1
|
Thi Thu Trinh M, Thoa THK, Thi Phuong Thao D. Neuronal effect of 0.3 % DMSO and the synergism between 0.3 % DMSO and loss function of UCH-L1 on Drosophila melanogaster model. Toxicol Rep 2025; 14:101904. [PMID: 39897395 PMCID: PMC11783005 DOI: 10.1016/j.toxrep.2025.101904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Dimethyl sulfoxide (DMSO) is a polar aprotic solvent which is widely used in biological and medical studies and as a vehicle for pharmacological therapy. DMSO from 0.1 % to 0.5 %, particularly 0.3 % is commonly used as solvent to dissolve compounds when testing their effect on living cell, tissues including nerve cell. However, scientific data on the effects of DMSO on nervous system is limited. Here, we present our data of case study on investigation the effects of DMSO at 0.3 % concentration on nerve cell of Drosophila melanogaster model. We found that 0.3 % DMSO concentration had affected on the active zone and glutamate receptor. Notably, this study also revealed the synergistic effect of 0.3 % DMSO and loss function of dUCH (the homolog of Ubiquitin Carboxyl terminal Hydrolase -L1, UCH-L1 in D. melanogaster). This combination caused more serious abnormalities in synapse structure, particularly number of boutons on Neuromuscular Junction, NMJ. Furthermore, 0.3 % DMSO reduced the amount of ubiquitinylated protein aggregates in the indirect flight muscle of both normal and genectic defect fly model. Taken together, data in this sytudy indicated that 0.3 % DMSO caused the aberrant morphology of the synaptic structure and decreased the number of ubiquitinylated proteins in the indirect flight muscle of Drosophila. The data from the study contributed new evidence of the effects of DMSO on the nervous system. Signigicantly, this study revealed that DMSO affected on neuron cell at low concentration which widely used as pharmacological solvent.
Collapse
Affiliation(s)
- Mai Thi Thu Trinh
- Laboratory of Molecular Biotechnology, University of Science-VNU.HCM, Viet Nam
- Department of Molecular and Environmental Biotechnology, Faculty of Biology -Biotechnology, University of Science-VNU.HCM, Viet Nam
| | - Truong Huynh Kim Thoa
- Laboratory of Molecular Biotechnology, University of Science-VNU.HCM, Viet Nam
- Department of Molecular and Environmental Biotechnology, Faculty of Biology -Biotechnology, University of Science-VNU.HCM, Viet Nam
| | - Dang Thi Phuong Thao
- Laboratory of Molecular Biotechnology, University of Science-VNU.HCM, Viet Nam
- Department of Molecular and Environmental Biotechnology, Faculty of Biology -Biotechnology, University of Science-VNU.HCM, Viet Nam
| |
Collapse
|
2
|
Ehrhardt E, Whitehead SC, Namiki S, Minegishi R, Siwanowicz I, Feng K, Otsuna H, FlyLight Project Team, Meissner GW, Stern D, Truman J, Shepherd D, Dickinson MH, Ito K, Dickson BJ, Cohen I, Card GM, Korff W. Single-cell type analysis of wing premotor circuits in the ventral nerve cord of Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.05.31.542897. [PMID: 37398009 PMCID: PMC10312520 DOI: 10.1101/2023.05.31.542897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
To perform most behaviors, animals must send commands from higher-order processing centers in the brain to premotor circuits that reside in ganglia distinct from the brain, such as the mammalian spinal cord or insect ventral nerve cord. How these circuits are functionally organized to generate the great diversity of animal behavior remains unclear. An important first step in unraveling the organization of premotor circuits is to identify their constituent cell types and create tools to monitor and manipulate these with high specificity to assess their functions. This is possible in the tractable ventral nerve cord of the fly. To generate such a toolkit, we used a combinatorial genetic technique (split-GAL4) to create 195 sparse transgenic driver lines targeting 196 individual cell types in the ventral nerve cord. These included wing and haltere motoneurons, modulatory neurons, and interneurons. Using a combination of behavioral, developmental, and anatomical analyses, we systematically characterized the cell types targeted in our collection. In addition, we identified correspondences between the cells in this collection and a recent connectomic data set of the ventral nerve cord. Taken together, the resources and results presented here form a powerful toolkit for future investigations of neuronal circuits and connectivity of premotor circuits while linking them to behavioral outputs.
Collapse
Affiliation(s)
- Erica Ehrhardt
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
- Institute of Zoology, University of Cologne, Zülpicher Str 47b, 50674 Cologne, Germany
| | - Samuel C Whitehead
- Physics Department, Cornell University, 509 Clark Hall, Ithaca, New York 14853, USA
- California Institute of Technology, 1200 E California Blvd, Pasadena, California 91125, USA
| | - Shigehiro Namiki
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| | - Ryo Minegishi
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
- Queensland Brain Institute, University of Queensland, 79 Upland Rd, Brisbane, QLD, 4072, Australia
| | - Igor Siwanowicz
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| | - Kai Feng
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
- Queensland Brain Institute, University of Queensland, 79 Upland Rd, Brisbane, QLD, 4072, Australia
| | - Hideo Otsuna
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| | - FlyLight Project Team
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| | - Geoffrey W Meissner
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| | - David Stern
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| | - Jim Truman
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
- Department of Biology, University of Washington, Seattle, Washington 98195, USA
| | - David Shepherd
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Life Sciences Building, Southampton SO17 1BJ
| | - Michael H Dickinson
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
- California Institute of Technology, 1200 E California Blvd, Pasadena, California 91125, USA
| | - Kei Ito
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
- Institute of Zoology, University of Cologne, Zülpicher Str 47b, 50674 Cologne, Germany
| | - Barry J Dickson
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
- Queensland Brain Institute, University of Queensland, 79 Upland Rd, Brisbane, QLD, 4072, Australia
| | - Itai Cohen
- Physics Department, Cornell University, 509 Clark Hall, Ithaca, New York 14853, USA
| | - Gwyneth M Card
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| | - Wyatt Korff
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Dr, Ashburn, Virginia 20147, USA
| |
Collapse
|
3
|
Jones E, McLaughlin KA. A Novel Perspective on Neuronal Control of Anatomical Patterning, Remodeling, and Maintenance. Int J Mol Sci 2023; 24:13358. [PMID: 37686164 PMCID: PMC10488252 DOI: 10.3390/ijms241713358] [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: 07/20/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
While the nervous system may be best known as the sensory communication center of an organism, recent research has revealed a myriad of multifaceted roles for both the CNS and PNS from early development to adult regeneration and remodeling. These systems work to orchestrate tissue pattern formation during embryonic development and continue shaping pattering through transitional periods such as metamorphosis and growth. During periods of injury or wounding, the nervous system has also been shown to influence remodeling and wound healing. The neuronal mechanisms responsible for these events are largely conserved across species, suggesting this evidence may be important in understanding and resolving many human defects and diseases. By unraveling these diverse roles, this paper highlights the necessity of broadening our perspective on the nervous system beyond its conventional functions. A comprehensive understanding of the complex interactions and contributions of the nervous system throughout development and adulthood has the potential to revolutionize therapeutic strategies and open new avenues for regenerative medicine and tissue engineering. This review highlights an important role for the nervous system during the patterning and maintenance of complex tissues and provides a potential avenue for advancing biomedical applications.
Collapse
Affiliation(s)
| | - Kelly A. McLaughlin
- Department of Biology, Tufts University, 200 Boston Avenue, Suite 4700, Medford, MA 02155, USA;
| |
Collapse
|
4
|
Püffel F, Meyer L, Imirzian N, Roces F, Johnston R, Labonte D. Developmental biomechanics and age polyethism in leaf-cutter ants. Proc Biol Sci 2023; 290:20230355. [PMID: 37312549 PMCID: PMC10265030 DOI: 10.1098/rspb.2023.0355] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/05/2023] [Indexed: 06/15/2023] Open
Abstract
Many social insects display age polyethism: young workers stay inside the nest, and only older workers forage. This behavioural transition is accompanied by genetic and physiological changes, but the mechanistic origin of it remains unclear. To investigate if the mechanical demands on the musculoskeletal system effectively prevent young workers from foraging, we studied the biomechanical development of the bite apparatus in Atta vollenweideri leaf-cutter ants. Fully matured foragers generated peak in vivo bite forces of around 100 mN, more than one order of magnitude in excess of those measured for freshly eclosed callows of the same size. This change in bite force was accompanied by a sixfold increase in the volume of the mandible closer muscle, and by a substantial increase of the flexural rigidity of the head capsule, driven by a significant increase in both average thickness and indentation modulus of the head capsule cuticle. Consequently, callows lack the muscle force capacity required for leaf-cutting, and their head capsule is so compliant that large muscle forces would be likely to cause damaging deformations. On the basis of these results, we speculate that continued biomechanical development post eclosion may be a key factor underlying age polyethism, wherever foraging is associated with substantial mechanical demands.
Collapse
Affiliation(s)
- Frederik Püffel
- Department of Bioengineering, Imperial College London, London, UK
| | - Lara Meyer
- Faculty of Nature and Engineering, City University of Applied Sciences Bremen, Bremen, Germany
| | - Natalie Imirzian
- Department of Bioengineering, Imperial College London, London, UK
| | - Flavio Roces
- Department of Behavioural Physiology and Sociobiology, University of Würzburg, Würzburg, Germany
| | | | - David Labonte
- Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
5
|
Du K, Zhao X, Li Y, Wu Z, Sun W, Wang J, Jia X, Chen S, Lai S. Genome-Wide Identification and Characterization of Circular RNAs during Skeletal Muscle Development in Meat Rabbits. Animals (Basel) 2022; 12:ani12172208. [PMID: 36077928 PMCID: PMC9454498 DOI: 10.3390/ani12172208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/14/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Our knowledge of circRNAs regulating skeletal muscle development remains largely unknown in meat rabbits. Therefore, we collected the leg muscle tissues of ZIKA rabbits at three key growth stages. A combination of circRNA assembly from a circRNA-seq library and the whole-transcriptome sequencing data identified credible circRNAs in our samples. We found these circRNAs were more conserved between rabbits and humans than between rabbits and mice. A prediction of circRNA–microRNA–mRNAs networks revealed that circRNAs might be the regulators that mainly functioned in rabbits’ muscle neuron development and metabolic processes. Our work provides a catalog of circRNAs regulating skeletal muscle development at key growth stages in rabbits and might give a new insight into rabbit breeding. Abstract Skeletal muscle development plays a vital role in muscle quality and yield in meat rabbits. Circular RNAs (circRNAs) are a new type of single-stranded endogenous non-coding RNAs involved in different biological processes. However, our knowledge of circRNAs regulating skeletal muscle development remains largely unknown in meat rabbits. In this study, we collected the leg muscle tissues of ZIKA rabbits at three key growth stages. By performing whole-transcriptome sequencing, we found the sequential expression of day 0- (D0-), D35-, and D70-selective mRNAs mainly functioned in muscle development, nervous development, and immune response during skeletal muscle development, respectively. Then, a combination of circRNA assembly from a circRNA-seq library and the whole-transcriptome sequencing data identified 6845 credible circRNAs in our samples. Most circRNAs were transcribed from exons of known genes, contained few exons, and showed short length, and these circRNAs were more conserved between rabbits and humans than between rabbits and mice. The upregulated circRNAs, which were synchronously changed with host genes, primarily played roles in MAPK signaling pathways and fatty acid biosynthesis. The prediction of circRNA–microRNA–mRNAs networks revealed that circRNAs might be the regulators that mainly functioned in rabbits’ muscle neuron development and metabolic processes. Our work provides a catalog of circRNAs regulating skeletal muscle development at key growth stages in rabbits and might give a new insight into rabbit breeding.
