dendrite elimination without affecting dendrite addition. In summary, we successfully established a photo-switchable assay system in both developing and mature neurons and used WldS as a test case to study the mechanisms underlying dendrite regeneration and repair.
Collapse
|
24
|
Shree S, Sutradhar S, Trottier O, Tu Y, Liang X, Howard J. Dynamic instability of dendrite tips generates the highly branched morphologies of sensory neurons. SCIENCE ADVANCES 2022; 8:eabn0080. [PMID: 35767611 PMCID: PMC9242452 DOI: 10.1126/sciadv.abn0080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
The highly ramified arbors of neuronal dendrites provide the substrate for the high connectivity and computational power of the brain. Altered dendritic morphology is associated with neuronal diseases. Many molecules have been shown to play crucial roles in shaping and maintaining dendrite morphology. However, the underlying principles by which molecular interactions generate branched morphologies are not understood. To elucidate these principles, we visualized the growth of dendrites throughout larval development of Drosophila sensory neurons and found that the tips of dendrites undergo dynamic instability, transitioning rapidly and stochastically between growing, shrinking, and paused states. By incorporating these measured dynamics into an agent-based computational model, we showed that the complex and highly variable dendritic morphologies of these cells are a consequence of the stochastic dynamics of their dendrite tips. These principles may generalize to branching of other neuronal cell types, as well as to branching at the subcellular and tissue levels.
Collapse
Affiliation(s)
- Sonal Shree
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Olivier Trottier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Department of Physics, Yale University, New Haven, CT 06511, USA
| | - Yuhai Tu
- IBM T.J. Watson Research Center, Yorktown Heights, NY 10598, USA
| | - Xin Liang
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Department of Physics, Yale University, New Haven, CT 06511, USA
- Quantitative Biology Institute, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
25
|
The branching code: A model of actin-driven dendrite arborization. Cell Rep 2022; 39:110746. [PMID: 35476974 DOI: 10.1016/j.celrep.2022.110746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/24/2021] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
The cytoskeleton is crucial for defining neuronal-type-specific dendrite morphologies. To explore how the complex interplay of actin-modulatory proteins (AMPs) can define neuronal types in vivo, we focused on the class III dendritic arborization (c3da) neuron of Drosophila larvae. Using computational modeling, we reveal that the main branches (MBs) of c3da neurons follow general models based on optimal wiring principles, while the actin-enriched short terminal branches (STBs) require an additional growth program. To clarify the cellular mechanisms that define this second step, we thus concentrated on STBs for an in-depth quantitative description of dendrite morphology and dynamics. Applying these methods systematically to mutants of six known and novel AMPs, we revealed the complementary roles of these individual AMPs in defining STB properties. Our data suggest that diverse dendrite arbors result from a combination of optimal-wiring-related growth and individualized growth programs that are neuron-type specific.
Collapse
|
26
|
Shrestha BR, Burgos A, Grueber WB. The Immunoglobulin Superfamily Member Basigin Is Required for Complex Dendrite Formation in Drosophila. Front Cell Neurosci 2021; 15:739741. [PMID: 34803611 PMCID: PMC8600269 DOI: 10.3389/fncel.2021.739741] [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: 07/11/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Coordination of dendrite growth with changes in the surrounding substrate occurs widely in the nervous system and is vital for establishing and maintaining neural circuits. However, the molecular basis of this important developmental process remains poorly understood. To identify potential mediators of neuron-substrate interactions important for dendrite morphogenesis, we undertook an expression pattern-based screen in Drosophila larvae, which revealed many proteins with expression in dendritic arborization (da) sensory neurons and in neurons and their epidermal substrate. We found that reporters for Basigin, a cell surface molecule of the immunoglobulin (Ig) superfamily previously implicated in cell-cell and cell-substrate interactions, are expressed in da sensory neurons and epidermis. Loss of Basigin in da neurons led to defects in morphogenesis of the complex dendrites of class IV da neurons. Classes of sensory neurons with simpler branching patterns were unaffected by loss of Basigin. Structure-function analyses showed that a juxtamembrane KRR motif is critical for this function. Furthermore, knock down of Basigin in the epidermis led to defects in dendrite elaboration of class IV neurons, suggesting a non-autonomous role. Together, our findings support a role for Basigin in complex dendrite morphogenesis and interactions between dendrites and the adjacent epidermis.
Collapse
Affiliation(s)
- Brikha R Shrestha
- Department of Neuroscience, Columbia University Medical Center, New York, NY, United States
| | - Anita Burgos
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| | - Wesley B Grueber
- Department of Neuroscience, Columbia University Medical Center, New York, NY, United States.,Department of Neuroscience, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States.,Department of Physiology and Cellular Biophysics, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
| |
Collapse
|
27
|
Kilo L, Stürner T, Tavosanis G, Ziegler AB. Drosophila Dendritic Arborisation Neurons: Fantastic Actin Dynamics and Where to Find Them. Cells 2021; 10:2777. [PMID: 34685757 PMCID: PMC8534399 DOI: 10.3390/cells10102777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 01/27/2023] Open
Abstract
Neuronal dendrites receive, integrate, and process numerous inputs and therefore serve as the neuron's "antennae". Dendrites display extreme morphological diversity across different neuronal classes to match the neuron's specific functional requirements. Understanding how this structural diversity is specified is therefore important for shedding light on information processing in the healthy and diseased nervous system. Popular models for in vivo studies of dendrite differentiation are the four classes of dendritic arborization (c1da-c4da) neurons of Drosophila larvae with their class-specific dendritic morphologies. Using da neurons, a combination of live-cell imaging and computational approaches have delivered information on the distinct phases and the time course of dendrite development from embryonic stages to the fully developed dendritic tree. With these data, we can start approaching the basic logic behind differential dendrite development. A major role in the definition of neuron-type specific morphologies is played by dynamic actin-rich processes and the regulation of their properties. This review presents the differences in the growth programs leading to morphologically different dendritic trees, with a focus on the key role of actin modulatory proteins. In addition, we summarize requirements and technological progress towards the visualization and manipulation of such actin regulators in vivo.
Collapse
Affiliation(s)
- Lukas Kilo
- Dendrite Differentiation, German Center for Neurodegenerative Diseases, 53115 Bonn, Germany; (L.K.); (G.T.)
| | - Tomke Stürner
- Department of Zoology, University of Cambridge, Cambridge CB2 1TN, UK;
| | - Gaia Tavosanis
- Dendrite Differentiation, German Center for Neurodegenerative Diseases, 53115 Bonn, Germany; (L.K.); (G.T.)
