1
|
Li T, Shi W, Ho MS, Zhang YQ. A Pvr-AP-1-Mmp1 signaling pathway is activated in astrocytes upon traumatic brain injury. eLife 2024; 12:RP87258. [PMID: 39480704 PMCID: PMC11527428 DOI: 10.7554/elife.87258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Abstract
Traumatic brain injury (TBI) caused by external mechanical forces is a major health burden worldwide, but the underlying mechanism in glia remains largely unclear. We report herein that Drosophila adults exhibit a defective blood-brain barrier, elevated innate immune responses, and astrocyte swelling upon consecutive strikes with a high-impact trauma device. RNA sequencing (RNA-seq) analysis of these astrocytes revealed upregulated expression of genes encoding PDGF and VEGF receptor-related (Pvr, a receptor tyrosine kinase), adaptor protein complex 1 (AP-1, a transcription factor complex of the c-Jun N-terminal kinase pathway) composed of Jun-related antigen (Jra) and kayak (kay), and matrix metalloproteinase 1 (Mmp1) following TBI. Interestingly, Pvr is both required and sufficient for AP-1 and Mmp1 upregulation, while knockdown of AP-1 expression in the background of Pvr overexpression in astrocytes rescued Mmp1 upregulation upon TBI, indicating that Pvr acts as the upstream receptor for the downstream AP-1-Mmp1 transduction. Moreover, dynamin-associated endocytosis was found to be an important regulatory step in downregulating Pvr signaling. Our results identify a new Pvr-AP-1-Mmp1 signaling pathway in astrocytes in response to TBI, providing potential targets for developing new therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Tingting Li
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Wenwen Shi
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| | - Margaret S Ho
- Institute of Neuroscience, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Brain Research Center, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yong Q Zhang
- Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, University of Chinese Academy of Sciences, Chinese Academy of SciencesBeijingChina
| |
Collapse
|
2
|
Alcalde Anton A, Young FJ, Melo-Flórez L, Couto A, Cross S, McMillan WO, Montgomery SH. Adult neurogenesis does not explain the extensive post-eclosion growth of Heliconius mushroom bodies. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230755. [PMID: 37885989 PMCID: PMC10598442 DOI: 10.1098/rsos.230755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023]
Abstract
Among butterflies, Heliconius have a unique behavioural profile, being the sole genus to actively feed on pollen. Heliconius learn the location of pollen resources, and have enhanced visual memories and expanded mushroom bodies, an insect learning and memory centre, relative to related genera. These structures also show extensive post-eclosion growth and developmental sensitivity to environmental conditions. However, whether this reflects plasticity in neurite growth, or an extension of neurogenesis into the adult stage, is unknown. Adult neurogenesis has been described in some Lepidoptera, and could provide one route to the increased neuron number observed in Heliconius. Here, we compare volumetric changes in the mushroom bodies of freshly eclosed and aged Heliconius erato and Dryas iulia, and estimate the number of intrinsic mushroom body neurons using a new and validated automated method to count nuclei. Despite extensive volumetric variation associated with age, our data show that neuron number is remarkably constant in both species, suggesting a lack of adult neurogenesis in the mushroom bodies. We support this conclusion with assays of mitotic cells, which reveal very low levels of post-eclosion cell division. Our analyses provide an insight into the evolution of neural plasticity, and can serve as a basis for continued exploration of the potential mechanisms behind brain development and maturation.
Collapse
Affiliation(s)
| | - Fletcher J. Young
- School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | | | - Antoine Couto
- School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
| | - Stephen Cross
- Wolfson Bioimaging Centre, University of Bristol, Bristol, UK
| | | | - Stephen H. Montgomery
- School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
- Smithsonian Tropical Research Institute, Gamboa, Panama
| |
Collapse
|
3
|
Villalobos-Cantor S, Barrett RM, Condon AF, Arreola-Bustos A, Rodriguez KM, Cohen MS, Martin I. Rapid cell type-specific nascent proteome labeling in Drosophila. eLife 2023; 12:83545. [PMID: 37092974 PMCID: PMC10125018 DOI: 10.7554/elife.83545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 04/09/2023] [Indexed: 04/25/2023] Open
Abstract
Controlled protein synthesis is required to regulate gene expression and is often carried out in a cell type-specific manner. Protein synthesis is commonly measured by labeling the nascent proteome with amino acid analogs or isotope-containing amino acids. These methods have been difficult to implement in vivo as they require lengthy amino acid replacement procedures. O-propargyl-puromycin (OPP) is a puromycin analog that incorporates into nascent polypeptide chains. Through its terminal alkyne, OPP can be conjugated to a fluorophore-azide for directly visualizing nascent protein synthesis, or to a biotin-azide for capture and identification of newly-synthesized proteins. To achieve cell type-specific OPP incorporation, we developed phenylacetyl-OPP (PhAc-OPP), a puromycin analog harboring an enzyme-labile blocking group that can be removed by penicillin G acylase (PGA). Here, we show that cell type-specific PGA expression in Drosophila can be used to achieve OPP labeling of newly-synthesized proteins in targeted cell populations within the brain. Following a brief 2 hr incubation of intact brains with PhAc-OPP, we observe robust imaging and affinity purification of OPP-labeled nascent proteins in PGA-targeted cell populations. We apply this method to show a pronounced age-related decline in neuronal protein synthesis in the fly brain, demonstrating the capability of PhAc-OPP to quantitatively capture in vivo protein synthesis states. This method, which we call POPPi (PGA-dependent OPP incorporation), should be applicable for rapidly visualizing protein synthesis and identifying nascent proteins synthesized under diverse physiological and pathological conditions with cellular specificity in vivo.
Collapse
Affiliation(s)
- Stefanny Villalobos-Cantor
- Jungers Center for Neurosciences, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Ruth M Barrett
- Jungers Center for Neurosciences, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Alec F Condon
- Jungers Center for Neurosciences, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Alicia Arreola-Bustos
- Jungers Center for Neurosciences, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Kelsie M Rodriguez
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, United States
| | - Michael S Cohen
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, United States
| | - Ian Martin
- Jungers Center for Neurosciences, Department of Neurology, Oregon Health and Science University, Portland, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, United States
- Parkinson Center of Oregon, Oregon Health and Science University, Portland, United States
| |
Collapse
|
4
|
Kundu S, Singh S. What Happens in TBI? A Wide Talk on Animal Models and Future Perspective. Curr Neuropharmacol 2023; 21:1139-1164. [PMID: 35794772 PMCID: PMC10286592 DOI: 10.2174/1570159x20666220706094248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/05/2022] [Accepted: 05/11/2022] [Indexed: 11/22/2022] Open
Abstract
Traumatic brain injury (TBI) is a global healthcare concern and a leading cause of death. The most common causes of TBI include road accidents, sports injuries, violence in warzones, and falls. TBI induces neuronal cell death independent of age, gender, and genetic background. TBI survivor patients often experience long-term behavioral changes like cognitive and emotional changes. TBI affects social activity, reducing the quality and duration of life. Over the last 40 years, several rodent models have been developed to mimic different clinical outcomes of human TBI for a better understanding of pathophysiology and to check the efficacy of drugs used for TBI. However, promising neuroprotective approaches that have been used preclinically have been found to be less beneficial in clinical trials. So, there is an urgent need to find a suitable animal model for establishing a new therapeutic intervention useful for TBI. In this review, we have demonstrated the etiology of TBI and post- TBI social life alteration, and also discussed various preclinical TBI models of rodents, zebrafish, and drosophila.