Collapse
|
6
|
Gunderson JT, Peppriell AE, Krout IN, Vorojeikina D, Rand MD. Neuroligin-1 Is a Mediator of Methylmercury Neuromuscular Toxicity. Toxicol Sci 2021; 184:236-251. [PMID: 34546366 DOI: 10.1093/toxsci/kfab114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Methylmercury (MeHg) is a developmental toxicant capable of eliciting neurocognitive and neuromuscular deficits in children with in utero exposure. Previous research in Drosophila melanogaster uncovered that developmental MeHg exposure simultaneously targets the developing musculature and innervating motor neuron in the embryo, along with identifying Drosophila neuroligin 1 (nlg1) as a gene associated with developmental MeHg sensitivity. Nlg1 and its transsynaptic partner neurexin 1 (Nrx1) are critical for axonal arborization and NMJ maturation. We investigated the effects of MeHg exposure on indirect flight muscle (IFM) morphogenesis, innervation, and function via flight assays and monitored the expression of NMJ-associated genes to characterize the role of Nlg1 mediating the neuromuscular toxicity of MeHg. Developmental MeHg exposure reduced the innervation of the IFMs, which corresponded with reduced flight ability. In addition, nlg1 expression was selectively reduced during early metamorphosis, whereas a subsequent increase was observed in other NMJ-associated genes, including nrx1, in late metamorphosis. Developmental MeHg exposure also resulted in persistent reduced expression of most nlg and nrx genes during the first 11 days of adulthood. Transgenic modulation of nlg1 and nrx1 revealed that developing muscle is particularly sensitive to nlg1 levels, especially during the 20-36-h window of metamorphosis with reduced nlg1 expression resulting in adult flight deficits. Muscle-specific overexpression of nlg1 partially rescued MeHg-induced deficits in eclosion and flight. We identified Nlg1 as a muscle-specific, NMJ structural component that can mediate MeHg neuromuscular toxicity resulting from early life exposure.
Collapse
Affiliation(s)
- Jakob T Gunderson
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Ashley E Peppriell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Ian N Krout
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| |
Collapse
|
7
|
Development of the indirect flight muscles of Aedes aegypti, a main arbovirus vector. BMC DEVELOPMENTAL BIOLOGY 2021; 21:11. [PMID: 34445959 PMCID: PMC8394598 DOI: 10.1186/s12861-021-00242-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 08/08/2021] [Indexed: 11/22/2022]
Abstract
Background Flying is an essential function for mosquitoes, required for mating and, in the case of females, to get a blood meal and consequently function as a vector. Flight depends on the action of the indirect flight muscles (IFMs), which power the wings beat. No description of the development of IFMs in mosquitoes, including Aedes aegypti, is available.
Methods A. aegypti thoraces of larvae 3 and larvae 4 (L3 and L4) instars were analyzed using histochemistry and bright field microscopy. IFM primordia from L3 and L4 and IFMs from pupal and adult stages were dissected and processed to detect F-actin labelling with phalloidin-rhodamine or TRITC, or to immunodetection of myosin and tubulin using specific antibodies, these samples were analyzed by confocal microscopy. Other samples were studied using transmission electron microscopy. Results At L3–L4, IFM primordia for dorsal-longitudinal muscles (DLM) and dorsal–ventral muscles (DVM) were identified in the expected locations in the thoracic region: three primordia per hemithorax corresponding to DLM with anterior to posterior orientation were present. Other three primordia per hemithorax, corresponding to DVM, had lateral position and dorsal to ventral orientation. During L3 to L4 myoblast fusion led to syncytial myotubes formation, followed by myotendon junctions (MTJ) creation, myofibrils assembly and sarcomere maturation. The formation of Z-discs and M-line during sarcomere maturation was observed in pupal stage and, the structure reached in teneral insects a classical myosin thick, and actin thin filaments arranged in a hexagonal lattice structure. Conclusions A general description of A. aegypti IFM development is presented, from the myoblast fusion at L3 to form myotubes, to sarcomere maturation at adult stage. Several differences during IFM development were observed between A. aegypti (Nematoceran) and Drosophila melanogaster (Brachyceran) and, similitudes with Chironomus sp. were observed as this insect is a Nematoceran, which is taxonomically closer to A. aegypti and share the same number of larval stages. Supplementary Information The online version contains supplementary material available at 10.1186/s12861-021-00242-8.
Collapse
|
8
|
Kinold JC, Brenner M, Aberle H. Misregulation of Drosophila Sidestep Leads to Uncontrolled Wiring of the Adult Neuromuscular System and Severe Locomotion Defects. Front Neural Circuits 2021; 15:658791. [PMID: 34149366 PMCID: PMC8209334 DOI: 10.3389/fncir.2021.658791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/10/2021] [Indexed: 11/29/2022] Open
Abstract
Holometabolic organisms undergo extensive remodelling of their neuromuscular system during metamorphosis. Relatively, little is known whether or not the embryonic guidance of molecules and axonal growth mechanisms are re-activated for the innervation of a very different set of adult muscles. Here, we show that the axonal attractant Sidestep (Side) is re-expressed during Drosophila metamorphosis and is indispensable for neuromuscular wiring. Mutations in side cause severe innervation defects in all legs. Neuromuscular junctions (NMJs) show a reduced density or are completely absent at multi-fibre muscles. Misinnervation strongly impedes, but does not completely abolish motor behaviours, including walking, flying, or grooming. Overexpression of Side in developing muscles induces similar innervation defects; for example, at indirect flight muscles, it causes flightlessness. Since muscle-specific overexpression of Side is unlikely to affect the central circuits, the resulting phenotypes seem to correlate with faulty muscle wiring. We further show that mutations in beaten path Ia (beat), a receptor for Side, results in similar weaker adult innervation and locomotion phenotypes, indicating that embryonic guidance pathways seem to be reactivated during metamorphosis.
Collapse
Affiliation(s)
- Jaqueline C Kinold
- Department of Biology, Institute for Functional Cell Morphology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marcel Brenner
- Department of Biology, Institute for Functional Cell Morphology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hermann Aberle
- Department of Biology, Institute for Functional Cell Morphology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
9
|
Fluorescence based rapid optical volume screening system (OVSS) for interrogating multicellular organisms. Sci Rep 2021; 11:7616. [PMID: 33828140 PMCID: PMC8027194 DOI: 10.1038/s41598-021-86951-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 03/22/2021] [Indexed: 11/08/2022] Open
Abstract
Continuous monitoring of large specimens for long durations requires fast volume imaging. This is essential for understanding the processes occurring during the developmental stages of multicellular organisms. One of the key obstacles of fluorescence based prolonged monitoring and data collection is photobleaching. To capture the biological processes and simultaneously overcome the effect of bleaching, we developed single- and multi-color lightsheet based OVSS imaging technique that enables rapid screening of multiple tissues in an organism. Our approach based on OVSS imaging employs quantized step rotation of the specimen to record 2D angular data that reduces data acquisition time when compared to the existing light sheet imaging system (SPIM). A co-planar multicolor light sheet PSF is introduced to illuminate the tissues labelled with spectrally-separated fluorescent probes. The detection is carried out using a dual-channel sub-system that can simultaneously record spectrally separate volume stacks of the target organ. Arduino-based control systems were employed to automatize and control the volume data acquisition process. To illustrate the advantages of our approach, we have noninvasively imaged the Drosophila larvae and Zebrafish embryo. Dynamic studies of multiple organs (muscle and yolk-sac) in Zebrafish for a prolonged duration (5 days) were carried out to understand muscle structuring (Dystrophin, microfibers), primitive Macrophages (in yolk-sac) and inter-dependent lipid and protein-based metabolism. The volume-based study, intensity line-plots and inter-dependence ratio analysis allowed us to understand the transition from lipid-based metabolism to protein-based metabolism during early development (Pharyngula period with a critical transition time, [Formula: see text] h post-fertilization) in Zebrafish. The advantage of multicolor lightsheet illumination, fast volume scanning, simultaneous visualization of multiple organs and an order-less photobleaching makes OVSS imaging the system of choice for rapid monitoring and real-time assessment of macroscopic biological organisms with microscopic resolution.
Collapse
|
10
|
Sidisky JM, Weaver D, Hussain S, Okumus M, Caratenuto R, Babcock D. Mayday sustains trans-synaptic BMP signaling required for synaptic maintenance with age. eLife 2021; 10:e54932. [PMID: 33667157 PMCID: PMC7935490 DOI: 10.7554/elife.54932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/23/2021] [Indexed: 01/12/2023] Open
Abstract
Maintaining synaptic structure and function over time is vital for overall nervous system function and survival. The processes that underly synaptic development are well understood. However, the mechanisms responsible for sustaining synapses throughout the lifespan of an organism are poorly understood. Here, we demonstrate that a previously uncharacterized gene, CG31475, regulates synaptic maintenance in adult Drosophila NMJs. We named CG31475 mayday due to the progressive loss of flight ability and synapse architecture with age. Mayday is functionally homologous to the human protein Cab45, which sorts secretory cargo from the Trans Golgi Network (TGN). We find that Mayday is required to maintain trans-synaptic BMP signaling at adult NMJs in order to sustain proper synaptic structure and function. Finally, we show that mutations in mayday result in the loss of both presynaptic motor neurons as well as postsynaptic muscles, highlighting the importance of maintaining synaptic integrity for cell viability.