- LIMES-Institute, University of Bonn, 53115 Bonn, Germany
| | - Anna B. Ziegler
- Institute of Neuro- and Behavioral Biology, University of Münster, 48149 Münster, Germany
| |
Collapse
|
28
|
Yin C, Peterman E, Rasmussen JP, Parrish JZ. Transparent Touch: Insights From Model Systems on Epidermal Control of Somatosensory Innervation. Front Cell Neurosci 2021; 15:680345. [PMID: 34135734 PMCID: PMC8200473 DOI: 10.3389/fncel.2021.680345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/28/2021] [Indexed: 12/28/2022] Open
Abstract
Somatosensory neurons (SSNs) densely innervate our largest organ, the skin, and shape our experience of the world, mediating responses to sensory stimuli including touch, pressure, and temperature. Historically, epidermal contributions to somatosensation, including roles in shaping innervation patterns and responses to sensory stimuli, have been understudied. However, recent work demonstrates that epidermal signals dictate patterns of SSN skin innervation through a variety of mechanisms including targeting afferents to the epidermis, providing instructive cues for branching morphogenesis, growth control and structural stability of neurites, and facilitating neurite-neurite interactions. Here, we focus onstudies conducted in worms (Caenorhabditis elegans), fruit flies (Drosophila melanogaster), and zebrafish (Danio rerio): prominent model systems in which anatomical and genetic analyses have defined fundamental principles by which epidermal cells govern SSN development.
Collapse
Affiliation(s)
| | | | | | - Jay Z. Parrish
- Department of Biology, University of Washington, Seattle, WA, United States
| |
Collapse
|
29
|
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
|
30
|
Alizzi RA, Xu D, Tenenbaum CM, Wang W, Gavis ER. The ELAV/Hu protein Found in neurons regulates cytoskeletal and ECM adhesion inputs for space-filling dendrite growth. PLoS Genet 2020; 16:e1009235. [PMID: 33370772 PMCID: PMC7793258 DOI: 10.1371/journal.pgen.1009235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 01/08/2021] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
Dendritic arbor morphology influences how neurons receive and integrate extracellular signals. We show that the ELAV/Hu family RNA-binding protein Found in neurons (Fne) is required for space-filling dendrite growth to generate highly branched arbors of Drosophila larval class IV dendritic arborization neurons. Dendrites of fne mutant neurons are shorter and more dynamic than in wild-type, leading to decreased arbor coverage. These defects result from both a decrease in stable microtubules and loss of dendrite-substrate interactions within the arbor. Identification of transcripts encoding cytoskeletal regulators and cell-cell and cell-ECM interacting proteins as Fne targets using TRIBE further supports these results. Analysis of one target, encoding the cell adhesion protein Basigin, indicates that the cytoskeletal defects contributing to branch instability in fne mutant neurons are due in part to decreased Basigin expression. The ability of Fne to coordinately regulate the cytoskeleton and dendrite-substrate interactions in neurons may shed light on the behavior of cancer cells ectopically expressing ELAV/Hu proteins. Different types of neurons have different sizes and shapes that are tailored to their particular functions. In the fruit fly larva, a set of sensory neurons called class IV da neurons have highly branched trees of dendrites that cover the epidermis to sense potentially harmful stimuli. Neurons whose dendrites completely fill the territory they sample are also found in zebrafish, worms, mice and humans. We show that an RNA-binding protein called Fne plays an important role in coordinating different contributions to dendrite branching in class IV da neurons by impacting the organization of the cytoskeleton within the neuron and the ability of the dendrite to contact the substratum outside of it. The identification of mRNAs that code for cytoskeleton regulators and adhesive proteins as targets of Fne using a genome-wide approach further supports these results. While the ability of Fne to exert control over such proteins is crucial to generating the space-filling growth of neurons, it can be deleterious if not properly employed, such as when the homologs of Fne are expressed in cancer cells.
Collapse
Affiliation(s)
- Rebecca A. Alizzi
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Derek Xu
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Conrad M. Tenenbaum
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Wei Wang
- Lewis-Sigler Institute, Princeton University, Princeton, New Jersey, United States of America
| | - Elizabeth R. Gavis
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
31
|
Ferreira Castro A, Baltruschat L, Stürner T, Bahrami A, Jedlicka P, Tavosanis G, Cuntz H. Achieving functional neuronal dendrite structure through sequential stochastic growth and retraction. eLife 2020; 9:e60920. [PMID: 33241995 PMCID: PMC7837678 DOI: 10.7554/elife.60920] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/15/2020] [Indexed: 02/06/2023] Open
Abstract
Class I ventral posterior dendritic arborisation (c1vpda) proprioceptive sensory neurons respond to contractions in the Drosophila larval body wall during crawling. Their dendritic branches run along the direction of contraction, possibly a functional requirement to maximise membrane curvature during crawling contractions. Although the molecular machinery of dendritic patterning in c1vpda has been extensively studied, the process leading to the precise elaboration of their comb-like shapes remains elusive. Here, to link dendrite shape with its proprioceptive role, we performed long-term, non-invasive, in vivo time-lapse imaging of c1vpda embryonic and larval morphogenesis to reveal a sequence of differentiation stages. We combined computer models and dendritic branch dynamics tracking to propose that distinct sequential phases of stochastic growth and retraction achieve efficient dendritic trees both in terms of wire and function. Our study shows how dendrite growth balances structure-function requirements, shedding new light on general principles of self-organisation in functionally specialised dendrites.