Collapse
Affiliation(s)
- Satyabrata Kundu
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shamsher Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
5
|
Hermans L, Kaynak M, Braun J, Ríos VL, Chen CL, Friedberg A, Günel S, Aymanns F, Sakar MS, Ramdya P. Microengineered devices enable long-term imaging of the ventral nerve cord in behaving adult Drosophila. Nat Commun 2022; 13:5006. [PMID: 36008386 PMCID: PMC9411199 DOI: 10.1038/s41467-022-32571-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 08/04/2022] [Indexed: 11/09/2022] Open
Abstract
The dynamics and connectivity of neural circuits continuously change on timescales ranging from milliseconds to an animal's lifetime. Therefore, to understand biological networks, minimally invasive methods are required to repeatedly record them in behaving animals. Here we describe a suite of devices that enable long-term optical recordings of the adult Drosophila melanogaster ventral nerve cord (VNC). These consist of transparent, numbered windows to replace thoracic exoskeleton, compliant implants to displace internal organs, a precision arm to assist implantation, and a hinged stage to repeatedly tether flies. To validate and illustrate our toolkit we (i) show minimal impact on animal behavior and survival, (ii) follow the degradation of chordotonal organ mechanosensory nerve terminals over weeks after leg amputation, and (iii) uncover waves of neural activity caffeine ingestion. Thus, our long-term imaging toolkit opens up the investigation of premotor and motor circuit adaptations in response to injury, drug ingestion, aging, learning, and disease.
Collapse
Affiliation(s)
- Laura Hermans
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland
- Microbiorobotic Systems Laboratory, Institute of Mechanical Engineering & Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Murat Kaynak
- Microbiorobotic Systems Laboratory, Institute of Mechanical Engineering & Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Jonas Braun
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Victor Lobato Ríos
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Chin-Lin Chen
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Adam Friedberg
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Semih Günel
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland
- Computer Vision Laboratory, EPFL, Lausanne, Switzerland
| | - Florian Aymanns
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland
| | - Mahmut Selman Sakar
- Microbiorobotic Systems Laboratory, Institute of Mechanical Engineering & Institute of Bioengineering, EPFL, Lausanne, Switzerland.
| | - Pavan Ramdya
- Neuroengineering Laboratory, Brain Mind Institute & Institute of Bioengineering, EPFL, Lausanne, Switzerland.
| |
Collapse
|
6
|
Coupe D, Bossing T. Insights into nervous system repair from the fruit fly. Neuronal Signal 2022; 6:NS20210051. [PMID: 35474685 PMCID: PMC9008705 DOI: 10.1042/ns20210051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Millions of people experience injury to the central nervous system (CNS) each year, many of whom are left permanently disabled, providing a challenging hurdle for the field of regenerative medicine. Repair of damage in the CNS occurs through a concerted effort of phagocytosis of debris, cell proliferation and differentiation to produce new neurons and glia, distal axon/dendrite degeneration, proximal axon/dendrite regeneration and axon re-enwrapment. In humans, regeneration is observed within the peripheral nervous system, while in the CNS injured axons exhibit limited ability to regenerate. This has also been described for the fruit fly Drosophila. Powerful genetic tools available in Drosophila have allowed the response to CNS insults to be probed and novel regulators with mammalian orthologs identified. The conservation of many regenerative pathways, despite considerable evolutionary separation, stresses that these signals are principal regulators and may serve as potential therapeutic targets. Here, we highlight the role of Drosophila CNS injury models in providing key insight into regenerative processes by exploring the underlying pathways that control glial and neuronal activation in response to insult, and their contribution to damage repair in the CNS.
Collapse
Affiliation(s)
- David Coupe
- Peninsula Medical School, University of Plymouth, John Bull Building, 16 Research Way, Plymouth PL6 8BU, U.K
| | - Torsten Bossing
- Peninsula Medical School, University of Plymouth, John Bull Building, 16 Research Way, Plymouth PL6 8BU, U.K
| |
Collapse
|
7
|
Aggarwal P, Thapliyal D, Sarkar S. The past and present of Drosophila models of Traumatic Brain Injury. J Neurosci Methods 2022; 371:109533. [DOI: 10.1016/j.jneumeth.2022.109533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 11/30/2022]
|
8
|
Farder-Gomes CF, Fernandes KM, Bernardes RC, Bastos DSS, Oliveira LLD, Martins GF, Serrão JE. Harmful effects of fipronil exposure on the behavior and brain of the stingless bee Partamona helleri Friese (Hymenoptera: Meliponini). THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 794:148678. [PMID: 34225147 DOI: 10.1016/j.scitotenv.2021.148678] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
Fipronil is a pesticide widely used to control agricultural and household insect pests. However, fipronil is highly toxic to non-target insects, including pollinators. In this study, we investigated the acute effects of fipronil on the behavior, brain morphology, antioxidant activity, and proteins related to signaling pathways on the brain of workers of the stingless bee Partamona helleri. The ingestion of fipronil increases both the walking distance and velocity and causes enlarged intercellular spaces in the Kenyon cells and intense vacuolization in the neuropils of the brain. Moreover, fipronil decreases the activity of catalase (CAT) and increases the activity of glutathione S-transferase (GST). However, there is no difference in superoxide dismutase (SOD) activity between the control and fipronil. Regarding immunofluorescence analysis, bees exposed to fipronil showed an increase in the number of cells positive for cleaved caspase-3 and peroxidase, but a reduction in the number of cells positive for ERK 1/2, JNK and Notch, suggesting neuron death and impaired brain function. Our results demonstrate that fipronil has harmful effects on the behavior and brain of a stingless bee, which may threaten the individuals and colonies of this pollinator.