Collapse
Affiliation(s)
- Jessica M Sidisky
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Daniel Weaver
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Sarrah Hussain
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Meryem Okumus
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Russell Caratenuto
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| | - Daniel Babcock
- Department of Biological Sciences, Lehigh UniversityBethlehemUnited States
| |
Collapse
|
11
|
Ludwig JC, Trimmer BA. Metamorphosis in Insect Muscle: Insights for Engineering Muscle-Based Actuators. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:330-340. [PMID: 33012237 DOI: 10.1089/ten.teb.2020.0204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
One of the major limitations to advancing the development of soft robots is the absence of lightweight, effective soft actuators. While synthetic systems, such as pneumatics and shape memory alloys, have created important breakthroughs in soft actuation, they typically rely on large external power sources and some rigid components. Muscles provide an ideal actuator for soft constructs, as they are lightweight, deformable, biodegradable, silent, and powered by energy-dense hydrocarbons such as glucose. Vertebrate cell lines and embryonic cultures have allowed critical foundational work to this end, but progress there is limited by the difficulty of identifying individual pathways in embryonic development, and the divergence of immortal cell lines from these normal developmental programs. An alternative to culturing muscles from embryonic cells is to exploit the advantages of species with metamorphic stages. In these animals, muscles develop from a predefined pool of myoblasts with well-characterized contacts to other tissues. In addition, the endocrine triggers for development into adult muscles are often known and tractable for experimental manipulation. This is particularly true for metamorphic muscle development in holometabolous insects, which provide exciting new avenues for tissue engineering. Using insect tissues for actuator development confers additional benefits; insect muscles are more robust to varying pH, temperature, and oxygenation than are vertebrate cells. Given that biohybrid robots are likely to be used in ambient conditions and changing environments, this sort of hardiness is likely to be required for practical use. In this study, we summarize key processes and signals in metamorphic muscle development, drawing attention to those pathways that offer entry points for manipulation. By focusing on lessons learned from in vivo insect development, we propose that future culture designs will be able to use more systematic, hypothesis-driven approaches to optimizing engineered muscle. Impact statement This review summarizes our current understanding of metamorphic muscle development in insects. It provides a framework for engineering muscle-based actuators that can be used in robotic applications in a wide range of ambient conditions. The focus is on identifying key processes that might be manipulated to solve current challenges in controlling tissue development such as myoblast proliferation, myotube formation and fusion, cytoskeletal alignment, myotendinous attachment and full differentiation. An important goal is to gather findings that cross disciplinary boundaries and to promote the development of better bioactuators for nonclinical applications.
Collapse
|
12
|
Gunderson JT, Peppriell AE, Vorojeikina D, Rand MD. Tissue-specific Nrf2 signaling protects against methylmercury toxicity in Drosophila neuromuscular development. Arch Toxicol 2020; 94:4007-4022. [PMID: 32816092 DOI: 10.1007/s00204-020-02879-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
Methylmercury (MeHg) can elicit cognitive and motor deficits due to its developmental neuro- and myotoxic properties. While previous work has demonstrated that Nrf2 antioxidant signaling protects from MeHg toxicity, in vivo tissue-specific studies are lacking. In Drosophila, MeHg exposure shows greatest developmental toxicity in the pupal stage resulting in failed eclosion (emergence of adults) and an accompanying 'myosphere' phenotype in indirect flight muscles (IFMs). To delineate tissue-specific contributions to MeHg-induced motor deficits, we investigated the potential of Nrf2 signaling in either muscles or neurons to moderate MeHg toxicity. Larva were exposed to various concentrations of MeHg (0-20 µM in food) in combination with genetic modulation of the Nrf2 homolog cap-n-collar C (CncC), or its negative regulator Keap1. Eclosion behavior was evaluated in parallel with the morphology of two muscle groups, the thoracic IFMs and the abdominal dorsal internal oblique muscles (DIOMs). CncC signaling activity was reported with an antioxidant response element construct (ARE-GFP). We observed that DIOMs are distinguished by elevated endogenous ARE-GFP expression, which is only transiently seen in the IFMs. Dose-dependent MeHg reductions in eclosion behavior parallel formation of myospheres in the DIOMs and IFMs, while also increasing ARE-GFP expression in the DIOMs. Modulating CncC signaling via muscle-specific Keap1 knockdown and upregulation gives a rescue and exacerbation, respectively, of MeHg effects on eclosion and myospheres. Interestingly, muscle-specific CncC upregulation and knockdown both induce lethality. In contrast, neuron-specific upregulation of CncC, as well as Keap1 knockdown, rescued MeHg effects on eclosion and myospheres. Our findings indicate that enhanced CncC signaling localized to either muscles or neurons is sufficient to rescue muscle development and neuromuscular function from a MeHg insult. Additionally, there may be distinct roles for CncC signaling in myo-morphogenesis.
Collapse
Affiliation(s)
- Jakob T Gunderson
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Ashley E Peppriell
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Daria Vorojeikina
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
13
|
Chaturvedi D, Prabhakar S, Aggarwal A, Atreya KB, VijayRaghavan K. Adult Drosophila muscle morphometry through microCT reveals dynamics during ageing. Open Biol 2019; 9:190087. [PMID: 31238820 PMCID: PMC6597753 DOI: 10.1098/rsob.190087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Indirect flight muscles (IFMs) in adult Drosophila provide the key power stroke for wing beating. They also serve as a valuable model for studying muscle development. An age-dependent decline in Drosophila free flight has been documented, but its relation to gross muscle structure has not yet been explored satisfactorily. Such analyses are impeded by conventional histological preparations and imaging techniques that limit exact morphometry of flight muscles. In this study, we employ microCT scanning on a tissue preparation that retains muscle morphology under homeostatic conditions. Focusing on a subset of IFMs called the dorsal longitudinal muscles (DLMs), we find that DLM volumes increase with age, partially due to the increased separation between myofibrillar fascicles, in a sex-dependent manner. We have uncovered and quantified asymmetry in the size of these muscles on either side of the longitudinal midline. Measurements of this resolution and scale make substantive studies that test the connection between form and function possible. We also demonstrate the application of this method to other insect species making it a valuable tool for histological analysis of insect biodiversity.
Collapse
Affiliation(s)
- Dhananjay Chaturvedi
- 1 National Center for Biological Sciences, TIFR , GKVK Campus, Bellary Road, Bengaluru 560065 , India
| | - Sunil Prabhakar
- 2 microCT and EM Facility, National Center for Biological Sciences, TIFR , GKVK Campus, Bellary Road, Bengaluru 560065 , India
| | - Aman Aggarwal
- 1 National Center for Biological Sciences, TIFR , GKVK Campus, Bellary Road, Bengaluru 560065 , India.,3 Manipal Academy of Higher Education , Manipal, Karnataka 576104 , India
| | - Krishan B Atreya
- 1 National Center for Biological Sciences, TIFR , GKVK Campus, Bellary Road, Bengaluru 560065 , India
| | - K VijayRaghavan
- 1 National Center for Biological Sciences, TIFR , GKVK Campus, Bellary Road, Bengaluru 560065 , India
| |
Collapse
|
14
|
Prince LM, Rand MD. Methylmercury exposure causes a persistent inhibition of myogenin expression and C2C12 myoblast differentiation. Toxicology 2018; 393:113-122. [PMID: 29104120 PMCID: PMC5757876 DOI: 10.1016/j.tox.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/03/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
Methylmercury (MeHg) is a ubiquitous environmental toxicant, best known for its selective targeting of the developing nervous system. MeHg exposure has been shown to cause motor deficits such as impaired gait and coordination, muscle weakness, and muscle atrophy, which have been associated with disruption of motor neurons. However, recent studies have suggested that muscle may also be a target of MeHg toxicity, both in the context of developmental myogenic events and of low-level chronic exposures affecting muscle wasting in aging. We therefore investigated the effects of MeHg on myotube formation, using the C2C12 mouse myoblast model. We found that MeHg inhibits both differentiation and fusion, in a concentration-dependent manner. Furthermore, MeHg specifically and persistently inhibits myogenin (MyoG), a transcription factor involved in myocyte differentiation, within the first six hours of exposure. MeHg-induced reduction in MyoG expression is contemporaneous with a reduction of a number of factors involved in mitochondrial biogenesis and mtDNA transcription and translation, which may implicate a role for mitochondria in mediating MeHg-induced change in the differentiation program. Unexpectedly, inhibition of myoblast differentiation with MeHg parallels inhibition of Notch receptor signaling. Our research establishes muscle cell differentiation as a target for MeHg toxicity, which may contribute to the underlying etiology of motor deficits with MeHg toxicity.
Collapse
Affiliation(s)
- Lisa M Prince
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Department of Environmental Medicine, Rochester, NY, 14642, USA.
| |
Collapse
|
15
|
Prince LM, Rand MD. Notch Target Gene E(spl)mδ Is a Mediator of Methylmercury-Induced Myotoxicity in Drosophila. Front Genet 2018; 8:233. [PMID: 29379520 PMCID: PMC5775289 DOI: 10.3389/fgene.2017.00233] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/22/2017] [Indexed: 01/09/2023] Open
Abstract
Methylmercury (MeHg) is a ubiquitous environmental contaminant and neurotoxicant that has long been known to cause a variety of motor deficits. These motor deficits have primarily been attributed to MeHg targeting of developing neurons and induction of oxidative stress and calcium dysregulation. Few studies have looked at how MeHg may be affecting fundamental signaling mechanisms in development, particularly in developing muscle. Studies in Drosophila recently revealed that MeHg perturbs embryonic muscle formation and upregulates Notch target genes, reflected predominantly by expression of the downstream transcriptional repressor Enhancer of Split mdelta [E(spl)mδ]. An E(spl)mδ reporter gene shows expression primarily in the myogenic domain, and both MeHg exposure and genetic upregulation of E(spl)mδ can disrupt embryonic muscle development. Here, we tested the hypothesis that developing muscle is targeted by MeHg via upregulation of E(spl)mδ using genetic modulation of E(spl)mδ expression in combination with MeHg exposure in developing flies. Developmental MeHg exposure causes a decreased rate of eclosion that parallels gross disruption of indirect flight muscle (IFM) development. An increase in E(spl) expression across the pupal stages, with preferential E(spl)mδ upregulation occurring at early (p5) stages, is also observed. E(spl)mδ overexpression in myogenic lineages under the Mef2 promoter was seen to phenocopy eclosion and IFM effects of developmental MeHg exposure; whereas reduced expression of E(spl)mδ shows rescue of eclosion and IFM morphology effects of MeHg exposure. No effects were seen on eclosion with E(spl)mδ overexpression in neural and gut tissues. Our data indicate that muscle development is a target for MeHg and that E(spl)mδ is a muscle-specific mediator of this myotoxicity. This research advances our knowledge of the target pathways that mediate susceptibility to MeHg toxicity, as well as a potential muscle development-specific role for E(spl)mδ.
Collapse
Affiliation(s)
- Lisa M Prince
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
| | - Matthew D Rand
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
| |
Collapse
|
16
|
Jiang Q, Bao C, Yang Y, Liu A, Liu F, Huang H, Ye H. Transcriptome profiling of claw muscle of the mud crab (Scylla paramamosain) at different fattening stages. PLoS One 2017; 12:e0188067. [PMID: 29141033 PMCID: PMC5687733 DOI: 10.1371/journal.pone.0188067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 10/31/2017] [Indexed: 01/29/2023] Open
Abstract
In crustaceans, muscle growth and development is complicated, and to date substantial knowledge gaps exist. In this study, the claw muscle, hepatopancreas and nervous tissue of the mud crab (Scylla paramamosain) were collected at three fattening stages for sequence by the Illumina sequencing. A total of 127.87 Gb clean data with no less than 3.94 Gb generated for each sample and the cycleQ30 percentages were more than 86.13% for all samples. De Bruijn assembly of these clean data produced 94,853 unigenes, thereinto, 50,059 unigenes were found in claw muscle. A total of 121 differentially expressed genes (DEGs) were revealed in claw muscle from the three fattening stages with a Padj value < 0.01, including 63 genes with annotation. Functional annotation and enrichment analysis showed that the DEGs clusters represented the predominant gene catalog with roles in biochemical processes (glycolysis, phosphorylation and regulation of transcription), molecular function (ATP binding, 6-phosphofructokinase activity, and sequence-specific DNA binding) and cellular component (6-phosphofructokinase complex, plasma membrane, and integral component of membrane). qRT-PCR was employed to further validate certain DEGs. Single nucleotide polymorphism (SNP) analysis obtained 159,322, 125,963 and 166,279 potential SNPs from the muscle transcriptome at stage B, stage C and stage D, respectively. In addition, there were sixteen neuropeptide transcripts being predicted in the claw muscle. The present study provides a comprehensive transcriptome of claw muscle of S. paramamosain during fattening, providing a basis for screening the functional genes that may affect muscle growth of S. paramamosain.