Collapse
Affiliation(s)
- André Ferreira Castro
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
- Ernst Strüngmann Institute (ESI) for Neuroscience in cooperation with Max Planck SocietyFrankfurt am MainGermany
- Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | | | - Tomke Stürner
- Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| | | | - Peter Jedlicka
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
- Faculty of Medicine, ICAR3R – Interdisciplinary Centre for 3Rs in Animal Research, Justus Liebig University GiessenGiessenGermany
- Neuroscience Center, Institute of Clinical Neuroanatomy, Goethe UniversityFrankfurt am MainGermany
| | - Gaia Tavosanis
- Center for Neurodegenerative Diseases (DZNE)BonnGermany
- LIMES Institute, University of BonnBonnGermany
| | - Hermann Cuntz
- Frankfurt Institute for Advanced StudiesFrankfurt am MainGermany
- Ernst Strüngmann Institute (ESI) for Neuroscience in cooperation with Max Planck SocietyFrankfurt am MainGermany
| |
Collapse
|
32
|
Palavalli A, Tizón-Escamilla N, Rupprecht JF, Lecuit T. Deterministic and Stochastic Rules of Branching Govern Dendrite Morphogenesis of Sensory Neurons. Curr Biol 2020; 31:459-472.e4. [PMID: 33212017 DOI: 10.1016/j.cub.2020.10.054] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/22/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Dendrite morphology is necessary for the correct integration of inputs that neurons receive. The branching mechanisms allowing neurons to acquire their type-specific morphology remain unclear. Classically, axon and dendrite patterns were shown to be guided by molecules, providing deterministic cues. However, the extent to which deterministic and stochastic mechanisms, based upon purely statistical bias, contribute to the emergence of dendrite shape is largely unknown. We address this issue using the Drosophila class I vpda multi-dendritic neurons. Detailed quantitative analysis of vpda dendrite morphogenesis indicates that the primary branch grows very robustly in a fixed direction, though secondary branch numbers and lengths showed fluctuations characteristic of stochastic systems. Live-tracking dendrites and computational modeling revealed how neuron shape emerges from few local statistical parameters of branch dynamics. We report key opposing aspects of how tree architecture feedbacks on the local probability of branch shrinkage. Child branches promote stabilization of parent branches, although self-repulsion promotes shrinkage. Finally, we show that self-repulsion, mediated by the adhesion molecule Dscam1, indirectly patterns the growth of secondary branches by spatially restricting their direction of stable growth perpendicular to the primary branch. Thus, the stochastic nature of secondary branch dynamics and the existence of geometric feedback emphasize the importance of self-organization in neuronal dendrite morphogenesis.
Collapse
Affiliation(s)
- Amrutha Palavalli
- Aix Marseille Université and CNRS, IBDM - UMR7288 and Turing Centre for Living Systems Campus de Luminy Case 907, Marseille 13288, France
| | - Nicolás Tizón-Escamilla
- Aix-Marseille Université, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems Campus de Luminy Case 907, Marseille 13288, France
| | - Jean-François Rupprecht
- Aix-Marseille Université, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems Campus de Luminy Case 907, Marseille 13288, France.
| | - Thomas Lecuit
- Aix Marseille Université and CNRS, IBDM - UMR7288 and Turing Centre for Living Systems Campus de Luminy Case 907, Marseille 13288, France; Collège de France, 11 Place Marcelin Berthelot, Paris 75005, France.
| |
Collapse
|
33
|
Kim YY, Yoon JH, Um JH, Jeong DJ, Shin DJ, Hong YB, Kim JK, Kim DH, Kim C, Chung CG, Lee SB, Koh H, Yun J. PINK1 alleviates thermal hypersensitivity in a paclitaxel-induced Drosophila model of peripheral neuropathy. PLoS One 2020; 15:e0239126. [PMID: 32941465 PMCID: PMC7498067 DOI: 10.1371/journal.pone.0239126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 08/31/2020] [Indexed: 11/18/2022] Open
Abstract
Paclitaxel is a representative anticancer drug that induces chemotherapy-induced peripheral neuropathy (CIPN), a common side effect that limits many anticancer chemotherapies. Although PINK1, a key mediator of mitochondrial quality control, has been shown to protect neuronal cells from various toxic treatments, the role of PINK1 in CIPN has not been investigated. Here, we examined the effect of PINK1 expression on CIPN using a recently established paclitaxel-induced peripheral neuropathy model in Drosophila larvae. We found that the class IV dendritic arborization (C4da) sensory neuron-specific expression of PINK1 significantly ameliorated the paclitaxel-induced thermal hyperalgesia phenotype. In contrast, knockdown of PINK1 resulted in an increase in thermal hypersensitivity, suggesting a critical role for PINK1 in sensory neuron-mediated thermal nociceptive sensitivity. Interestingly, analysis of the C4da neuron morphology suggests that PINK1 expression alleviates paclitaxel-induced thermal hypersensitivity by means other than preventing alterations in sensory dendrites in C4da neurons. We found that paclitaxel induces mitochondrial dysfunction in C4da neurons and that PINK1 expression suppressed the paclitaxel-induced increase in mitophagy in C4da neurons. These results suggest that PINK1 mitigates paclitaxel-induced sensory dendrite alterations and restores mitochondrial homeostasis in C4da neurons and that improvement in mitochondrial quality control could be a promising strategy for the treatment of CIPN.
Collapse
Affiliation(s)
- Young Yeon Kim
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Jeong-Hyun Yoon
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Jee-Hyun Um
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Dae Jin Jeong
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Dong Jin Shin
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Young Bin Hong
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Jong Kuk Kim
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Neurology, College of Medicine, Dong‐A University, Busan, Republic of Korea
| | - Dong Hyun Kim
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A University, Busan, Republic of Korea
| | - Changsoo Kim
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea
| | - Chang Geon Chung
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Sung Bae Lee
- Department of Pharmacology, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Hyongjong Koh
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Pharmacology, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Jeanho Yun
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Republic of Korea
- * E-mail:
| |
Collapse
|
34
|
Kitatani Y, Tezuka A, Hasegawa E, Yanagi S, Togashi K, Tsuji M, Kondo S, Parrish JZ, Emoto K. Drosophila miR-87 promotes dendrite regeneration by targeting the transcriptional repressor Tramtrack69. PLoS Genet 2020; 16:e1008942. [PMID: 32764744 PMCID: PMC7439810 DOI: 10.1371/journal.pgen.1008942] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/19/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
To remodel functional neuronal connectivity, neurons often alter dendrite arbors through elimination and subsequent regeneration of dendritic branches. However, the intrinsic mechanisms underlying this developmentally programmed dendrite regeneration and whether it shares common machinery with injury-induced regeneration remain largely unknown. Drosophila class IV dendrite arborization (C4da) sensory neurons regenerate adult-specific dendrites after eliminating larval dendrites during metamorphosis. Here we show that the microRNA miR-87 is a critical regulator of dendrite regeneration in Drosophila. miR-87 knockout impairs dendrite regeneration after developmentally-programmed pruning, whereas miR-87 overexpression in C4da neurons leads to precocious initiation of dendrite regeneration. Genetic analyses indicate that the transcriptional repressor Tramtrack69 (Ttk69) is a functional target for miR-87-mediated repression as ttk69 expression is increased in miR-87 knockout neurons and reducing ttk69 expression restores dendrite regeneration to mutants lacking miR-87 function. We further show that miR-87 is required for dendrite regeneration after acute injury in the larval stage, providing a mechanistic link between developmentally programmed and injury-induced dendrite regeneration. These findings thus indicate that miR-87 promotes dendrite regrowth during regeneration at least in part through suppressing Ttk69 in Drosophila sensory neurons and suggest that developmental and injury-induced dendrite regeneration share a common intrinsic mechanism to reactivate dendrite growth. Dendrites are the primary sites for synaptic and sensory inputs. To remodel or repair neuronal connectivity, dendrites often exhibit large-scale structural changes that can be triggered by developmental signals, alterations in sensory inputs, or injury. Despite the importance of dendritic remodeling to nervous system function, the molecular basis for this remodeling is largely unknown. Here we used an unbiased genetic screen and in vivo imaging in Drosophila sensory neurons to demonstrate that the microRNA miR-87 is a critical factor required in neurons to reactivate dendritic growth both in developmental remodeling and following injury. Our work supports the model that miR-87 promotes dendrite regeneration by blocking expression of the transcriptional repressor Tramtrack69 in neurons. This study thus establishes a role for miRNAs in temporal control of dendrite regeneration.