Collapse
Affiliation(s)
| | - Kenner Morais Fernandes
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | | | - Daniel Silva Sena Bastos
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | | | - Gustavo Ferreira Martins
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| | - José Eduardo Serrão
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais 36570-900, Brazil.
| |
Collapse
|
9
|
Buhlman LM, Krishna G, Jones TB, Thomas TC. Drosophila as a model to explore secondary injury cascades after traumatic brain injury. Biomed Pharmacother 2021; 142:112079. [PMID: 34463269 PMCID: PMC8458259 DOI: 10.1016/j.biopha.2021.112079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022] Open
Abstract
Drosophilae are emerging as a valuable model to study traumatic brain injury (TBI)-induced secondary injury cascades that drive persisting neuroinflammation and neurodegenerative pathology that imposes significant risk for long-term neurological deficits. As in mammals, TBI in Drosophila triggers axonal injury, metabolic crisis, oxidative stress, and a robust innate immune response. Subsequent neurodegeneration stresses quality control systems and perpetuates an environment for neuroprotection, regeneration, and delayed cell death via highly conserved cell signaling pathways. Fly injury models continue to be developed and validated for both whole-body and head-specific injury to isolate, evaluate, and modulate these parallel pathways. In conjunction with powerful genetic tools, the ability for longitudinal evaluation, and associated neurological deficits that can be tested with established behavioral tasks, Drosophilae are an attractive model to explore secondary injury cascades and therapeutic intervention after TBI. Here, we review similarities and differences between mammalian and fly pathophysiology and highlight strategies for their use in translational neurotrauma research.
Collapse
Affiliation(s)
- Lori M Buhlman
- Biomedical Sciences Program, Midwestern University, Glendale, AZ, USA.
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA
| | - T Bucky Jones
- Department of Anatomy, Midwestern University, Glendale, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Phoenix VA Health Care System, Phoenix, AZ, USA.
| |
Collapse
|
10
|
Sieriebriennikov B, Reinberg D, Desplan C. A molecular toolkit for superorganisms. Trends Genet 2021; 37:846-859. [PMID: 34116864 PMCID: PMC8355152 DOI: 10.1016/j.tig.2021.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022]
Abstract
Social insects, such as ants, bees, wasps, and termites, draw biologists' attention due to their distinctive lifestyles. As experimental systems, they provide unique opportunities to study organismal differentiation, division of labor, longevity, and the evolution of development. Ants are particularly attractive because several ant species can be propagated in the laboratory. However, the same lifestyle that makes social insects interesting also hampers the use of molecular genetic techniques. Here, we summarize the efforts of the ant research community to surmount these hurdles and obtain novel mechanistic insight into the biology of social insects. We review current approaches and propose novel ones involving genomics, transcriptomics, chromatin and DNA methylation profiling, RNA interference (RNAi), and genome editing in ants and discuss future experimental strategies.
Collapse
Affiliation(s)
- Bogdan Sieriebriennikov
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Danny Reinberg
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Howard Hughes Medical Institute, NYU Grossman School of Medicine, New York, NY, USA.
| | - Claude Desplan
- Department of Biology, New York University, New York, NY, USA.
| |
Collapse
|
11
|
Tzouanas CN, Kim S, Badhiwala KN, Avants BW, Robinson JT. Hydra vulgaris shows stable responses to thermal stimulation despite large changes in the number of neurons. iScience 2021; 24:102490. [PMID: 34095784 PMCID: PMC8164038 DOI: 10.1016/j.isci.2021.102490] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/03/2021] [Accepted: 04/27/2021] [Indexed: 11/27/2022] Open
Abstract
Many animals that lose neural tissue to injury or disease can maintain behavioral repertoires by regenerating new neurons or reorganizing existing neural circuits. However, most neuroscience small model organisms lack this high degree of neural plasticity. We show that Hydra vulgaris can maintain stable sensory-motor behaviors despite 2-fold changes in neuron count, due to naturally occurring size variation or surgical resection. Specifically, we find that both behavioral and neural responses to rapid temperature changes are maintained following these perturbations. We further describe possible mechanisms for the observed neural activity and argue that Hydra's radial symmetry may allow it to maintain stable behaviors when changes in the numbers of neurons do not selectively eliminate any specific neuronal cell type. These results suggest that Hydra provides a powerful model for studying how animals maintain stable sensory-motor responses within dynamic neural circuits and may lead to the development of general principles for injury-tolerant neural architectures.
Collapse
Affiliation(s)
| | - Soonyoung Kim
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Krishna N. Badhiwala
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Benjamin W. Avants
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| | - Jacob T. Robinson
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Department of Electrical and Computer Engineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
12
|
Liu C, Liu J, Liu C, Zhou Q, Zhou Y, Zhang B, Saijilafu. The intrinsic axon regenerative properties of mature neurons after injury. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1-9. [PMID: 33258872 DOI: 10.1093/abbs/gmaa148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Indexed: 01/07/2023] Open
Abstract
Thousands of nerve injuries occur in the world each year. Axon regeneration is a very critical process for the restoration of the injured nervous system's function. However, the precise molecular mechanism or signaling cascades that control axon regeneration are not clearly understood, especially in mammals. Therefore, there is almost no ideal treatment method to repair the nervous system's injury until now. Mammalian axonal regeneration requires multiple signaling pathways to coordinately regulate gene expression in soma and assembly of the cytoskeleton protein in the growth cone. A better understanding of their molecular mechanisms, such as axon regeneration regulatory signaling cascades, will be helpful in developing new treatment strategies for promoting axon regeneration. In this review, we mainly focus on describing these regeneration-associated signaling cascades, which regulate axon regeneration.
Collapse
Affiliation(s)
- Chunfeng Liu
- Department of Orthopedic Surgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou 215000, China
| | - Jinlian Liu
- Department of Orthopedic Surgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou 215000, China
| | - Chaoqun Liu
- Department of Orthopedic Surgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou 215000, China
| | - Qing Zhou
- Department of Orthopedic Surgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou 215000, China
| | - Yaodong Zhou
- Department of Orthopedic Surgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University Medical School, Suzhou 215000, China
| | - Boyin Zhang
- Orthopedics Surgery Department, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Saijilafu
- Department of Orthopaedics, The First Affiliated Hospital, Orthopaedic Institute, Soochow University, Suzhou 215007, China
| |
Collapse
|
13
|
Bicker G, Stern M. Structural and Functional Plasticity in the Regenerating Olfactory System of the Migratory Locust. Front Physiol 2020; 11:608661. [PMID: 33424632 PMCID: PMC7793960 DOI: 10.3389/fphys.2020.608661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022] Open
Abstract
Regeneration after injury is accompanied by transient and lasting changes in the neuroarchitecture of the nervous system and, thus, a form of structural plasticity. In this review, we introduce the olfactory pathway of a particular insect as a convenient model to visualize neural regeneration at an anatomical level and study functional recovery at an electrophysiological level. The olfactory pathway of the locust (Locusta migratoria) is characterized by a multiglomerular innervation of the antennal lobe by olfactory receptor neurons. These olfactory afferents were axotomized by crushing the base of the antenna. The resulting degeneration and regeneration in the antennal lobe could be quantified by size measurements, dye labeling, and immunofluorescence staining of cell surface proteins implicated in axonal guidance during development. Within 3 days post lesion, the antennal lobe volume was reduced by 30% and from then onward regained size back to normal by 2 weeks post injury. The majority of regenerating olfactory receptor axons reinnervated the glomeruli of the antennal lobe. A few regenerating axons project erroneously into the mushroom body on a pathway that is normally chosen by second-order projection neurons. Based on intracellular responses of antennal lobe output neurons to odor stimulation, regenerated fibers establish functional synapses again. Following complete absence after nerve crush, responses to odor stimuli return to control level within 10–14 days. On average, regeneration of afferents, and re-established synaptic connections appear faster in younger fifth instar nymphs than in adults. The initial degeneration of olfactory receptor axons has a trans-synaptic effect on a second order brain center, leading to a transient size reduction of the mushroom body calyx. Odor-evoked oscillating field potentials, absent after nerve crush, were restored in the calyx, indicative of regenerative processes in the network architecture. We conclude that axonal regeneration in the locust olfactory system appears to be possible, precise, and fast, opening an avenue for future mechanistic studies. As a perspective of biomedical importance, the current evidence for nitric oxide/cGMP signaling as positive regulator of axon regeneration in connectives of the ventral nerve cord is considered in light of particular regeneration studies in vertebrate central nervous systems.