Collapse
Affiliation(s)
- Qingling Jiang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Chenchang Bao
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ya’nan Yang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - An Liu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fang Liu
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Huiyang Huang
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Haihui Ye
- College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Collaborative Innovation Center for Development and Utilization of Marine Biological Resources, Xiamen, China
- * E-mail:
| |
Collapse
|
17
|
Venkatesh H, Monje M. Neuronal Activity in Ontogeny and Oncology. Trends Cancer 2017; 3:89-112. [PMID: 28718448 DOI: 10.1016/j.trecan.2016.12.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 01/06/2023]
Abstract
The nervous system plays a central role in regulating the stem cell niche in many organs, and thereby pivotally modulates development, homeostasis, and plasticity. A similarly powerful role for neural regulation of the cancer microenvironment is emerging. Neurons promote the growth of cancers of the brain, skin, prostate, pancreas, and stomach. Parallel mechanisms shared in development and cancer suggest that neural modulation of the tumor microenvironment may prove a universal theme, although the mechanistic details of such modulation remain to be discovered for many malignancies. We review here what is known about the influences of active neurons on stem cell and cancer microenvironments across a broad range of tissues, and we discuss emerging principles of neural regulation of development and cancer.
Collapse
Affiliation(s)
- Humsa Venkatesh
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA; Cancer Biology Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle Monje
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
18
|
Rai M, Katti P, Nongthomba U. Spatio-temporal coordination of cell cycle exit, fusion and differentiation of adult muscle precursors by Drosophila Erect wing (Ewg). Mech Dev 2016; 141:109-118. [DOI: 10.1016/j.mod.2016.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/16/2016] [Accepted: 03/25/2016] [Indexed: 12/12/2022]
|
19
|
Schulman VK, Dobi KC, Baylies MK. Morphogenesis of the somatic musculature in Drosophila melanogaster. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:313-34. [PMID: 25758712 DOI: 10.1002/wdev.180] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 01/28/2015] [Accepted: 01/30/2015] [Indexed: 12/22/2022]
Abstract
In Drosophila melanogaster, the somatic muscle system is first formed during embryogenesis, giving rise to the larval musculature. Later during metamorphosis, this system is destroyed and replaced by an entirely new set of muscles in the adult fly. Proper formation of the larval and adult muscles is critical for basic survival functions such as hatching and crawling (in the larva), walking and flying (in the adult), and feeding (at both larval and adult stages). Myogenesis, from mononucleated muscle precursor cells to multinucleated functional muscles, is driven by a number of cellular processes that have begun to be mechanistically defined. Once the mesodermal cells destined for the myogenic lineage have been specified, individual myoblasts fuse together iteratively to form syncytial myofibers. Combining cytoplasmic contents demands a level of intracellular reorganization that, most notably, leads to redistribution of the myonuclei to maximize internuclear distance. Signaling from extending myofibers induces terminal tendon cell differentiation in the ectoderm, which results in secure muscle-tendon attachments that are critical for muscle contraction. Simultaneously, muscles become innervated and undergo sarcomerogenesis to establish the contractile apparatus that will facilitate movement. The cellular mechanisms governing these morphogenetic events share numerous parallels to mammalian development, and the basic unit of all muscle, the myofiber, is conserved from flies to mammals. Thus, studies of Drosophila myogenesis and comparisons to muscle development in other systems highlight conserved regulatory programs of biomedical relevance to general muscle biology and studies of muscle disease.
Collapse
Affiliation(s)
- Victoria K Schulman
- Cell and Developmental Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA.,Program in Developmental Biology, Sloan Kettering Institute, New York, NY, USA
| | - Krista C Dobi
- Program in Developmental Biology, Sloan Kettering Institute, New York, NY, USA
| | - Mary K Baylies
- Cell and Developmental Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA.,Program in Developmental Biology, Sloan Kettering Institute, New York, NY, USA
| |
Collapse
|
20
|
Ryglewski S, Kilo L, Duch C. Sequential acquisition of cacophony calcium currents, sodium channels and voltage-dependent potassium currents affects spike shape and dendrite growth during postembryonic maturation of an identified Drosophila motoneuron. Eur J Neurosci 2014; 39:1572-85. [PMID: 24620836 DOI: 10.1111/ejn.12517] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 01/16/2014] [Accepted: 01/20/2014] [Indexed: 11/28/2022]
Abstract
During metamorphosis the CNS undergoes profound changes to accommodate the switch from larval to adult behaviors. In Drosophila and other holometabolous insects, adult neurons differentiate either from respecified larval neurons, newly born neurons, or are born embryonically but remain developmentally arrested until differentiation during pupal life. This study addresses the latter in the identified Drosophila flight motoneuron 5. In situ patch-clamp recordings, intracellular dye fills and immunocytochemistry address the interplay between dendritic shape, excitability and ionic current development. During pupal life, changes in excitability and spike shape correspond to a stereotyped, progressive appearance of voltage-gated ion channels. High-voltage-activated calcium current is the first current to appear at pupal stage P4, prior to the onset of dendrite growth. This is followed by voltage-gated sodium as well as transient potassium channel expression, when first dendrites grow, and sodium-dependent action potentials can be evoked by somatic current injection. Sustained potassium current appears later than transient potassium current. During the early stages of rapid dendritic growth, sodium-dependent action potentials are broadened by a calcium component. Narrowing of spike shape coincides with sequential increases in transient and sustained potassium currents during stages when dendritic growth ceases. Targeted RNAi knockdown of pupal calcium current significantly reduces dendritic growth. These data indicate that the stereotyped sequential acquisition of different voltage-gated ion channels affects spike shape and excitability such that activity-dependent calcium influx serves as a partner of genetic programs during critical stages of motoneuron dendrite growth.
Collapse
Affiliation(s)
- Stefanie Ryglewski
- Institute of Zoology III - Neurobiology, University of Mainz, D-55128, Mainz, Germany
| | | | | |
Collapse
|
21
|
Piccirillo R, Demontis F, Perrimon N, Goldberg AL. Mechanisms of muscle growth and atrophy in mammals and Drosophila. Dev Dyn 2014; 243:201-15. [PMID: 24038488 PMCID: PMC3980484 DOI: 10.1002/dvdy.24036] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/01/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The loss of skeletal muscle mass (atrophy) that accompanies disuse and systemic diseases is highly debilitating. Although the pathogenesis of this condition has been primarily studied in mammals, Drosophila is emerging as an attractive system to investigate some of the mechanisms involved in muscle growth and atrophy. RESULTS In this review, we highlight the outstanding unsolved questions that may benefit from a combination of studies in both flies and mammals. In particular, we discuss how different environmental stimuli and signaling pathways influence muscle mass and strength and how a variety of disease states can cause muscle wasting. CONCLUSIONS Studies in Drosophila and mammals should help identify molecular targets for the treatment of muscle wasting in humans.
Collapse
Affiliation(s)
- Rosanna Piccirillo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Department of Oncology, IRCCS - Mario Negri Institute for Pharmacological Research, Milano, Italy
| | - Fabio Demontis
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Department of Developmental Neurobiology, Division of Developmental Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
22
|
Abstract
Many aspects of skeletal muscle biology are remarkably similar between mammals and tiny insects, and experimental models of mice and flies (Drosophila) provide powerful tools to understand factors controlling the growth, maintenance, degeneration (atrophy and necrosis), and regeneration of normal and diseased muscles, with potential applications to the human condition. This review compares the limb muscles of mice and the indirect flight muscles of flies, with respect to the mechanisms of adult myofiber formation, homeostasis, atrophy, hypertrophy, and the response to muscle degeneration, with some comment on myogenic precursor cells and common gene regulatory pathways. There is a striking similarity between the species for events related to muscle atrophy and hypertrophy, without contribution of any myoblast fusion. Since the flight muscles of adult flies lack a population of reserve myogenic cells (equivalent to satellite cells), this indicates that such cells are not required for maintenance of normal muscle function. However, since satellite cells are essential in postnatal mammals for myogenesis and regeneration in response to myofiber necrosis, the extent to which such regeneration might be possible in flight muscles of adult flies remains unclear. Common cellular and molecular pathways for both species are outlined related to neuromuscular disorders and to age-related loss of skeletal muscle mass and function (sarcopenia). The commonality of events related to skeletal muscles in these disparate species (with vast differences in size, growth duration, longevity, and muscle activities) emphasizes the combined value and power of these experimental animal models.
Collapse
|
23
|
Herrera-Valdez MA, McKiernan EC, Berger SD, Ryglewski S, Duch C, Crook S. Relating ion channel expression, bifurcation structure, and diverse firing patterns in a model of an identified motor neuron. J Comput Neurosci 2013; 34:211-29. [PMID: 22878689 PMCID: PMC6595220 DOI: 10.1007/s10827-012-0416-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 06/19/2012] [Accepted: 07/09/2012] [Indexed: 01/12/2023]
Abstract
Neurons show diverse firing patterns. Even neurons belonging to a single chemical or morphological class, or the same identified neuron, can display different types of electrical activity. For example, motor neuron MN5, which innervates a flight muscle of adult Drosophila, can show distinct firing patterns under the same recording conditions. We developed a two-dimensional biophysical model and show that a core complement of just two voltage-gated channels is sufficient to generate firing pattern diversity. We propose Shab and DmNa v to be two candidate genes that could encode these core currents, and find that changes in Shab channel expression in the model can reproduce activity resembling the main firing patterns observed in MN5 recordings. We use bifurcation analysis to describe the different transitions between rest and spiking states that result from variations in Shab channel expression, exposing a connection between ion channel expression, bifurcation structure, and firing patterns in models of membrane potential dynamics.