Collapse
Affiliation(s)
- Yasuko Kitatani
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Akane Tezuka
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Eri Hasegawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Satoyoshi Yanagi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Kazuya Togashi
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Masato Tsuji
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shu Kondo
- Genetic Strains Research Center, National Institute of Genetics, Yata, Mishima, Shizuoka, Japan
| | - Jay Z. Parrish
- Department of Biology, University of Washington, Seattle, Washington, United States of America
- * E-mail: (JZP); (KE)
| | - Kazuo Emoto
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), University of Tokyo, Hongo, Bunkyo-ku, Tokyo, Japan
- * E-mail: (JZP); (KE)
| |
Collapse
|
35
|
Ho CH, Treisman JE. Specific Isoforms of the Guanine-Nucleotide Exchange Factor dPix Couple Neuromuscular Synapse Growth to Muscle Growth. Dev Cell 2020; 54:117-131.e5. [PMID: 32516570 DOI: 10.1016/j.devcel.2020.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/09/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022]
Abstract
Developmental growth requires coordination between the growth rates of individual tissues and organs. Here, we examine how Drosophila neuromuscular synapses grow to match the size of their target muscles. We show that changes in muscle growth driven by autonomous modulation of insulin receptor signaling produce corresponding changes in synapse size, with each muscle affecting only its presynaptic motor neuron branches. This scaling growth is mechanistically distinct from synaptic plasticity driven by neuronal activity and requires increased postsynaptic differentiation induced by insulin receptor signaling in muscle. We identify the guanine-nucleotide exchange factor dPix as an effector of insulin receptor signaling. Alternatively spliced dPix isoforms that contain a specific exon are necessary and sufficient for postsynaptic differentiation and scaling growth, and their mRNA levels are regulated by insulin receptor signaling. These findings define a mechanism by which the same signaling pathway promotes both autonomous muscle growth and non-autonomous synapse growth.
Collapse
Affiliation(s)
- Cheuk Hei Ho
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
36
|
Wiebe KF, Elebute OO, LeMoine CMR, Cassone BJ. A Day in the Life: Identification of Developmentally Regulated MicroRNAs in the Colorado Potato Beetle (Leptinotarsa decemlineata; Coleoptera: Chrysomelidae). JOURNAL OF ECONOMIC ENTOMOLOGY 2020; 113:1445-1454. [PMID: 32150604 DOI: 10.1093/jee/toaa020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Indexed: 06/10/2023]
Abstract
The Colorado potato beetle (Leptinotarsa decemlineata (Say)) is an important pest of the cultivated potato (Solanum tuberosum (L.) [Solanales: Solanaceae]). With its broad resistance toward commonly used insecticides, it is clear that more sophisticated control strategies are needed. Due to their importance in insect development, microRNAs (miRNAs) represent a potential tool to employ in insect control strategies. However, most studies conducted in this area have focused on model species with well-annotated genomes. In this study, next-generation sequencing was used to catalogue the miRNAs produced by L. decemlineata across all eight stages of its development, from eggs to adults. For most stages, the length of miRNAs peaked between 21 and 22 nt, though it was considerably longer for the egg stage (26 nt). Global profiling of miRNAs revealed three distinct developmental clusters: 1) egg stage; 2) early stage (first, second, and third instar); and 3) late stage (fourth instar, prepupae, pupae, and adult). We identified 86 conserved miRNAs and 33 bonafide novel miRNAs, including stage-specific miRNAs and those not previously identified in L. decemlineata. Most of the conserved miRNAs were found in multiple developmental stages, whereas the novel miRNAs were often stage specific with the bulk identified in the egg stage. The identified miRNAs have a myriad of putative functions, including growth, reproduction, and insecticide resistance. We discuss the putative roles of some of the most notable miRNAs in the regulation of L. decemlineata development, as well as the potential applications of this research in Colorado potato beetle management.
Collapse
Affiliation(s)
- K F Wiebe
- Department of Biology, Brandon University, Brandon, Canada
| | - O O Elebute
- Department of Biology, Brandon University, Brandon, Canada
| | - C M R LeMoine
- Department of Biology, Brandon University, Brandon, Canada
| | - B J Cassone
- Department of Biology, Brandon University, Brandon, Canada
| |
Collapse
|
37
|
Poe AR, Xu Y, Zhang C, Lei J, Li K, Labib D, Han C. Low FoxO expression in Drosophila somatosensory neurons protects dendrite growth under nutrient restriction. eLife 2020; 9:53351. [PMID: 32427101 PMCID: PMC7308081 DOI: 10.7554/elife.53351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/18/2020] [Indexed: 12/22/2022] Open
Abstract
During prolonged nutrient restriction, developing animals redistribute vital nutrients to favor brain growth at the expense of other organs. In Drosophila, such brain sparing relies on a glia-derived growth factor to sustain proliferation of neural stem cells. However, whether other aspects of neural development are also spared under nutrient restriction is unknown. Here we show that dynamically growing somatosensory neurons in the Drosophila peripheral nervous system exhibit organ sparing at the level of arbor growth: Under nutrient stress, sensory dendrites preferentially grow as compared to neighboring non-neural tissues, resulting in dendrite overgrowth. These neurons express lower levels of the stress sensor FoxO than neighboring epidermal cells, and hence exhibit no marked induction of autophagy and a milder suppression of Tor signaling under nutrient stress. Preferential dendrite growth allows for heightened animal responses to sensory stimuli, indicative of a potential survival advantage under environmental challenges.