Collapse
Affiliation(s)
- Gerd Bicker
- Division of Cell Biology, Institute of Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Michael Stern
- Division of Cell Biology, Institute of Physiology and Cell Biology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
14
|
Bolus H, Crocker K, Boekhoff-Falk G, Chtarbanova S. Modeling Neurodegenerative Disorders in Drosophila melanogaster. Int J Mol Sci 2020; 21:E3055. [PMID: 32357532 PMCID: PMC7246467 DOI: 10.3390/ijms21093055] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Drosophila melanogaster provides a powerful genetic model system in which to investigate the molecular mechanisms underlying neurodegenerative diseases. In this review, we discuss recent progress in Drosophila modeling Alzheimer's Disease, Parkinson's Disease, Amyotrophic Lateral Sclerosis (ALS), Huntington's Disease, Ataxia Telangiectasia, and neurodegeneration related to mitochondrial dysfunction or traumatic brain injury. We close by discussing recent progress using Drosophila models of neural regeneration and how these are likely to provide critical insights into future treatments for neurodegenerative disorders.
Collapse
Affiliation(s)
- Harris Bolus
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA;
| | - Kassi Crocker
- Genetics Graduate Training Program, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA;
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Grace Boekhoff-Falk
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA
| | | |
Collapse
|
15
|
Duan S, Gao W, Chen Z, Li Z, Li S, Gan J, Chen X, Li J. Crystal structure of human archease, a key cofactor of tRNA splicing ligase complex. Int J Biochem Cell Biol 2020; 122:105744. [PMID: 32234548 DOI: 10.1016/j.biocel.2020.105744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 11/26/2022]
Abstract
The human archease, hereafter named HArch, is identified as a key cofactor of the tRNA-splicing ligase complex, and a potential therapeutic target for treating nervous system injuries. However, little is known about the structural basis of HArch in tRNA maturation, mRNA splicing, and RNA repair. Here we report the crystal structures of HArch and its two mutants D51A and D178A with resolutions ranging from 1.96 Å to 3.4 Å. HArch is composed of an extended N-terminal protrusion domain (NTD) and one compacted C-terminal domain (CTD). Unlike previously reported homologous proteins, the NTD of the first subunit interacts with the CTD of the second one, and this interaction might be important for maintaining protein stability. Moreover, HArch interacts and colocalizes with RNA ligase RTCB in cells. Our current study reveals the atomic structure of HArch and may help us understand its function in mRNA splicing.
Collapse
Affiliation(s)
- Shuyan Duan
- State Key Laboratory of Genetic Engineering, Department of Neurology, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, 200438, China
| | - Wenqing Gao
- State Key Laboratory of Genetic Engineering, Department of Neurology, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, 200438, China
| | - Zijun Chen
- State Key Laboratory of Genetic Engineering, Department of Neurology, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, 200438, China
| | - Zhengyang Li
- State Key Laboratory of Genetic Engineering, Department of Neurology, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, 200438, China
| | - Suhua Li
- State Key Laboratory of Genetic Engineering, Department of Neurology, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, 200438, China
| | - Jianhua Gan
- State Key Laboratory of Genetic Engineering, Department of Neurology, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, 200438, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jixi Li
- State Key Laboratory of Genetic Engineering, Department of Neurology, School of Life Sciences and Huashan Hospital, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
16
|
Mattedi F, Vagnoni A. Temporal Control of Axonal Transport: The Extreme Case of Organismal Ageing. Front Cell Neurosci 2019; 13:393. [PMID: 31555095 PMCID: PMC6716446 DOI: 10.3389/fncel.2019.00393] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/09/2019] [Indexed: 01/05/2023] Open
Abstract
A fundamental question in cell biology is how cellular components are delivered to their destination with spatial and temporal precision within the crowded cytoplasmic environment. The long processes of neurons represent a significant spatial challenge and make these cells particularly dependent on mechanisms for long-range cytoskeletal transport of proteins, RNA and organelles. Although many studies have substantiated a role for defective transport of axonal cargoes in the pathogenesis of neurodevelopmental and neurodegenerative diseases, remarkably little is known about how transport is regulated throughout ageing. The scale of the challenge posed by ageing is considerable because, in this case, the temporal regulation of transport is ultimately dictated by the length of organismal lifespan, which can extend to days, years or decades. Recent methodological advances to study live axonal transport during ageing in situ have provided new tools to scratch beneath the surface of this complex problem and revealed that age-dependent decline in the transport of mitochondria is a common feature across different neuronal populations of several model organisms. In certain instances, the molecular pathways that affect transport in ageing animals have begun to emerge. However, the functional implications of these observations are still not fully understood. Whether transport decline is a significant determinant of neuronal ageing or a mere consequence of decreased cellular fitness remains an open question. In this review, we discuss the latest developments in axonal trafficking in the ageing nervous system, along with the early studies that inaugurated this new area of research. We explore the possibility that the interplay between mitochondrial function and motility represents a crucial driver of ageing in neurons and put forward the hypothesis that declining axonal transport may be legitimately considered a hallmark of neuronal ageing.