Collapse
Affiliation(s)
- Marco A Herrera-Valdez
- Institute of Interdisciplinary Research, University of Puerto Rico at Cayey, 205 Antonio R. Barcelo Ave., Cayey, PR 00736, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Modzelewska K, Lauritzen A, Hasenoeder S, Brown L, Georgiou J, Moghal N. Neurons refine the Caenorhabditis elegans body plan by directing axial patterning by Wnts. PLoS Biol 2013; 11:e1001465. [PMID: 23319891 PMCID: PMC3539944 DOI: 10.1371/journal.pbio.1001465] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 11/16/2012] [Indexed: 12/14/2022] Open
Abstract
Metazoans display remarkable conservation of gene families, including growth factors, yet somehow these genes are used in different ways to generate tremendous morphological diversity. While variations in the magnitude and spatio-temporal aspects of signaling by a growth factor can generate different body patterns, how these signaling variations are organized and coordinated during development is unclear. Basic body plans are organized by the end of gastrulation and are refined as limbs, organs, and nervous systems co-develop. Despite their proximity to developing tissues, neurons are primarily thought to act after development, on behavior. Here, we show that in Caenorhabditis elegans, the axonal projections of neurons regulate tissue progenitor responses to Wnts so that certain organs develop with the correct morphology at the right axial positions. We find that foreshortening of the posteriorly directed axons of the two canal-associated neurons (CANs) disrupts mid-body vulval morphology, and produces ectopic vulval tissue in the posterior epidermis, in a Wnt-dependent manner. We also provide evidence that suggests that the posterior CAN axons modulate the location and strength of Wnt signaling along the anterior-posterior axis by employing a Ror family Wnt receptor to bind posteriorly derived Wnts, and hence, refine their distributions. Surprisingly, despite high levels of Ror expression in many other cells, these cells cannot substitute for the CAN axons in patterning the epidermis, nor can cells expressing a secreted Wnt inhibitor, SFRP-1. Thus, unmyelinated axon tracts are critical for patterning the C. elegans body. Our findings suggest that the evolution of neurons not only improved metazoans by increasing behavioral complexity, but also by expanding the diversity of developmental patterns generated by growth factors such as Wnts.
Collapse
Affiliation(s)
- Katarzyna Modzelewska
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Amara Lauritzen
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Stefan Hasenoeder
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Louise Brown
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - John Georgiou
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Nadeem Moghal
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Tang JM, Yuan J, Li Q, Wang JN, Kong X, Zheng F, Zhang L, Chen L, Guo LY, Huang YH, Yang JY, Chen SY. Acetylcholine induces mesenchymal stem cell migration via Ca2+ /PKC/ERK1/2 signal pathway. J Cell Biochem 2012; 113:2704-13. [PMID: 22441978 DOI: 10.1002/jcb.24148] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Acetylcholine (ACh) plays an important role in neural and non-neural function, but its role in mesenchymal stem cell (MSC) migration remains to be determined. In the present study, we have found that ACh induces MSC migration via muscarinic acetylcholine receptors (mAChRs). Among several mAChRs, MSCs express mAChR subtype 1 (m1AChR). ACh induces MSC migration via interaction with mAChR1. MEK1/2 inhibitor PD98059 blocks ERK1/2 phosphorylation while partially inhibiting the ACh-induced MSC migration. InsP3Rs inhibitor 2-APB that inhibits MAPK/ERK phosphorylation completely blocks ACh-mediated MSC migration. Interestingly, intracellular Ca(2+) ATPase-specific inhibitor thapsigargin also completely blocks ACh-induced MSC migration through the depletion of intracellular Ca(2+) storage. PKCα or PKCβ inhibitor or their siRNAs only partially inhibit ACh-induced MSC migration, but PKC-ζ siRNA completely inhibits ACh-induced MSC migration via blocking ERK1/2 phosphorylation. These results indicate that ACh induces MSC migration via Ca(2+), PKC, and ERK1/2 signal pathways.
Collapse
Affiliation(s)
- Jun-Ming Tang
- Institute of Clinical Medicine and Department of Cardiology, Renmin Hospital, Hubei University of Medicine, Hubei 442000, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Brierley DJ, Rathore K, VijayRaghavan K, Williams DW. Developmental origins and architecture of Drosophila leg motoneurons. J Comp Neurol 2012; 520:1629-49. [PMID: 22120935 DOI: 10.1002/cne.23003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Motoneurons are key points of convergence within motor networks, acting as the "output channels" that directly control sets of muscles to maintain posture and generate movement. Here we use genetic mosaic techniques to reveal the origins and architecture of the leg motoneurons of Drosophila. We show that a small number of leg motoneurons are born in the embryo but most are generated during larval life. These postembryonic leg motoneurons are produced by five neuroblasts per hemineuromere, and each lineage generates stereotyped lineage-specific projection patterns. Two of these postembryonic neuroblasts generate solely motoneurons that are the bulk of the leg motoneurons. Within the largest lineage, lineage 15, we see distinct birth-order differences in projection patterns. A comparison of the central projections of leg motoneurons and the muscles they innervate reveals a stereotyped architecture and the existence of a myotopic map. Timeline analysis of axonal outgrowth reveals that leg motoneurons reach their sites of terminal arborization in the leg at the time when their dendrites are elaborating their subtype-specific shapes. Our findings provide a comprehensive description of the origin, development, and architecture of leg motoneurons that will aid future studies exploring the link between the assembly and organization of connectivity within the leg motor system of Drosophila.
Collapse
Affiliation(s)
- D J Brierley
- MRC Centre for Developmental Neurobiology, King's College London, London, SE1 1UL, UK
| | | | | | | |
Collapse
|
27
|
Abstract
Cell ablation is a powerful tool in the study of eukaryotic developmental biology. The selective removal of cells by ablation may provide much information about their origin, fate, or function in the developing organism. Laser-based techniques have an advantage over genetic or chemical ablation methods in that the operations can be performed in essentially any cell pattern and at any time in development. This protocol describes the methods needed to target and ablate specific cells of interest in Drosophila embryos with lasers.
Collapse
|
28
|
Sweeney ST, Hidalgo A, de Belle JS, Keshishian H. Setup for functional cell ablation with lasers: coupling of a laser to a microscope. Cold Spring Harb Protoc 2012; 2012:726-32. [PMID: 22661442 DOI: 10.1101/pdb.ip068387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The selective removal of cells by ablation is a powerful tool in the study of eukaryotic developmental biology, providing much information about their origin, fate, or function in the developing organism. In Drosophila, three main methods have been used to ablate cells: chemical, genetic, and laser ablation. Each method has its own applicability with regard to developmental stage and the cells to be ablated, and its own limitations. The primary advantage of laser-based ablation is the flexibility provided by the method: The operations can be performed in any cell pattern and at any time in development. Laser-based techniques permit manipulation of structures within cells, even to the molecular level. They can also be used for gene activation. However, laser ablation can be expensive, labor-intensive, and time-consuming. Although live cells can be difficult to image in Drosophila embryos, the use of vital fluorescent imaging methods has made laser-mediated cell manipulation methods more appealing; the methods are relatively straightforward. This article provides the information necessary for setting up and using a laser microscope for lasesr ablation studies.
Collapse
|
29
|
Salvi SS, Kumar RP, Ramachandra NB, Sparrow JC, Nongthomba U. Mutations in Drosophila myosin rod cause defects in myofibril assembly. J Mol Biol 2012; 419:22-40. [PMID: 22370558 DOI: 10.1016/j.jmb.2012.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 01/17/2012] [Accepted: 02/17/2012] [Indexed: 11/15/2022]
Abstract
The roles of myosin during muscle contraction are well studied, but how different domains of this protein are involved in myofibril assembly in vivo is far less understood. The indirect flight muscles (IFMs) of Drosophila melanogaster provide a good model for understanding muscle development and function in vivo. We show that two missense mutations in the rod region of the myosin heavy-chain gene, Mhc, give rise to IFM defects and abnormal myofibrils. These defects likely result from thick filament abnormalities that manifest during early sarcomere development or later by hypercontraction. The thick filament defects are accompanied by marked reduction in accumulation of flightin, a myosin binding protein, and its phosphorylated forms, which are required to stabilise thick filaments. We investigated with purified rod fragments whether the mutations affect the coiled-coil structure, rod aggregate size or rod stability. No significant changes in these parameters were detected, except for rod thermodynamic stability in one mutation. Molecular dynamics simulations suggest that these mutations may produce localised rod instabilities. We conclude that the aberrant myofibrils are a result of thick filament defects, but that these in vivo effects cannot be detected in vitro using the biophysical techniques employed. The in vivo investigation of these mutant phenotypes in IFM development and function provides a useful platform for studying myosin rod and thick filament formation generically, with application to the aetiology of human myosin rod myopathies.
Collapse
Affiliation(s)
- Sheetal S Salvi
- Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560 012, India
| | | | | | | | | |
Collapse
|
30
|
Ren H, Li L, Su H, Xu L, Wei C, Zhang L, Li H, Liu W, Du L. Histological and transcriptome-wide level characteristics of fetal myofiber hyperplasia during the second half of gestation in Texel and Ujumqin sheep. BMC Genomics 2011; 12:411. [PMID: 21838923 PMCID: PMC3173453 DOI: 10.1186/1471-2164-12-411] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Accepted: 08/14/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Whether myofibers increase with a pulsed-wave mode at particular developmental stages or whether they augment evenly across developmental stages in large mammals is unclear. Additionally, the molecular mechanisms of myostatin in myofiber hyperplasia at the fetal stage in sheep remain unknown. Using the first specialized transcriptome-wide sheep oligo DNA microarray and histological methods, we investigated the gene expression profile and histological characteristics of developing fetal ovine longissimus muscle in Texel sheep (high muscle and low fat), as a myostatin model of natural mutation, and Ujumqin sheep (low muscle and high fat). Fetal skeletal muscles were sampled at 70, 85, 100, 120, and 135 d of gestation. RESULTS Myofiber number increased sharply with a pulsed-wave mode at certain developmental stages but was not augmented evenly across developmental stages in fetal sheep. The surges in myofiber hyperplasia occurred at 85 and 120 d in Texel sheep, whereas a unique proliferative surge appeared at 100 d in Ujumqin sheep. Analysis of the microarray demonstrated that immune and hematological systems' development and function, lipid metabolism, and cell communication were the biological functions that were most differentially expressed between Texel and Ujumqin sheep during muscle development. Pathways associated with myogenesis and the proliferation of myoblasts, such as calcium signaling, chemokine (C-X-C motif) receptor 4 signaling, and vascular endothelial growth factor signaling, were affected significantly at specific fetal stages, which underpinned fetal myofiber hyperplasia and postnatal muscle hypertrophy. Moreover, we identified some differentially expressed genes between the two breeds that could be potential myostatin targets for further investigation. CONCLUSIONS Proliferation of myofibers proceeded in a pulsed-wave mode at particular fetal stages in the sheep. The myostatin mutation changed the gene expression pattern in skeletal muscle at a transcriptome-wide level, resulting in variation in myofiber phenotype between Texel and Ujumqin sheep during the second half of gestation. Our findings provide a novel and dynamic description of the effect of myostatin on skeletal muscle development, which contributes to understanding the biology of muscle development in large mammals.