Collapse
Affiliation(s)
- Amy R Poe
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Yineng Xu
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Christine Zhang
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Joyce Lei
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Kailyn Li
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - David Labib
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Chun Han
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| |
Collapse
|
38
|
Yang WK, Chien CT. Beyond being innervated: the epidermis actively shapes sensory dendritic patterning. Open Biol 2020; 9:180257. [PMID: 30914004 PMCID: PMC6451362 DOI: 10.1098/rsob.180257] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sensing environmental cues requires well-built neuronal circuits linked to the body surface. Sensory neurons generate dendrites to innervate surface epithelium, thereby making it the largest sensory organ in the body. Previous studies have illustrated that neuronal type, physiological function and branching patterns are determined by intrinsic factors. Perhaps for effective sensation or protection, sensory dendrites bind to or are surrounded by the substrate epidermis. Recent studies have shed light on the mechanisms by which dendrites interact with their substrates. These interactions suggest that substrates can regulate dendrite guidance, arborization and degeneration. In this review, we focus on recent studies of Drosophila and Caenorhabditis elegans that demonstrate how epidermal cells can regulate dendrites in several aspects.
Collapse
Affiliation(s)
- Wei-Kang Yang
- Institute of Molecular Biology, Academia Sinica , Taipei 115 , Taiwan
| | - Cheng-Ting Chien
- Institute of Molecular Biology, Academia Sinica , Taipei 115 , Taiwan
| |
Collapse
|
39
|
Hu C, Kanellopoulos AK, Richter M, Petersen M, Konietzny A, Tenedini FM, Hoyer N, Cheng L, Poon CLC, Harvey KF, Windhorst S, Parrish JZ, Mikhaylova M, Bagni C, Calderon de Anda F, Soba P. Conserved Tao Kinase Activity Regulates Dendritic Arborization, Cytoskeletal Dynamics, and Sensory Function in Drosophila. J Neurosci 2020; 40:1819-1833. [PMID: 31964717 PMCID: PMC7046460 DOI: 10.1523/jneurosci.1846-19.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/13/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Dendritic arborization is highly regulated and requires tight control of dendritic growth, branching, cytoskeletal dynamics, and ion channel expression to ensure proper function. Abnormal dendritic development can result in altered network connectivity, which has been linked to neurodevelopmental disorders, including autism spectrum disorders (ASDs). How neuronal growth control programs tune dendritic arborization to ensure function is still not fully understood. Using Drosophila dendritic arborization (da) neurons as a model, we identified the conserved Ste20-like kinase Tao as a negative regulator of dendritic arborization. We show that Tao kinase activity regulates cytoskeletal dynamics and sensory channel localization required for proper sensory function in both male and female flies. We further provide evidence for functional conservation of Tao kinase, showing that its ASD-linked human ortholog, Tao kinase 2 (Taok2), could replace Drosophila Tao and rescue dendritic branching, dynamic microtubule alterations, and behavioral defects. However, several ASD-linked Taok2 variants displayed impaired rescue activity, suggesting that Tao/Taok2 mutations can disrupt sensory neuron development and function. Consistently, we show that Tao kinase activity is required in developing and as well as adult stages for maintaining normal dendritic arborization and sensory function to regulate escape and social behavior. Our data suggest an important role for Tao kinase signaling in cytoskeletal organization to maintain proper dendritic arborization and sensory function, providing a strong link between developmental sensory aberrations and behavioral abnormalities relevant for Taok2-dependent ASDs.SIGNIFICANCE STATEMENT Autism spectrum disorders (ASDs) are linked to abnormal dendritic arbors. However, the mechanisms of how dendritic arbors develop to promote functional and proper behavior are unclear. We identified Drosophila Tao kinase, the ortholog of the ASD risk gene Taok2, as a regulator of dendritic arborization in sensory neurons. We show that Tao kinase regulates cytoskeletal dynamics, controls sensory ion channel localization, and is required to maintain somatosensory function in vivo Interestingly, ASD-linked human Taok2 mutations rendered it nonfunctional, whereas its WT form could restore neuronal morphology and function in Drosophila lacking endogenous Tao. Our findings provide evidence for a conserved role of Tao kinase in dendritic development and function of sensory neurons, suggesting that aberrant sensory function might be a common feature of ASDs.
Collapse
Affiliation(s)
- Chun Hu
- Neuronal Patterning and Connectivity Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | | - Melanie Richter
- Neuronal Development Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Meike Petersen
- Neuronal Patterning and Connectivity Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Anja Konietzny
- Neuronal Protein Transport Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Federico M Tenedini
- Neuronal Patterning and Connectivity Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Nina Hoyer
- Neuronal Patterning and Connectivity Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Lin Cheng
- Neuronal Patterning and Connectivity Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Carole L C Poon
- Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, 3000 Victoria, Australia
- Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, 3800 Victoria, Australia
| | - Sabine Windhorst
- Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, 98195 Washington, and
| | - Marina Mikhaylova
- Neuronal Protein Transport Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Froylan Calderon de Anda
- Neuronal Development Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Peter Soba
- Neuronal Patterning and Connectivity Laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany,
| |
Collapse
|
40
|
Arnés M, Romero N, Casas-Tintó S, Acebes Á, Ferrús A. PI3K activation prevents Aβ42-induced synapse loss and favors insoluble amyloid deposit formation. Mol Biol Cell 2019; 31:244-260. [PMID: 31877058 PMCID: PMC7183762 DOI: 10.1091/mbc.e19-05-0303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Excess of Aβ42 peptide is considered a hallmark of the disease. Here we express the human Aβ42 peptide to assay the neuroprotective effects of PI3K in adult Drosophila melanogaster. The neuronal expression of the human peptide elicits progressive toxicity in the adult fly. The pathological traits include reduced axonal transport, synapse loss, defective climbing ability and olfactory perception, as well as lifespan reduction. The Aβ42-dependent synapse decay does not involve transcriptional changes in the core synaptic protein encoding genes bruchpilot, liprin and synaptobrevin. All toxicity features, however, are suppressed by the coexpression of PI3K. Moreover, PI3K activation induces a significant increase of 6E10 and thioflavin-positive amyloid deposits. Mechanistically, we suggest that Aβ42-Ser26 could be a candidate residue for direct or indirect phosphorylation by PI3K. Along with these in vivo experiments, we further analyze Aβ42 toxicity and its suppression by PI3K activation in in vitro assays with SH-SY5Y human neuroblastoma cell cultures, where Aβ42 aggregation into large insoluble deposits is reproduced. Finally, we show that the Aβ42 toxicity syndrome includes the transcriptional shut down of PI3K expression. Taken together, these results uncover a potential novel pharmacological strategy against this disease through the restoration of PI3K activity.