Collapse
Affiliation(s)
| | - Alessio Vagnoni
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, IoPPN, King’s College London, London, United Kingdom
| |
Collapse
|
17
|
Ding C, Hammarlund M. Aberrant information transfer interferes with functional axon regeneration. eLife 2018; 7:e38829. [PMID: 30371349 PMCID: PMC6231761 DOI: 10.7554/elife.38829] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/26/2018] [Indexed: 12/16/2022] Open
Abstract
Functional axon regeneration requires regenerating neurons to restore appropriate synaptic connectivity and circuit function. To model this process, we developed an assay in Caenorhabditis elegans that links axon and synapse regeneration of a single neuron to recovery of behavior. After axon injury and regeneration of the DA9 neuron, synapses reform at their pre-injury location. However, these regenerated synapses often lack key molecular components. Further, synaptic vesicles accumulate in the dendrite in response to axon injury. Dendritic vesicle release results in information misrouting that suppresses behavioral recovery. Dendritic synapse formation depends on dynein and jnk-1. But even when information transfer is corrected, axonal synapses fail to adequately transmit information. Our study reveals unexpected plasticity during functional regeneration. Regeneration of the axon is not sufficient for the reformation of correct neuronal circuits after injury. Rather, synapse reformation and function are also key variables, and manipulation of circuit reformation improves behavioral recovery.
Collapse
Affiliation(s)
- Chen Ding
- Department of NeuroscienceYale UniversityNew HavenUnited States
| | - Marc Hammarlund
- Department of NeuroscienceYale UniversityNew HavenUnited States
- Department of GeneticsYale UniversityNew HavenUnited States
| |
Collapse
|
18
|
Curcio M, Bradke F. Axon Regeneration in the Central Nervous System: Facing the Challenges from the Inside. Annu Rev Cell Dev Biol 2018; 34:495-521. [DOI: 10.1146/annurev-cellbio-100617-062508] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
After an injury in the adult mammalian central nervous system (CNS), lesioned axons fail to regenerate. This failure to regenerate contrasts with axons’ remarkable potential to grow during embryonic development and after an injury in the peripheral nervous system (PNS). Several intracellular mechanisms—including cytoskeletal dynamics, axonal transport and trafficking, signaling and transcription of regenerative programs, and epigenetic modifications—control axon regeneration. In this review, we describe how manipulation of intrinsic mechanisms elicits a regenerative response in different organisms and how strategies are implemented to form the basis of a future regenerative treatment after CNS injury.
Collapse
Affiliation(s)
- Michele Curcio
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany;,
| | - Frank Bradke
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany;,
| |
Collapse
|
19
|
Lossi L, Merighi A. The Use of ex Vivo Rodent Platforms in Neuroscience Translational Research With Attention to the 3Rs Philosophy. Front Vet Sci 2018; 5:164. [PMID: 30073174 PMCID: PMC6060265 DOI: 10.3389/fvets.2018.00164] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/29/2018] [Indexed: 01/08/2023] Open
Abstract
The principles of the 3Rs—Replacement, Reduction, and Refinement—are at the basis of most advanced national and supranational (EU) regulations on animal experimentation and welfare. In the perspective to reduce and refine the use of these animals in translational research, we here discuss the use of rodent acute and organotypically cultured central nervous system slices. We describe novel applications of these ex vivo platforms in medium-throughput screening of neuroactive molecules of potential pharmacological interest, with particular attention to more recent developments that permit to fully exploit the potential of direct genetic engineering of organotypic cultures using transfection techniques. We then describe the perspectives for expanding the use ex vivo platforms in neuroscience studies under the 3Rs philosophy using the following approaches: (1) Use of co-cultures of two brain regions physiologically connected to each other (source-target) to analyze axon regeneration and reconstruction of circuitries; (2) Microinjection or co-cultures of primary cells and/or cell lines releasing one or more neuroactive molecules to screen their physiological and/or pharmacological effects onto neuronal survival and slice circuitry. Microinjected or co-cultured cells are ideally made fluorescent after transfection with a plasmid construct encoding green or red fluorescent protein under the control of a general promoter such as hCMV; (3) Use of “sniffer” cells sensing the release of biologically active molecules from organotypic cultures by means of fluorescent probes. These cells can be prepared with activatable green fluorescent protein, a unique chromophore that remains in a “dark” state because its maturation is inhibited, and can be made fluorescent (de-quenched) if specific cellular enzymes, such as proteases or kinases, are activated.
Collapse
Affiliation(s)
- Laura Lossi
- Laboratory of Neurobiology, Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Adalberto Merighi
- Laboratory of Neurobiology, Department of Veterinary Sciences, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Tissue and cellular rigidity and mechanosensitive signaling activation in Alexander disease. Nat Commun 2018; 9:1899. [PMID: 29765022 PMCID: PMC5954157 DOI: 10.1038/s41467-018-04269-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 04/12/2018] [Indexed: 12/22/2022] Open
Abstract
Glial cells have increasingly been implicated as active participants in the pathogenesis of neurological diseases, but critical pathways and mechanisms controlling glial function and secondary non-cell autonomous neuronal injury remain incompletely defined. Here we use models of Alexander disease, a severe brain disorder caused by gain-of-function mutations in GFAP, to demonstrate that misregulation of GFAP leads to activation of a mechanosensitive signaling cascade characterized by activation of the Hippo pathway and consequent increased expression of A-type lamin. Importantly, we use genetics to verify a functional role for dysregulated mechanotransduction signaling in promoting behavioral abnormalities and non-cell autonomous neurodegeneration. Further, we take cell biological and biophysical approaches to suggest that brain tissue stiffness is increased in Alexander disease. Our findings implicate altered mechanotransduction signaling as a key pathological cascade driving neuronal dysfunction and neurodegeneration in Alexander disease, and possibly also in other brain disorders characterized by gliosis. Alexander disease is a rare neurodegeneration caused by mutations in a glial gene GFAP. Here, Wang and colleagues show in animal models of Alexander disease that GFAP mutant brain and cells have greater tissue and cellular stiffness and greater activation of mechanosensitive signaling cascade.
Collapse
|
21
|
Koch M, Nicolas M, Zschaetzsch M, de Geest N, Claeys A, Yan J, Morgan MJ, Erfurth ML, Holt M, Schmucker D, Hassan BA. A Fat-Facets-Dscam1-JNK Pathway Enhances Axonal Growth in Development and after Injury. Front Cell Neurosci 2018; 11:416. [PMID: 29472843 PMCID: PMC5809495 DOI: 10.3389/fncel.2017.00416] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/12/2017] [Indexed: 11/13/2022] Open
Abstract
Injury to the adult central nervous systems (CNS) can result in severe long-term disability because damaged CNS connections fail to regenerate after trauma. Identification of regulators that enhance the intrinsic growth capacity of severed axons is a first step to restore function. Here, we conducted a gain-of-function genetic screen in Drosophila to identify strong inducers of axonal growth after injury. We focus on a novel axis the Down Syndrome Cell Adhesion Molecule (Dscam1), the de-ubiquitinating enzyme Fat Facets (Faf)/Usp9x and the Jun N-Terminal Kinase (JNK) pathway transcription factor Kayak (Kay)/Fos. Genetic and biochemical analyses link these genes in a common signaling pathway whereby Faf stabilizes Dscam1 protein levels, by acting on the 3'-UTR of its mRNA, and Dscam1 acts upstream of the growth-promoting JNK signal. The mammalian homolog of Faf, Usp9x/FAM, shares both the regenerative and Dscam1 stabilizing activities, suggesting a conserved mechanism.