Collapse
Affiliation(s)
- Hangxing Ren
- National Center for Molecular Genetics and Breeding of Animal, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Coming of age in an ant colony: cephalic muscle maturation accompanies behavioral development in Pheidole dentata. Naturwissenschaften 2011; 98:783-93. [DOI: 10.1007/s00114-011-0828-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 06/13/2011] [Accepted: 07/13/2011] [Indexed: 10/18/2022]
|
32
|
Sen A, Yokokura T, Kankel MW, Dimlich DN, Manent J, Sanyal S, Artavanis-Tsakonas S. Modeling spinal muscular atrophy in Drosophila links Smn to FGF signaling. ACTA ACUST UNITED AC 2011; 192:481-95. [PMID: 21300852 PMCID: PMC3101100 DOI: 10.1083/jcb.201004016] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
FGF signaling in neurons is regulated by Survival Motor Neuron, a component of a complex that regulates snRNP biogenesis and FGF receptor expression. Spinal muscular atrophy (SMA), a devastating neurodegenerative disorder characterized by motor neuron loss and muscle atrophy, has been linked to mutations in the Survival Motor Neuron (SMN) gene. Based on an SMA model we developed in Drosophila, which displays features that are analogous to the human pathology and vertebrate SMA models, we functionally linked the fibroblast growth factor (FGF) signaling pathway to the Drosophila homologue of SMN, Smn. Here, we characterize this relationship and demonstrate that Smn activity regulates the expression of FGF signaling components and thus FGF signaling. Furthermore, we show that alterations in FGF signaling activity are able to modify the neuromuscular junction defects caused by loss of Smn function and that muscle-specific activation of FGF is sufficient to rescue Smn-associated abnormalities.
Collapse
Affiliation(s)
- Anindya Sen
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Kapelnikov A, Rivlin PK, Hoy RR, Heifetz Y. Tissue remodeling: a mating-induced differentiation program for the Drosophila oviduct. BMC DEVELOPMENTAL BIOLOGY 2008; 8:114. [PMID: 19063748 PMCID: PMC2636784 DOI: 10.1186/1471-213x-8-114] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Accepted: 12/08/2008] [Indexed: 12/11/2022]
Abstract
BACKGROUND In both vertebrates and invertebrates, the oviduct is an epithelial tube surrounded by visceral muscles that serves as a conduit for gamete transport between the ovary and uterus. While Drosophila is a model system for tubular organ development, few studies have addressed the development of the fly's oviduct. Recent studies in Drosophila have identified mating-responsive genes and proteins whose levels in the oviduct are altered by mating. Since many of these molecules (e.g. Muscle LIM protein 84B, Coracle, Neuroglian) have known roles in the differentiation of muscle and epithelia of other organs, mating may trigger similar differentiation events in the oviduct. This led us to hypothesize that mating mediates the last stages of oviduct differentiation in which organ-specific specializations arise. RESULTS Using electron- and confocal-microscopy we identified tissue-wide post-mating changes in the oviduct including differentiation of cellular junctions, remodeling of extracellular matrix, increased myofibril formation, and increased innervation. Analysis of once- and twice-mated females reveals that some mating-responsive proteins respond only to the first mating, while others respond to both matings. CONCLUSION We uncovered ultrastructural changes in the mated oviduct that are consistent with the roles that mating-responsive proteins play in muscle and epithelial differentiation elsewhere. This suggests that mating triggers the late differentiation of the oviduct. Furthermore, we suggest that mating-responsive proteins that respond only to the first mating are involved in the final maturation of the oviduct while proteins that remain responsive to later matings are also involved in maintenance and ongoing function of the oviduct. Taken together, our results establish the oviduct as an attractive system to address mechanisms that regulate the late stages of differentiation and maintenance of a tubular organ.
Collapse
Affiliation(s)
- Anat Kapelnikov
- Department of Entomology, The Hebrew University, Rehovot, Israel.
| | | | | | | |
Collapse
|
34
|
Soldier-specific modification of the mandibular motor neurons in termites. PLoS One 2008; 3:e2617. [PMID: 18612458 PMCID: PMC2435624 DOI: 10.1371/journal.pone.0002617] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Accepted: 06/05/2008] [Indexed: 11/19/2022] Open
Abstract
Social insects exhibit a variety of caste-specific behavioral tendencies that constitute the basis of division of labor within the colony. In termites, the soldier caste display distinctive defense behaviors, such as aggressively attacking enemies with well-developed mandibles, while the other castes retreat into the colony without exhibiting any aggressive response. It is thus likely that some form of soldier-specific neuronal modification exists in termites. In this study, the authors compared the brain (cerebral ganglion) and the suboesophageal ganglion (SOG) of soldiers and pseudergates (workers) in the damp-wood termite, Hodotermopsis sjostedti. The size of the SOG was significantly larger in soldiers than in pseudergates, but no difference in brain size was apparent between castes. Furthermore, mandibular nerves were thicker in soldiers than in pseudergates. Retrograde staining revealed that the somata sizes of the mandibular motor neurons (MdMNs) in soldiers were more than twice as large as those of pseudergates. The enlargement of MdMNs was also observed in individuals treated with a juvenile hormone analogue (JHA), indicating that MdMNs become enlarged in response to juvenile hormone (JH) action during soldier differentiation. This enlargement is likely to have two functions: a behavioral function in which soldier termites will be able to defend more effectively through relatively faster and stronger mandibular movements, and a developmental function that associates with the development of soldier-specific mandibular muscle morphogenesis in termite head. The soldier-specific enlargement of mandibular motor neurons was observed in all examined species in five termite families that have different mechanisms of defense, suggesting that such neuronal modification was already present in the common ancestor of termites and is significant for soldier function.
Collapse
|
35
|
Founder cells regulate fiber number but not fiber formation during adult myogenesis in Drosophila. Dev Biol 2008; 321:123-40. [PMID: 18616937 DOI: 10.1016/j.ydbio.2008.06.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2007] [Revised: 05/22/2008] [Accepted: 06/04/2008] [Indexed: 11/22/2022]
Abstract
During insect myogenesis, myoblasts are organized into a pre-pattern by specialized organizer cells. In the Drosophila embryo, these cells have been termed founder cells and play important roles in specifying muscle identity and in serving as targets for myoblast fusion. A group of adult muscles, the dorsal longitudinal (flight) muscles, DLMs, is patterned by persistent larval scaffolds; the second set, the dorso-ventral muscles, DVMs is patterned by mono-nucleate founder cells (FCs) that are much larger than the surrounding myoblasts. Both types of organizer cells express Dumbfounded, which is known to regulate fusion during embryonic myogenesis. The role of DVM founder cells as well as the DLM scaffolds was tested in genetic ablation studies using the UAS/Gal4 system of targeted transgene expression. In both cases, removal of organizer cells prior to fusion, causes formation of supernumerary fibers, suggesting that cells in the myoblast pool have the capacity to initiate fiber formation, which is normally inhibited by the organizers. In addition to the large DVM FCs, some (smaller) cells in the myoblast pool also express Dumbfounded. We propose that these cells are responsible for seeding supernumerary fibers, when DVM FCs are eliminated prior to fusion. When these cells are also eliminated, myogenesis fails to occur. In the second set of studies, targeted expression of constitutively active Ras(V12) also resulted in the appearance of supernumerary fibers. In this case, the original DVM FCs are present, suggesting alterations in cell fate. Taken together, these data suggest that DVM myoblasts are able to respond to cues other than the original founder cell, to initiate fusion and fiber formation. Thus, the role of the large DVM founder cells is to generate the correct number of fibers, but they are not required for fiber formation itself. We also present evidence that the DVM FCs may arise from the leg imaginal disc.
Collapse
|
36
|
Rao GN, Kulkarni SS, Koushika SP, Rau KR. In vivo nanosecond laser axotomy: cavitation dynamics and vesicle transport. OPTICS EXPRESS 2008; 16:9884-9894. [PMID: 18575558 DOI: 10.1364/oe.16.009884] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Nanosecond laser pulses (lambda = 355 nm) were used to cut mechanosensory neurons in Caenorhabditis elegans and motorneurons in Drosophila melanogaster larvae. A pulse energy range of 0.8-1.2 microJ and < 20 pulses in single shot mode were sufficient to generate axonal cuts. Viability post-surgery was >95% for C. elegans and 60% for Drosophila. Cavitation bubble dynamics generated due to laser-induced plasma formation were observed in vivo by time-resolved imaging in both organisms. Bubble oscillations were severely damped in vivo and cavitation dynamics were complete within 100 ns in C. elegans and 800 ns in Drosophila. We report the use of this system to study axonal transport for the first time and discuss advantages of nanosecond lasers compared to femtosecond sources for such procedures.
Collapse
Affiliation(s)
- G Nageswara Rao
- National Centre for Biological Sciences-TIFR, Bangalore 560065, India
| | | | | | | |
Collapse
|
37
|
Hebbar S, Hall RE, Demski SA, Subramanian A, Fernandes JJ. The adult abdominal neuromuscular junction of Drosophila: a model for synaptic plasticity. ACTA ACUST UNITED AC 2006; 66:1140-55. [PMID: 16838368 DOI: 10.1002/neu.20279] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
During its life cycle, Drosophila makes two sets of neuromuscular junctions (NMJs), embryonic/larval and adult, which serve distinct stage-specific functions. During metamorphosis, the larval NMJs are restructured to give rise to their adult counterparts, a process that is integrated into the overall remodeling of the nervous system. The NMJs of the prothoracic muscles and the mesothoracic dorsal longitudinal (flight) muscles have been previously described. Given the diversity and complexity of adult muscle groups, we set out to examine the less complex abdominal muscles. The large bouton sizes of these NMJs are particularly advantageous for easy visualization. Specifically, we have characterized morphological attributes of the ventral abdominal NMJ and show that an embryonic motor neuron identity gene, dHb9, is expressed at these adult junctions. We quantified bouton numbers and size and examined the localization of synaptic markers. We have also examined the formation of boutons during metamorphosis and examined the localization of presynaptic markers at these stages. To test the usefulness of the ventral abdominal NMJs as a model system, we characterized the effects of altering electrical activity and the levels of the cell adhesion molecule, FasciclinII (FasII). We show that both manipulations affect NMJ formation and that the effects are specific as they can be rescued genetically. Our results indicate that both activity and FasII affect development at the adult abdominal NMJ in ways that are distinct from their larval and adult thoracic counterparts
Collapse
Affiliation(s)
- Sarita Hebbar
- Department of Zoology, Miami University, Oxford, Ohio 45056, USA
| | | | | | | | | |
Collapse
|
38
|
Fernandes JJ, Atreya KB, Desai KM, Hall RE, Patel MD, Desai AA, Benham AE, Mable JL, Straessle JL. A dominant negative form of Rac1 affects myogenesis of adult thoracic muscles in Drosophila. Dev Biol 2005; 285:11-27. [PMID: 16125691 DOI: 10.1016/j.ydbio.2005.05.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2004] [Revised: 04/25/2005] [Accepted: 05/25/2005] [Indexed: 11/29/2022]
Abstract
Blocking Rac1 function in precursors of the indirect flight muscle of Drosophila severely disrupts muscle formation. The DLM fibers that develop using larval scaffolds are reduced in number and fiber size, while the DVMs, which develop using founder cells, are mostly absent. These adult muscle phenotypes are in part due to a reduced myoblast pool present at the third larval instar. BrDU labeling studies indicated that this is primarily due to a reduction in proliferation. In addition, DVM myoblasts display altered morphology and are unable to segregate into primordia. This defect precedes the evident block in fusion. We also show that the recently described DVM founder cells can be labeled with 22C10 and beta-3 tubulin, and that they are present under conditions of dominant negative Rac1(N17) expression. Despite the presence of founder cells, DVM fiber formation is rarely observed. Although DLM myoblasts are able to segregate around their larval scaffolds, the pace of fusion is reduced and consequently there is a delay in DLM fiber formation. Thus, in addition to its well-established role in fusion, Rac1 is also involved in the regulation of myoblast proliferation and segregation during adult myogenesis. These are two new roles for Rac1 in Drosophila.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Cell Fusion
- Cell Proliferation
- Drosophila/cytology
- Drosophila/genetics
- Drosophila/growth & development
- Drosophila/physiology
- Drosophila Proteins/deficiency
- Drosophila Proteins/genetics
- Drosophila Proteins/physiology
- Female
- Flight, Animal
- Gene Expression Regulation, Developmental
- Gene Targeting
- Genes, Dominant
- Genes, Insect
- Larva/cytology
- Larva/growth & development
- Male
- Muscle Development/genetics
- Muscle, Skeletal/cytology
- Muscle, Skeletal/growth & development
- Myoblasts, Skeletal/cytology
- Phenotype
- Wings, Animal/cytology
- Wings, Animal/growth & development
- rac GTP-Binding Proteins/deficiency
- rac GTP-Binding Proteins/genetics
- rac GTP-Binding Proteins/physiology
Collapse
Affiliation(s)
- Joyce J Fernandes
- Center for Neuroscience, 250 Pearson Hall, Miami University, Oxford, OH 45056, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hebbar S, Fernandes JJ. A role for Fas II in the stabilization of motor neuron branches during pruning in Drosophila. Dev Biol 2005; 285:185-99. [PMID: 16055111 DOI: 10.1016/j.ydbio.2005.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 06/16/2005] [Accepted: 06/16/2005] [Indexed: 11/18/2022]
Abstract
During insect metamorphosis, the nervous system is extensively remodeled resulting in the development of new circuits that will execute adult-specific behaviors. The peripheral remodeling seen during development of innervation to the Dorsal Longitudinal (flight) Muscle (DLM) in Drosophila involves an initial retraction of larval neuromuscular junctions followed by adult-specific branch outgrowth. Subsequently, a phase of pruning occurs during which motor neuron branches are pruned back to reveal the stereotypic pattern of multiple contact points (or arbors) along the length of each DLM fiber. In this study, we show that the cell adhesion molecule, Fasciclin II (Fas II), is important for generating the stereotypic pattern. In Fas II hypomorphs, the number of contact points is increased, and the phenotype is rescued by targeted expression of Fas II in either synaptic partner. Arbor development has three distinct phases: outgrowth and elaboration, pruning and stabilization, and expansion of stabilized arbors. Fas II is expressed during the first two phases. A subset of branches is labeled during the elaboration phase, which is likely to initiate a stabilization pathway allowing branches to survive the pruning phase. However, since not all Fas II positive branches are retained, we propose that it primes branches for stabilization. Our data suggest that Fas II functions to restrict branch length and arbor expanse.