Collapse
Affiliation(s)
| | - Ninovska Romero
- Department of Basic Medical Sciences, Institute of Biomedical Technologies, University of La Laguna, Tenerife 38200, Spain
| | | | - Ángel Acebes
- Cajal Institute (CSIC), Madrid 28002, Spain.,Department of Basic Medical Sciences, Institute of Biomedical Technologies, University of La Laguna, Tenerife 38200, Spain
| | | |
Collapse
|
41
|
Glia-derived exosomal miR-274 targets Sprouty in trachea and synaptic boutons to modulate growth and responses to hypoxia. Proc Natl Acad Sci U S A 2019; 116:24651-24661. [PMID: 31666321 PMCID: PMC6900535 DOI: 10.1073/pnas.1902537116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Secreted exosomal microRNAs (miRNAs) mediate interorgan/tissue communications by modulating target gene expression, thereby regulating developmental and physiological functions. However, the source, route, and function in target cells have not been formally established for specific miRNAs. Here, we show that glial miR-274 non-cell-autonomously modulates the growth of synaptic boutons and tracheal branches. Whereas the precursor form of miR-274 is expressed in glia, the mature form of miR-274 distributes broadly, including in synaptic boutons, muscle cells, and tracheal cells. Mature miR-274 is secreted from glia to the circulating hemolymph as an exosomal cargo, a process requiring ESCRT components in exosome biogenesis and Rab11 and Syx1A in exosome release. We further show that miR-274 can function in the neurons or tracheal cells to modulate the growth of synaptic boutons and tracheal branches, respectively. Also, miR-274 uptake into the target cells by AP-2-dependent mechanisms modulates target cell growth. In the target cells, miR-274 down-regulates Sprouty (Sty) through a targeting sequence at the sty 3' untranslated region, thereby enhancing MAPK signaling and promoting cell growth. miR-274 expressed in glia of an mir-274 null mutant is released as an exosomal cargo in the circulating hemolymph, and such glial-specific expression resets normal levels of Sty and MAPK signaling and modulates target cell growth. mir-274 mutant larvae are hypersensitive to hypoxia, which is suppressed by miR-274 expression in glia or by increasing tracheal branches. Thus, glia-derived miR-274 coordinates growth of synaptic boutons and tracheal branches to modulate larval hypoxia responses.
Collapse
|
42
|
Wang LH, Baker NE. Salvador-Warts-Hippo pathway regulates sensory organ development via caspase-dependent nonapoptotic signaling. Cell Death Dis 2019; 10:669. [PMID: 31511495 PMCID: PMC6739336 DOI: 10.1038/s41419-019-1924-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 08/03/2019] [Accepted: 08/27/2019] [Indexed: 12/19/2022]
Abstract
The fundamental roles for the Salvador-Warts-Hippo (SWH) pathway are widely characterized in growth regulation and organ size control. However, the function of SWH pathway is less known in cell fate determination. Here we uncover a novel role of the SWH signaling pathway in determination of cell fate during neural precursor (sensory organ precursor, SOP) development. Inactivation of the SWH pathway in SOP of the wing imaginal discs affects caspase-dependent bristle patterning in an apoptosis-independent process. Such nonapoptotic functions of caspases have been implicated in inflammation, proliferation, cellular remodeling, and cell fate determination. Our data indicate an effect on the Wingless (Wg)/Wnt pathway. Previously, caspases were proposed to cleave and activate a negative regulator of Wg/Wnt signaling, Shaggy (Sgg)/GSK3β. Surprisingly, we found that a noncleavable form of Sgg encoded from the endogenous locus after CRISPR-Cas9 modification supported almost normal bristle patterning, indicating that Sgg might not be the main target of the caspase-dependent nonapoptotic process. Collectively, our results outline a new function of SWH signaling that crosstalks to caspase-dependent nonapoptotic signaling and Wg/Wnt signaling in neural precursor development, which might be implicated in neuronal pathogenesis.
Collapse
Affiliation(s)
- Lan-Hsin Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, 161 Sec 6, Minquan E. Rd, Taipei, 11490, Taiwan.
| | - Nicholas E Baker
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|
43
|
Tenedini FM, Sáez González M, Hu C, Pedersen LH, Petruzzi MM, Spitzweck B, Wang D, Richter M, Petersen M, Szpotowicz E, Schweizer M, Sigrist SJ, Calderon de Anda F, Soba P. Maintenance of cell type-specific connectivity and circuit function requires Tao kinase. Nat Commun 2019; 10:3506. [PMID: 31383864 PMCID: PMC6683158 DOI: 10.1038/s41467-019-11408-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/13/2019] [Indexed: 01/05/2023] Open
Abstract
Sensory circuits are typically established during early development, yet how circuit specificity and function are maintained during organismal growth has not been elucidated. To gain insight we quantitatively investigated synaptic growth and connectivity in the Drosophila nociceptive network during larval development. We show that connectivity between primary nociceptors and their downstream neurons scales with animal size. We further identified the conserved Ste20-like kinase Tao as a negative regulator of synaptic growth required for maintenance of circuit specificity and connectivity. Loss of Tao kinase resulted in exuberant postsynaptic specializations and aberrant connectivity during larval growth. Using functional imaging and behavioral analysis we show that loss of Tao-induced ectopic synapses with inappropriate partner neurons are functional and alter behavioral responses in a connection-specific manner. Our data show that fine-tuning of synaptic growth by Tao kinase is required for maintaining specificity and behavioral output of the neuronal network during animal growth.