Collapse
Affiliation(s)
- Marta Koch
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium
| | - Maya Nicolas
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium
| | - Marlen Zschaetzsch
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium
| | - Natalie de Geest
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium
| | - Annelies Claeys
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium
| | - Jiekun Yan
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium
| | - Matthew J Morgan
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium
| | - Maria-Luise Erfurth
- Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium.,Neuronal Wiring Lab, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Matthew Holt
- Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium.,Laboratory of Glia Biology, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Dietmar Schmucker
- Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium.,Neuronal Wiring Lab, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium
| | - Bassem A Hassan
- Laboratory of Neurogenetics, Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie (VIB), Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, KU Leuven, Leuven, Belgium.,Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut du Cerveau et de la Moelle Epinière, Hôpital Pitié-Salpêtrière, UPMC, Sorbonne Universités, Paris, France
| |
Collapse
|
22
|
Sabado V, Nagoshi E. Single-cell Resolution Fluorescence Live Imaging of Drosophila Circadian Clocks in Larval Brain Culture. J Vis Exp 2018. [PMID: 29443100 DOI: 10.3791/57015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The circadian pacemaker circuit orchestrates rhythmic behavioral and physiological outputs coordinated with environmental cues, such as day/night cycles. The molecular clock within each pacemaker neuron generates circadian rhythms in gene expression, which underlie the rhythmic neuronal functions essential to the operation of the circuit. Investigation of the properties of the individual molecular oscillators in different subclasses of pacemaker neurons and their interaction with neuronal signaling yields a better understanding of the circadian pacemaker circuit. Here, we present a time-lapse fluorescent microscopy approach developed to monitor the molecular clockwork in clock neurons of cultured Drosophila larval brain. This method allows the multi-day recording of the rhythms of genetically encoded fluorescent circadian reporters at single-cell resolution. This setup can be combined with pharmacological manipulations to closely analyze real-time response of the molecular clock to various compounds. Beyond circadian rhythms, this multipurpose method in combination with powerful Drosophila genetic techniques offers the possibility to study diverse neuronal or molecular processes in live brain tissue.
Collapse
Affiliation(s)
| | - Emi Nagoshi
- Department of Genetics and Evolution, University of Geneva;
| |
Collapse
|
23
|
Moore EL, Arvidson R, Banks C, Urenda JP, Duong E, Mohammed H, Adams ME. Ampulexins: A New Family of Peptides in Venom of the Emerald Jewel Wasp, Ampulex compressa. Biochemistry 2018; 57:1907-1916. [DOI: 10.1021/acs.biochem.7b00916] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
24
|
Lou WPK, Mateos A, Koch M, Klussman S, Yang C, Lu N, Kumar S, Limpert S, Göpferich M, Zschaetzsch M, Sliwinski C, Kenzelmann M, Seedorf M, Maillo C, Senis E, Grimm D, Puttagunta R, Mendez R, Liu K, Hassan BA, Martin-Villalba A. Regulation of Adult CNS Axonal Regeneration by the Post-transcriptional Regulator Cpeb1. Front Mol Neurosci 2018; 10:445. [PMID: 29379413 PMCID: PMC5770975 DOI: 10.3389/fnmol.2017.00445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022] Open
Abstract
Adult mammalian central nervous system (CNS) neurons are unable to regenerate following axonal injury, leading to permanent functional impairments. Yet, the reasons underlying this regeneration failure are not fully understood. Here, we studied the transcriptome and translatome shortly after spinal cord injury. Profiling of the total and ribosome-bound RNA in injured and naïve spinal cords identified a substantial post-transcriptional regulation of gene expression. In particular, transcripts associated with nervous system development were down-regulated in the total RNA fraction while remaining stably loaded onto ribosomes. Interestingly, motif association analysis of post-transcriptionally regulated transcripts identified the cytoplasmic polyadenylation element (CPE) as enriched in a subset of these transcripts that was more resistant to injury-induced reduction at the transcriptome level. Modulation of these transcripts by overexpression of the CPE binding protein, Cpeb1, in mouse and Drosophila CNS neurons promoted axonal regeneration following injury. Our study uncovered a global evolutionarily conserved post-transcriptional mechanism enhancing regeneration of injured CNS axons.
Collapse
Affiliation(s)
- Wilson Pak-Kin Lou
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Alvaro Mateos
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
| | - Marta Koch
- VIB Center for the Biology of Disease and Center for Human Genetics, VIB and KU Leuven, Leuven, Belgium
| | - Stefan Klussman
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Chao Yang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Na Lu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Sachin Kumar
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Stefanie Limpert
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
| | - Manuel Göpferich
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Marlen Zschaetzsch
- VIB Center for the Biology of Disease and Center for Human Genetics, VIB and KU Leuven, Leuven, Belgium
| | - Christopher Sliwinski
- Department of Neuroregeneration, University Hospital Heidelberg, Heidelberg, Germany
| | - Marc Kenzelmann
- Division of Molecular Biology of the Cell I, German Cancer Research Center, Heidelberg, Germany
| | - Matthias Seedorf
- Zentrum für Molekulare Biologie, University of Heidelberg, Heidelberg, Germany
| | - Carlos Maillo
- Translational Control of Cell Cycle and Differentiation, Institute for Research in Biomedicine, Barcelona, Spain
| | - Elena Senis
- Virus Host Interaction, Heidelberg University Hospital, Center for Infectious Diseases/Virology, Cluster of Excellence CellNetworks, BioQuant, Heidelberg, Germany
| | - Dirk Grimm
- Virus Host Interaction, Heidelberg University Hospital, Center for Infectious Diseases/Virology, Cluster of Excellence CellNetworks, BioQuant, Heidelberg, Germany
| | - Radhika Puttagunta
- Department of Neuroregeneration, University Hospital Heidelberg, Heidelberg, Germany
| | - Raul Mendez
- Translational Control of Cell Cycle and Differentiation, Institute for Research in Biomedicine, Barcelona, Spain
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, Hong Kong
- Center of Systems Biology and Human Health, School of Science and Institute for Advanced Study, Hong Kong University of Science and Technology, Hong Kong, Hong Kong
| | - Bassem A. Hassan
- VIB Center for the Biology of Disease and Center for Human Genetics, VIB and KU Leuven, Leuven, Belgium
- Sorbonne Universités, UPMC Univ Paris 06, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut du Cerveau et de la Moelle epiniere - Hôpital Pitié-Salpêtrière, Paris, France
| | - Ana Martin-Villalba
- Division of Molecular Neurobiology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
25
|
Mezan S, Feuz JD, Deplancke B, Kadener S. PDF Signaling Is an Integral Part of the Drosophila Circadian Molecular Oscillator. Cell Rep 2017; 17:708-719. [PMID: 27732848 PMCID: PMC5081397 DOI: 10.1016/j.celrep.2016.09.048] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 08/12/2016] [Accepted: 09/15/2016] [Indexed: 12/30/2022] Open
Abstract
Circadian clocks generate 24-hr rhythms in physiology and behavior. Despite numerous studies, it is still uncertain how circadian rhythms emerge from their molecular and neural constituents. Here, we demonstrate a tight connection between the molecular and neuronal circadian networks. Using fluorescent transcriptional reporters in a Drosophila ex vivo brain culture system, we identified a reciprocal negative regulation between the master circadian regulator CLK and expression of pdf, the main circadian neuropeptide. We show that PDF feedback is required for maintaining normal oscillation pattern in CLK-driven transcription. Interestingly, we found that CLK and neuronal firing suppresses pdf transcription, likely through a common pathway involving the transcription factors DHR38 and SR, establishing a direct link between electric activity and the circadian system. In sum, our work provides evidence for the existence of an uncharacterized CLK-PDF feedback loop that tightly wraps together the molecular oscillator with the circadian neuronal network in Drosophila. Monitoring circadian transcription ex vivo using fluorescent reporters CLK activation in the LNvs provokes downregulation in CLK activity in LNds and DNs Reciprocal negative regulation of CLK activity and pdf transcription and signaling PDF signaling is required for the normal oscillation pattern in CLK activity
Collapse
Affiliation(s)
- Shaul Mezan
- Biological Chemistry Department, Silberman Institute of Life Sciences, the Hebrew University, Jerusalem 91904, Israel
| | - Jean Daniel Feuz
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Bart Deplancke
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Sebastian Kadener
- Biological Chemistry Department, Silberman Institute of Life Sciences, the Hebrew University, Jerusalem 91904, Israel.