Collapse
Affiliation(s)
- Sarita Hebbar
- Department of Zoology, 250 Pearson Hall, Miami University, Oxford, OH 45056, USA
| | | |
Collapse
|
40
|
Knittel LM, Kent KS. Remodeling of an identified motoneuron during metamorphosis: hormonal influences on the growth of dendrites and axon terminals. ACTA ACUST UNITED AC 2005; 63:106-25. [PMID: 15702475 DOI: 10.1002/neu.20121] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
During metamorphosis of the tobacco hawkmoth Manduca sexta, the femoral depressor motoneuron (FeDe MN) undergoes remodeling of its dendrites and motor terminals. Previous studies have established that remodeling of MNs during metamorphosis is mediated by the same hormones that control metamorphosis: the ecdysteroids and juvenile hormone (JH). During the pupal stage, the ecdysteroids promote adult-specific growth of MNs in the absence of JH, but JH or its synthetic mimics can interfere with ecdysteroid-mediated growth if applied during early sensitive periods. Hence, the application of a JH mimic (JHM) either systemically or locally to a target muscle has been used to distinguish those aspects of motor-terminal remodeling that are controlled by ecdysteroid action on the CNS from those that are influenced by ecdysteroid action on the peripheral targets. Here, we have extended the analysis of central and peripheral hormonal influences on MN remodeling by injecting JHM locally into the CNS thus altering the hormonal environment of the FeDe MN soma without altering the hormonal environment of its target muscle. Our results demonstrate that adult dendritic growth and motor-terminal growth can be experimentally uncoupled, suggesting that each is regulated independently. JHM application to the CNS perturbed dendritic growth, but had no measurable impact on motor-terminal growth. Peripheral actions of ecdysteroids, therefore, appear sufficient to promote the development of adult-specific motor terminals but not the development of an adult-specific dendritic arbor.
Collapse
Affiliation(s)
- Laura M Knittel
- Department of Integrative Biosciences, School of Dentistry, Oregon Health & Science University, Portland, 97239, USA
| | | |
Collapse
|
41
|
Wu Y, Kawasaki F, Ordway RW. Properties of short-term synaptic depression at larval neuromuscular synapses in wild-type and temperature-sensitive paralytic mutants of Drosophila. J Neurophysiol 2005; 93:2396-405. [PMID: 15845998 DOI: 10.1152/jn.01108.2004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The larval neuromuscular synapse of Drosophila serves as an important model for genetic and molecular analysis of synaptic development and function. Further functional characterization of this synapse, as well as adult neuromuscular synapses, will greatly enhance the impact of this model system on our understanding of synaptic transmission. Here we describe a form of short-term synaptic depression observed at larval, but not adult, neuromuscular synapses and explore the underlying mechanisms. Larval neuromuscular synapses exhibited a form of short-term depression that was strongly dependent on stimulation frequency over a narrow range of low frequencies (0.1-1 Hz). This form of synaptic depression, referred to here as low-frequency short-term depression (LF-STD), results from an activity-dependent reduction in neurotransmitter release. However, in contrast to the predictions of depletion models, the degree of depression was independent of the initial level of neurotransmitter release over a range of extracellular calcium concentrations. This conclusion was confirmed in two temperature-sensitive (TS) paralytic mutants, cacophony and shibire, which exhibit reduced neurotransmitter release resulting from conditional disruption of presynaptic calcium channels and dynamin, respectively. Higher stimulation frequencies (40 or 60 Hz) produced two components of depression that appeared to include LF-STD as well as a more conventional component of short-term depression. These findings reveal novel properties of short-term synaptic depression and suggest that complementary genetic analysis of larval and adult neuromuscular synapses will further define the in vivo mechanisms of neurotransmitter release and short-term synaptic plasticity.
Collapse
Affiliation(s)
- Ying Wu
- Department of Biology and Center for Cellular and Molecular Neuroscience, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | |
Collapse
|
42
|
Fernandes JJ, Keshishian H. Motoneurons regulate myoblast proliferation and patterning in Drosophila. Dev Biol 2005; 277:493-505. [PMID: 15617689 DOI: 10.1016/j.ydbio.2004.09.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2003] [Revised: 06/15/2004] [Accepted: 09/16/2004] [Indexed: 10/26/2022]
Abstract
Motoneurons directly influence the differentiation of muscle fibers, regulating features such as muscle fiber type and receptor development. Less well understood is whether motoneurons direct earlier events, such as the patterning of the musculature. In Drosophila, the denervation of indirect flight muscles results in a diminished myoblast population and smaller or missing muscle fibers. We have examined whether the neuron-dependent control of myoblast number is due to regulation of cell division, motoneuron-dependent apoptosis, or nerve-dependent localization and migration of myoblasts. We found that denervation resulted in a reduced rate of cell division, as revealed by BrDU incorporation. There was no change in the frequency of apoptotic myoblasts following denervation. Using time lapse imaging of GFP-expressing myoblasts in vivo in pupae, we observed that despite denervation, the migration and localization of myoblasts remained unchanged. In addition to reducing myoblast proliferation, denervation also altered the segregation of myoblasts into the de novo arising dorso-ventral muscles (DVMs). To address this effect on muscle patterning, we examined the expression of the founder-cell marker Dumbfounded/Kirre (Duf) in imaginal pioneer cells. We show that there is a strong correspondence between cells that express Dumbfounded/Kirre and the number of DVM fibers, consistent with a role for these cells in establishing adult muscles. In the absence of innervation the Duf-positive cells are no longer detected, and muscle patterning is severely disrupted. Our results support a model where specialized founder cells prefigure the adult muscle fibers under the control of the nervous system.
Collapse
Affiliation(s)
- Joyce J Fernandes
- Zoology Department, 250 Pearson Hall, Miami University, Oxford, OH 45056, USA
| | | |
Collapse
|
43
|
Consoulas C, Levine RB, Restifo LL. The steroid hormone-regulated geneBroad Complex is required for dendritic growth of motoneurons during metamorphosis ofDrosophila. J Comp Neurol 2005; 485:321-37. [PMID: 15803508 DOI: 10.1002/cne.20499] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Dendrites are subject to subtle modifications as well as extensive remodeling during the assembly and maturation of neural circuits in a wide variety of organisms. During metamorphosis, Drosophila flight motoneurons MN1-MN4 undergo dendritic regression, followed by regrowth, whereas MN5 differentiates de novo (Consoulas et al. [2002] J. Neurosci. 22:4906-4917). Many cellular changes during metamorphosis are triggered and orchestrated by the steroid hormone 20-hydroxyecdysone, which initiates a cascade of coordinated gene expression. Broad Complex (BRC), a primary response gene in the ecdysone cascade, encodes a family of transcription factors (BRC-Z1-Z4) that are essential for metamorphic reorganization of the central nervous system (CNS). Using neuron-filling techniques that reveal cellular morphology with very high resolution, we tested the hypothesis that BRC is required for metamorphic development of MN1-MN5. Through a combination of loss-of-function mutant analyses, genetic mapping, and transgenic rescue experiments, we found that 2Bc function, mediated by BRC-Z3, is required selectively for motoneuron dendritic regrowth (MN1-MN4) and de novo outgrowth (MN5), as well as for soma expansion of MN5. In contrast, larval development and dendritic regression of MN1-MN4 are BRC-independent. Surprisingly, BRC proteins are not expressed in the motoneurons, suggesting that BRC-Z3 exerts its effect in a non-cell-autonomous manner. The 2Bc mutants display no gross defects in overall thoracic CNS structure, or in peripheral structures such as target muscles or sensory neurons. Candidates for mediating the effect of BRC-Z3 on dendritic growth of MN1-MN5 include their synaptic inputs and non-neuronal CNS cells that interact with them through direct contact or diffusible factors.