Collapse
Affiliation(s)
- Federico Marcello Tenedini
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Maria Sáez González
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Chun Hu
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Lisa Hedegaard Pedersen
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Mabel Matamala Petruzzi
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Bettina Spitzweck
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Denan Wang
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Melanie Richter
- Neuronal Development laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Meike Petersen
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Emanuela Szpotowicz
- Electron microscopy unit, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Michaela Schweizer
- Electron microscopy unit, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Stephan J Sigrist
- Institute of Biology, Free University Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Froylan Calderon de Anda
- Neuronal Development laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany
| | - Peter Soba
- Neuronal Patterning and Connectivity laboratory, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Falkenried 94, 20251, Hamburg, Germany.
| |
Collapse
|
44
|
Rosa JB, Sagasti A. Developmental Neurobiology: It Takes Nrg to Separate Dendrites. Curr Biol 2019; 29:R327-R329. [PMID: 31063725 DOI: 10.1016/j.cub.2019.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The development of sensory receptive fields requires the coordinated spatial patterning of neurites from multiple sensory neuron subtypes. A new study identifies a role for neuron-skin cell interactions in preventing the bundling of dendritic arbors from distinct neurons.
Collapse
Affiliation(s)
- Jeffrey B Rosa
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA 90095-1606, USA
| | - Alvaro Sagasti
- Department of Molecular, Cell, and Developmental Biology, UCLA, Los Angeles, CA 90095-1606, USA.
| |
Collapse
|
45
|
Jiang N, Rasmussen JP, Clanton JA, Rosenberg MF, Luedke KP, Cronan MR, Parker ED, Kim HJ, Vaughan JC, Sagasti A, Parrish JZ. A conserved morphogenetic mechanism for epidermal ensheathment of nociceptive sensory neurites. eLife 2019; 8:42455. [PMID: 30855229 PMCID: PMC6450671 DOI: 10.7554/elife.42455] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
Interactions between epithelial cells and neurons influence a range of sensory modalities including taste, touch, and smell. Vertebrate and invertebrate epidermal cells ensheath peripheral arbors of somatosensory neurons, including nociceptors, yet the developmental origins and functional roles of this ensheathment are largely unknown. Here, we describe an evolutionarily conserved morphogenetic mechanism for epidermal ensheathment of somatosensory neurites. We found that somatosensory neurons in Drosophila and zebrafish induce formation of epidermal sheaths, which wrap neurites of different types of neurons to different extents. Neurites induce formation of plasma membrane phosphatidylinositol 4,5-bisphosphate microdomains at nascent sheaths, followed by a filamentous actin network, and recruitment of junctional proteins that likely form autotypic junctions to seal sheaths. Finally, blocking epidermal sheath formation destabilized dendrite branches and reduced nociceptive sensitivity in Drosophila. Epidermal somatosensory neurite ensheathment is thus a deeply conserved cellular process that contributes to the morphogenesis and function of nociceptive sensory neurons. Humans and other animals perceive and interact with the outside world through their sensory nervous system. Nerve cells, acting as the body’s ‘telegraph wires’, convey signals from sensory organs – like the eyes – to the brain, which then processes this information and tells the body how to respond. There are different kinds of sensory nerve cells that carry different types of information, but they all associate closely with the tissues and organs they connect to the brain. Human skin contains sensory nerve cells, which underpin our senses of touch and pain. There is a highly specialized, complex connection between some of these nerve cells and cells in the skin: the skin cells wrap tightly around the nerve cells’ free ends, forming sheath-like structures. This ‘ensheathment’ process happens in a wide range of animals, including those with a backbone, like fish and humans, and those without, like insects. Ensheathment is thought to be important for the skin’s nerve cells to work properly. Yet it remains unclear how or when these connections first appear. Jiang et al. therefore wanted to determine the developmental origins of ensheathment and to find out if these were also similar in animals with and without backbones. Experiments using fruit fly and zebrafish embryos revealed that nerve cells, not skin cells, were responsible for forming and maintaining the sheaths. In embryos where groups of sensory nerve cells were selectively killed – either using a laser or by making the cells produce a toxin – ensheathment did not occur. Further studies, using a variety of microscopy techniques, revealed that the molecular machinery required to stabilize the sheaths was similar in both fish and flies, and therefore likely to be conserved across different groups of animals. Removing sheaths in fly embryos led to nerve cells becoming unstable; the animals were also less sensitive to touch. This confirmed that ensheathment was indeed necessary for sensory nerve cells to work properly. By revealing how ensheathment first emerges, these findings shed new light on how the sensory nervous system develops and how its activity is controlled. In humans, skin cells ensheath the nerve cells responsible for sensing pain. A better understanding of how ensheathments first arise could therefore lead to new avenues for treating chronic pain and related conditions.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Biology, University of Washington, Seattle, United States
| | - Jeffrey P Rasmussen
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Joshua A Clanton
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Marci F Rosenberg
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Kory P Luedke
- Department of Biology, University of Washington, Seattle, United States
| | - Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University, Durham, United States
| | - Edward D Parker
- Department of Opthalmology, University of Washington, Seattle, United States
| | - Hyeon-Jin Kim
- Department of Chemistry, University of Washington, Seattle, United States.,Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, United States.,Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Alvaro Sagasti
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, United States
| |
Collapse
|
46
|
Low IIC, Williams CR, Chong MK, McLachlan IG, Wierbowski BM, Kolotuev I, Heiman MG. Morphogenesis of neurons and glia within an epithelium. Development 2019; 146:dev171124. [PMID: 30683663 PMCID: PMC6398450 DOI: 10.1242/dev.171124] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 01/15/2019] [Indexed: 12/13/2022]
Abstract
To sense the outside world, some neurons protrude across epithelia, the cellular barriers that line every surface of our bodies. To study the morphogenesis of such neurons, we examined the C. elegans amphid, in which dendrites protrude through a glial channel at the nose. During development, amphid dendrites extend by attaching to the nose via DYF-7, a type of protein typically found in epithelial apical ECM. Here, we show that amphid neurons and glia exhibit epithelial properties, including tight junctions and apical-basal polarity, and develop in a manner resembling other epithelia. We find that DYF-7 is a fibril-forming apical ECM component that promotes formation of the tube-shaped glial channel, reminiscent of roles for apical ECM in other narrow epithelial tubes. We also identify a requirement for FRM-2, a homolog of EPBL15/moe/Yurt that promotes epithelial integrity in other systems. Finally, we show that other environmentally exposed neurons share a requirement for DYF-7. Together, our results suggest that these neurons and glia can be viewed as part of an epithelium continuous with the skin, and are shaped by mechanisms shared with other epithelia.