| |
Collapse
|
26
|
Kelly SM, Elchert A, Kahl M. Dissection and Immunofluorescent Staining of Mushroom Body and Photoreceptor Neurons in Adult Drosophila melanogaster Brains. J Vis Exp 2017. [PMID: 29155751 PMCID: PMC5755316 DOI: 10.3791/56174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Nervous system development involves a sequential series of events that are coordinated by several signaling pathways and regulatory networks. Many of the proteins involved in these pathways are evolutionarily conserved between mammals and other eukaryotes, such as the fruit fly Drosophila melanogaster, suggesting that similar organizing principles exist during the development of these organisms. Importantly, Drosophila has been used extensively to identify cellular and molecular mechanisms regulating processes that are required in mammals including neurogenesis, differentiation, axonal guidance, and synaptogenesis. Flies have also been used successfully to model a variety of human neurodevelopmental diseases. Here we describe a protocol for the step-by-step microdissection, fixation, and immunofluorescent localization of proteins within the adult Drosophila brain. This protocol focuses on two example neuronal populations, mushroom body neurons and retinal photoreceptors, and includes optional steps to trace individual mushroom body neurons using Mosaic Analysis with a Repressible Cell Marker (MARCM) technique. Example data from both wild-type and mutant brains are shown along with a brief description of a scoring criteria for axonal guidance defects. While this protocol highlights two well-established antibodies for investigating the morphology of mushroom body and photoreceptor neurons, other Drosophila brain regions and the localization of proteins within other brain regions can also be investigated using this protocol.
Collapse
Affiliation(s)
- Seth M Kelly
- Program in Neuroscience, The College of Wooster; Department of Biology, The College of Wooster;
| | - Alexandra Elchert
- Program in Biochemistry, Cellular, and Molecular Biology, The College of Wooster
| | - Michael Kahl
- Department of Biology, The College of Wooster; Program in Biochemistry, Cellular, and Molecular Biology, The College of Wooster
| |
Collapse
|
27
|
Kato K, Losada-Perez M, Hidalgo A. Gene network underlying the glial regenerative response to central nervous system injury. Dev Dyn 2017; 247:85-93. [DOI: 10.1002/dvdy.24565] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 12/30/2022] Open
Affiliation(s)
- Kentaro Kato
- School of Medicine; Kyorin University; Tokyo Japan
| | | | - Alicia Hidalgo
- School of Biosciences; University of Birmingham; United Kingdom
| |
Collapse
|
28
|
Purice MD, Ray A, Münzel EJ, Pope BJ, Park DJ, Speese SD, Logan MA. A novel Drosophila injury model reveals severed axons are cleared through a Draper/MMP-1 signaling cascade. eLife 2017; 6. [PMID: 28825401 PMCID: PMC5565368 DOI: 10.7554/elife.23611] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 07/25/2017] [Indexed: 02/06/2023] Open
Abstract
Neural injury triggers swift responses from glia, including glial migration and phagocytic clearance of damaged neurons. The transcriptional programs governing these complex innate glial immune responses are still unclear. Here, we describe a novel injury assay in adult Drosophila that elicits widespread glial responses in the ventral nerve cord (VNC). We profiled injury-induced changes in VNC gene expression by RNA sequencing (RNA-seq) and found that responsive genes fall into diverse signaling classes. One factor, matrix metalloproteinase-1 (MMP-1), is induced in Drosophila ensheathing glia responding to severed axons. Interestingly, glial induction of MMP-1 requires the highly conserved engulfment receptor Draper, as well as AP-1 and STAT92E. In MMP-1 depleted flies, glia do not properly infiltrate neuropil regions after axotomy and, as a consequence, fail to clear degenerating axonal debris. This work identifies Draper-dependent activation of MMP-1 as a novel cascade required for proper glial clearance of severed axons.
Collapse
Affiliation(s)
- Maria D Purice
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Arpita Ray
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Eva Jolanda Münzel
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Bernard J Pope
- Melbourne Informatics, The University of Melbourne, Melbourne, Australia
| | - Daniel J Park
- Melbourne Informatics, The University of Melbourne, Melbourne, Australia
| | - Sean D Speese
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, Portland, United States
| | - Mary A Logan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, Portland, United States
| |
Collapse
|
29
|
Regeneration of synapses in the olfactory pathway of locusts after antennal deafferentation. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2017; 203:867-877. [PMID: 28685185 DOI: 10.1007/s00359-017-1199-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/31/2022]
Abstract
The olfactory pathway of the locust is capable of fast and precise regeneration on an anatomical level. Following deafferentation of the antenna either of young adult locusts, or of fifth instar nymphs, severed olfactory receptor neurons (ORNs) reinnervate the antennal lobe (AL) and arborize in AL microglomeruli. In the present study we tested whether these regenerated fibers establish functional synapses again. Intracellular recordings from AL projection neurons revealed that the first few odor stimulus evoked postsynaptic responses from regenerated ORNs from day 4-7 post crush on. On average, synaptic connections of regenerated afferents appeared faster in younger locusts operated as fifth instar nymphs than in adults. The proportions of response categories (excitatory vs. inhibitory) changed during regeneration, but were back to normal within 21 days. Odor-evoked oscillating extracellular local field potentials (LFP) were recorded in the mushroom body. These responses, absent after antennal nerve crush, reappeared, in a few animals as soon as 4 days post crush. Odor-induced oscillation patterns were restored within 7 days post crush. Both intra- and extracellular recordings indicate the capability of the locust olfactory system to re-establish synaptic contacts in the antennal lobe after antennal nerve lesion.