Collapse
Affiliation(s)
- Christos Consoulas
- ARL Division of Neurobiology, University of Arizona, Tucson, Arizona 85721, USA.
| | | | | |
Collapse
|
44
|
Hebbar S, Fernandes JJ. Pruning of motor neuron branches establishes the DLM innervation pattern in Drosophila. ACTA ACUST UNITED AC 2004; 60:499-516. [PMID: 15307154 DOI: 10.1002/neu.20031] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During the Drosophila life-cycle two sets of neuromuscular junctions are generated: the embryonic/larval NMJs develop during the first half, followed by the period of metamorphosis during which the adult counterpart is generated. Development of the adult innervation pattern is preceded by a withdrawal of larval NMJs, which occurs at the onset of metamorphosis, and is followed by adult-specific motor neuron outgrowth to innervate the newly developing adult fibers. Establishment of the adult innervation pattern occurs in the context of a broader restructuring of the nervous system, which results in the development of neural circuits that are necessary to carry out behaviors specific to the adult. In this article, we follow development of the dorsal longitudinal muscle (DLM) innervation pattern through metamorphosis. We find that the initial period of motor neuron elaboration is followed by a phase of extensive pruning resulting in a threefold reduction of neuromuscular contacts. This event establishes the adult pattern of second order branching. Subsequent higher order branching from the second order "contact" points generates the characteristic multiterminal innervation pattern of the DLMs. Boutons begin to appear after the pruning phase, and are much smaller than their larval counterparts. Additionally, we demonstrate that the DLM innervation is altered in the hyperexcitable double mutant, ether a go-go Shaker, and that the phenotype is suppressed by the hypoexcitable mutant, nap(ts1). Our results demonstrate that electrical activity regulates the patterning of DLM innervation during metamorphosis.
Collapse
Affiliation(s)
- Sarita Hebbar
- Department of Zoology, Miami University, Oxford, Ohio 45056, USA
| | | |
Collapse
|
45
|
Bernard F, Lalouette A, Gullaud M, Jeantet AY, Cossard R, Zider A, Ferveur JF, Silber J. Control of apterous by vestigial drives indirect flight muscle development in Drosophila. Dev Biol 2003; 260:391-403. [PMID: 12921740 DOI: 10.1016/s0012-1606(03)00255-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Drosophila thoracic muscles are comprised of both direct flight muscles (DFMs) and indirect flight muscles (IFMs). The IFMs can be further subdivided into dorsolongitudinal muscles (DLMs) and dorsoventral muscles (DVMs). The correct patterning of each category of muscles requires the coordination of specific executive regulatory programs. DFM development requires key regulatory genes such as cut (ct) and apterous (ap), whereas IFM development requires vestigial (vg). Using a new vg(null) mutant, we report that a total absence of vg leads to DLM degeneration through an apoptotic process and to a total absence of DVMs in the adult. We show that vg and scalloped (sd), the only known VG transcriptional coactivator, are coexpressed during IFM development. Moreover, we observed an ectopic expression of ct and ap, two markers of DFM development, in developing IFMs of vg(null) pupae. In addition, in vg(null) adult flies, degenerating DLMs express twist (twi) ectopically. We provide evidence that ap ectopic expression can induce per se ectopic twi expression and muscle degeneration. All these data seem to indicate that, in the absence of vg, the IFM developmental program switches into the DFM developmental program. Moreover, we were able to rescue the muscle phenotype of vg(null) flies by using the activity of ap promoter to drive VG expression. Thus, vg appears to be a key regulatory gene of IFM development.
Collapse
Affiliation(s)
- F Bernard
- Institut Jacques Monod, UMR 7592 CNRS, Université Paris 7, Tour 43, 2, place Jussieu, 75251 Paris Cedex 05, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Duch C, Mentel T. Stage-specific activity patterns affect motoneuron axonal retraction and outgrowth during the metamorphosis of Manduca sexta. Eur J Neurosci 2003; 17:945-62. [PMID: 12653971 DOI: 10.1046/j.1460-9568.2003.02523.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During the metamorphosis of holometabolous insects, most larval muscles and sensory neurons are replaced by new adult elements, whereas most motoneurons persist and are remodelled to serve new adult functions. In Manduca sexta, the formation of the anlagen of the adult dorsal longitudinal flight muscle (DLM) is characterized by retraction of axonal terminals and dendrites of persisting larval motoneurons, partial target muscle degeneration and myoblast accumulation during late larval life. Most of these structural changes have been attributed to hormonal control, not only because ecdysteroids govern metamorphosis, but also because motoneurons express ecdysteroid receptors and experimental manipulations of ecdysteroid titres perturb normal development. To test whether activity-dependent mechanisms also came into play, chronic extracellular recordings were conducted in vivo from the five future DLM motoneurons throughout the last 3 days of larval life. Motoneuron activity is regulated developmentally. The types of motoneurons recruited, the number of motor spikes and the duration of bursts change in a stereotypical fashion during different stages, indicating an internal control of motor activity. A characteristic cessation in the activity of the five future DLM motoneurons coincides in time with the retraction of their dendrites and their terminal arborizations, whereas their activation during ecdysis coincides with the onset of new outgrowth. Inducing advanced activity by stimulating the motoneurons selectively with ecdysis-like patterns results in significant outgrowth of their terminal arborizations. Therefore, steroids might act in concert with activity-dependent mechanisms during the postembryonic modifications of neuromuscular systems.
Collapse
Affiliation(s)
- C Duch
- Institute of Biology, Neurobiology, Free University Berlin, Koenigin-Luise Str 28-30, 14195 Berlin, Germany.
| | | |
Collapse
|
47
|
Patel K, Christ B, Stockdale FE. Control of muscle size during embryonic, fetal, and adult life. Results Probl Cell Differ 2003; 38:163-86. [PMID: 12132394 DOI: 10.1007/978-3-540-45686-5_8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Ketan Patel
- Department of Veterinary Basic Sciences, Royal Veterinary College, Royal College Street, London NW1 OTU, UK
| | | | | |
Collapse
|
48
|
Libersat F, Duch C. Morphometric analysis of dendritic remodeling in an identified motoneuron during postembryonic development. J Comp Neurol 2002; 450:153-66. [PMID: 12124760 DOI: 10.1002/cne.10318] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A detailed quantitative description of modifications in neuronal architecture is an important prerequisite to investigate the signals underlying behaviorally relevant changes in neuronal shape. Extensive morphological remodeling of neurons occurs during the metamorphosis of holometabolous insects, such as Manduca sexta, in which new adult behaviors develop postembryonically. In this study, a morphometric analysis of the structural changes of an identified Manduca motoneuron, MN5, was conducted by sampling its metric parameters at different developmental stages. The remodeling of MN5 is divided into three main phases. The regression of most larval dendrites (1) is followed by the formation of dendritic growth-cones (2), and subsequently, adult dendrite formation (3). In contrast, the cell body and link segment surface increase during dendritic regression and regrowth, indicating that different cell compartments receive different signals, or respond differently to the same signal. During dendritic growth-cone formation, the growth of the cell body and the link segment are arrested. Sholl and branch frequency analysis suggest two different modes of dendritic growth. During a first growth-cone-dependent phase, new branch formation occurs at all dendrites. The maximum path length of the major dendritic tree changes little, whereas branch order increases from 20 to 45. Changes in total dendritic length are correlated with strong changes in the number of nodes but with minor changes in the average dendritic segment length, indicating a mode of growth similar to that induced by steroid hormone application to cultured motoneurons. The second phase is growth-cone-independent, and branching is limited to high order dendrites.
Collapse
Affiliation(s)
- Frederic Libersat
- Zlotowski Center for Neuroscience and Department of Life Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel.
| | | |
Collapse
|
49
|
Bayline RJ, Duch C, Levine RB. Nerve-muscle interactions regulate motor terminal growth and myoblast distribution during muscle development. Dev Biol 2001; 231:348-63. [PMID: 11237464 DOI: 10.1006/dbio.2001.0158] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interactions between motoneurons and muscles influence many aspects of neuromuscular development in all animals. These interactions can be readily investigated during adult muscle development in holometabolous insects. In this study, the development of the dorsolongitudinal flight muscle (DLM) and its innervation is investigated in the moth, Manduca sexta, to address the specificity of neuromuscular interactions. The DLM develops from an anlage containing both regressed larval template fibers and imaginal myoblasts. In the adult, each fiber bundle (DLM1-5) is innervated by a single motoneuron (MN1-MN5), with the dorsal-most fiber bundle (DLM5) innervated by a mesothoracic motoneuron (MN5). The DLM failed to develop following complete denervation because myoblasts failed to accumulate in the DLM anlage. After lesioning MN1-4, MN5 retained its specificity for the DLM5 region of the anlage and failed to rescue DLM1-4. Thus specific innervation of the DLM fiber bundles does not depend on interactions among motoneurons. Myoblast accumulation, but not myonuclear proliferation, increased around the MN5 terminals, producing a hypertrophied adult DLM5. Therefore, motoneurons compete for uncommitted myoblasts. MN5 terminals subsequently grew more rapidly over the hypertrophied DLM5 anlage, indicating that motoneuron terminal expansion is regulated by the size of the target muscle anlage.
Collapse
Affiliation(s)
- R J Bayline
- Division of Neurobiology, University of Arizona, Room 611, Gould Simpson Building, Tucson, Arizona 85721, USA.
| | | | | |
Collapse
|
50
|
Jacobs K, Todman MG, Allen MJ, Davies JA, Bacon JP. Synaptogenesis in the giant-fibre system of Drosophila: interaction of the giant fibre and its major motorneuronal target. Development 2000; 127:5203-12. [PMID: 11060245 DOI: 10.1242/dev.127.23.5203] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The tergotrochanteral (jump) motorneuron is a major synaptic target of the Giant Fibre in Drosophila. These two neurons are major components of the fly's Giant-Fibre escape system. Our previous work has described the development of the Giant Fibre in early metamorphosis and the involvement of the shaking-B locus in the formation of its electrical synapses. In the present study, we have investigated the development of the tergotrochanteral motorneuron and its electrical synapses by transforming Drosophila with a Gal4 fusion construct containing sequences largely upstream of, but including, the shaking-B(lethal) promoter. This construct drives reporter gene expression in the tergotrochanteral motorneuron and some other neurons. Expression of green fluorescent protein in the motorneuron allows visualization of its cell body and its subsequent intracellular staining with Lucifer Yellow. These preparations provide high-resolution data on motorneuron morphogenesis during the first half of pupal development. Dye-coupling reveals onset of gap-junction formation between the tergotrochanteral motorneuron and other neurons of the Giant-Fibre System. The medial dendrite of the tergotrochanteral motorneuron becomes dye-coupled to the peripheral synapsing interneurons between 28 and 32 hours after puparium formation. Dye-coupling between tergotrochanteral motorneuron and Giant Fibre is first seen at 42 hours after puparium formation. All dye coupling is abolished in a shaking-B(neural) mutant. To investigate any interactions between the Giant Fibre and the tergotroachanteral motorneuron, we arrested the growth of the motorneuron's medial neurite by targeted expression of a constitutively active form of Dcdc42. This results in the Giant Fibre remaining stranded at the midline, unable to make its characteristic bend. We conclude that Giant Fibre morphogenesis normally relies on fasciculation with its major motorneuronal target.
Collapse
Affiliation(s)
- K Jacobs
- Sussex Centre for Neuroscience, School of Biological Sciences, University of Sussex, Brighton BN1 9QG, UK
| | | | | | | | | |
Collapse
|