Collapse
Affiliation(s)
- Isabel I C Low
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Claire R Williams
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Megan K Chong
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Ian G McLachlan
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Bradley M Wierbowski
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Irina Kolotuev
- Université de Rennes 1, Plateforme microscopie électronique, 35043 Rennes, France
| | - Maxwell G Heiman
- Department of Genetics, Blavatnik Institute, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| |
Collapse
|
47
|
Yoong LF, Pai YJ, Moore AW. Stages and transitions in dendrite arbor differentiation. Neurosci Res 2019; 138:70-78. [DOI: 10.1016/j.neures.2018.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022]
|
48
|
Clark SG, Graybeal LL, Bhattacharjee S, Thomas C, Bhattacharya S, Cox DN. Basal autophagy is required for promoting dendritic terminal branching in Drosophila sensory neurons. PLoS One 2018; 13:e0206743. [PMID: 30395636 PMCID: PMC6218061 DOI: 10.1371/journal.pone.0206743] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/18/2018] [Indexed: 01/22/2023] Open
Abstract
Dendrites function as the primary sites for synaptic input and integration with impairments in dendritic arborization being associated with dysfunctional neuronal circuitry. Post-mitotic neurons require high levels of basal autophagy to clear cytotoxic materials and autophagic dysfunction under native or cellular stress conditions has been linked to neuronal cell death as well as axo-dendritic degeneration. However, relatively little is known regarding the developmental role of basal autophagy in directing aspects of dendritic arborization or the mechanisms by which the autophagic machinery may be transcriptionally regulated to promote dendritic diversification. We demonstrate that autophagy-related (Atg) genes are positively regulated by the homeodomain transcription factor Cut, and that basal autophagy functions as a downstream effector pathway for Cut-mediated dendritic terminal branching in Drosophila multidendritic (md) sensory neurons. Further, loss of function analyses implicate Atg genes in promoting cell type-specific dendritic arborization and terminal branching, while gain of function studies suggest that excessive autophagy leads to dramatic reductions in dendritic complexity. We demonstrate that the Atg1 initiator kinase interacts with the dual leucine zipper kinase (DLK) pathway by negatively regulating the E3 ubiquitin ligase Highwire and positively regulating the MAPKKK Wallenda. Finally, autophagic induction partially rescues dendritic atrophy defects observed in a model of polyglutamine toxicity. Collectively, these studies implicate transcriptional control of basal autophagy in directing dendritic terminal branching and demonstrate the importance of homeostatic control of autophagic levels for dendritic arbor complexity under native or cellular stress conditions.
Collapse
Affiliation(s)
- Sarah G. Clark
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Lacey L. Graybeal
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Shatabdi Bhattacharjee
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Caroline Thomas
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Surajit Bhattacharya
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
49
|
Kane NS, Vora M, Padgett RW, Li Y. bantam microRNA is a negative regulator of the Drosophila decapentaplegic pathway. Fly (Austin) 2018; 12:105-117. [PMID: 30015555 PMCID: PMC6150632 DOI: 10.1080/19336934.2018.1499370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Decapentaplegic (Dpp), the Drosophila homolog of the vertebrate bone morphogenetic protein (BMP2/4), is crucial for patterning and growth in many developmental contexts. The Dpp pathway is regulated at many different levels to exquisitely control its activity. We show that bantam (ban), a microRNA, modulates Dpp signaling activity. Over expression of ban decreases phosphorylated Mothers against decapentaplegic (Mad) levels and negatively affects Dpp pathway transcriptional target genes, while null mutant clones of ban upregulate the pathway. We provide evidence that dpp upregulates ban in the wing imaginal disc, and attenuation of Dpp signaling results in a reduction of ban expression, showing that they function in a feedback loop. Furthermore, we show that this feedback loop is important for maintaining anterior-posterior compartment boundary stability in the wing disc through regulation of optomotor blind (omb), a known target of the pathway. Our results support a model that ban functions with dpp in a negative feedback loop.
Collapse
Affiliation(s)
- Nanci S Kane
- a Waksman Institute, Department of Molecular Biology and Biochemistry , Cancer Institute of New Jersey, Rutgers University , Piscataway , NJ , USA
| | - Mehul Vora
- a Waksman Institute, Department of Molecular Biology and Biochemistry , Cancer Institute of New Jersey, Rutgers University , Piscataway , NJ , USA
| | - Richard W Padgett
- a Waksman Institute, Department of Molecular Biology and Biochemistry , Cancer Institute of New Jersey, Rutgers University , Piscataway , NJ , USA
| | - Ying Li
- b Life Science Institute , Chongqing Medical University , Chongqing , China
| |
Collapse
|
50
|
Brazill JM, Cruz B, Zhu Y, Zhai RG. Nmnat mitigates sensory dysfunction in a Drosophila model of paclitaxel-induced peripheral neuropathy. Dis Model Mech 2018; 11:dmm.032938. [PMID: 29716954 PMCID: PMC6031360 DOI: 10.1242/dmm.032938] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/25/2018] [Indexed: 01/03/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is the major dose-limiting side effect of many commonly used chemotherapeutic agents, including paclitaxel. Currently, there are no neuroprotective or effective symptomatic treatments for CIPN. Lack of understanding of the in vivo mechanisms of CIPN has greatly impeded the identification of therapeutic targets. Here, we optimized a model of paclitaxel-induced peripheral neuropathy using Drosophila larvae that recapitulates aspects of chemotherapy-induced sensory dysfunction. We showed that nociceptive sensitivity is associated with disrupted organization of microtubule-associated MAP1B/Futsch and aberrant stabilization of peripheral sensory dendrites. These findings establish a robust and amenable model for studying peripheral mechanisms of CIPN. Using this model, we uncovered a critical role for nicotinamide mononucleotide adenylyltransferase (Nmnat) in maintaining the integrity and function of peripheral sensory neurons and uncovered Nmnat's therapeutic potential against diverse sensory symptoms of CIPN. Summary: Neurotoxic side effects of chemotherapy are poorly understood. Here, the authors optimize a Drosophila model of paclitaxel-induced sensory dysfunction, which is then used to explore the neuroprotective capacity of Nmnat.
Collapse
Affiliation(s)
- Jennifer M Brazill
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Beverley Cruz
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yi Zhu
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - R Grace Zhai
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA .,School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong 264005, China
| |
Collapse
|