Collapse
|
30
|
Hao Y, Collins C. Intrinsic mechanisms for axon regeneration: insights from injured axons in Drosophila. Curr Opin Genet Dev 2017; 44:84-91. [PMID: 28232273 PMCID: PMC5447494 DOI: 10.1016/j.gde.2017.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/17/2017] [Accepted: 01/26/2017] [Indexed: 12/18/2022]
Abstract
Axonal damage and loss are common and negative consequences of neuronal injuries, and also occur in some neurodegenerative diseases. For neurons to have a chance to repair their connections, they need to survive the damage, initiate new axonal growth, and ultimately establish new synaptic connections. This review discusses how recent work in Drosophila models have informed our understanding of the cellular pathways used by neurons to respond to axonal injuries. Similarly to mammalian neurons, Drosophila neurons appear to be more limited in their capacity regrow (regenerate) damaged axons in the central nervous system, but can undergo axonal regeneration to varying extents in the peripheral nervous system. Conserved cellular pathways are activated by axonal injury via mechanisms that are specific to axons but not dendrites, and new unanticipated inhibitors of axon regeneration can be identified via genetic screening. These findings, made predominantly via genetic and live imaging methods in Drosophila, emphasize the utility of this model organism for the identification and study of basic cellular mechanisms used for neuronal repair.
Collapse
Affiliation(s)
- Yan Hao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Catherine Collins
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA.
| |
Collapse
|
31
|
Regeneration of axotomized olfactory neurons in young and adult locusts quantified by fasciclin I immunofluorescence. Cell Tissue Res 2017; 368:1-12. [PMID: 28150067 DOI: 10.1007/s00441-016-2560-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/05/2016] [Indexed: 01/22/2023]
Abstract
The olfactory pathway of the locust Locusta migratoria is characterized by a multiglomerular innervation of the antennal lobe (AL) by olfactory receptor neurons (ORNs). After crushing the antenna and thereby severing ORN axons, changes in the AL were monitored. First, volume changes were measured at different times post-crush with scanning laser optical tomography in 5th instar nymphs. AL volume decreased significantly to a minimum volume at 4 days post-crush, followed by an increase. Second, anterograde labeling was used to visualize details in the AL and antennal nerve (AN) during de- and regeneration. Within 24 h post-crush (hpc) the ORN fragments distal to the lesion degenerated. After 48 hpc, regenerating fibers grew through the crush site. In the AL, labeled ORN projections disappeared completely and reappeared after a few days. A weak topographic match between ORN origin on the antenna and the position of innervated glomeruli that was present in untreated controls did not reappear after regeneration. Third, the cell surface marker fasciclin I that is expressed in ORNs was used for quantifying purposes. Immunofluorescence was measured in the AL during de- and regeneration in adults and 5th instar nymphs: after a rapid but transient, decrease, it reappeared. Both processes happen faster in 5th instar nymphs than in adults.
Collapse
|
32
|
Brace EJ, DiAntonio A. Models of axon regeneration in Drosophila. Exp Neurol 2017; 287:310-317. [PMID: 26996133 PMCID: PMC5026866 DOI: 10.1016/j.expneurol.2016.03.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 12/14/2022]
Abstract
Maintaining neuronal connectivity in the face of injury and disease is a major challenge for the nervous system. The great length of axons makes them particularly vulnerable to insult with dire consequences for neuronal function. In the peripheral nervous system there is a program of axonal regeneration that can reestablish connectivity. In the mammalian central nervous system, however, injured axons have little or no capacity to regenerate. The molecular mechanisms that promote axon regeneration have begun to be identified and many of the implicated pathways are evolutionarily conserved. Here we discuss Drosophila models of axonal regrowth, describe insights derived from these studies, and highlight future directions in the use of the fly for dissecting the mechanisms of axonal regeneration.
Collapse
Affiliation(s)
- E J Brace
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA, 63110
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA, 63110.
| |
Collapse
|
33
|
Losada-Perez M, Harrison N, Hidalgo A. Molecular mechanism of central nervous system repair by the Drosophila NG2 homologue kon-tiki. J Cell Biol 2016; 214:587-601. [PMID: 27551055 PMCID: PMC5004445 DOI: 10.1083/jcb.201603054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 07/25/2016] [Indexed: 11/22/2022] Open
Abstract
Glial cells help central nervous system injury repair, but this is limited by the failure of newly produced glial cells to differentiate. Here, Losada-Perez et al. identify the NG2-dependent mechanism modulating glial proliferation and differentiation after damage to promote repair, in the central nervous system of Drosophila. Neuron glia antigen 2 (NG2)–positive glia are repair cells that proliferate upon central nervous system (CNS) damage, promoting functional recovery. However, repair is limited because of the failure of the newly produced glial cells to differentiate. It is a key goal to discover how to regulate NG2 to enable glial proliferation and differentiation conducive to repair. Drosophila has an NG2 homologue called kon-tiki (kon), of unknown CNS function. We show that kon promotes repair and identify the underlying mechanism. Crush injury up-regulates kon expression downstream of Notch. Kon in turn induces glial proliferation and initiates glial differentiation by activating glial genes and prospero (pros). Two negative feedback loops with Notch and Pros allow Kon to drive the homeostatic regulation required for repair. By modulating Kon levels in glia, we could prevent or promote CNS repair. Thus, the functional links between Kon, Notch, and Pros are essential for, and can drive, repair. Analogous mechanisms could promote CNS repair in mammals.
Collapse
Affiliation(s)
- Maria Losada-Perez
- NeuroDevelopment Lab, School of Biosciences, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Neale Harrison
- NeuroDevelopment Lab, School of Biosciences, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Alicia Hidalgo
- NeuroDevelopment Lab, School of Biosciences, University of Birmingham, Birmingham B15 2TT, England, UK
| |
Collapse
|
34
|
Musashe DT, Purice MD, Speese SD, Doherty J, Logan MA. Insulin-like Signaling Promotes Glial Phagocytic Clearance of Degenerating Axons through Regulation of Draper. Cell Rep 2016; 16:1838-50. [PMID: 27498858 DOI: 10.1016/j.celrep.2016.07.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 05/23/2016] [Accepted: 07/09/2016] [Indexed: 01/15/2023] Open
Abstract
Neuronal injury triggers robust responses from glial cells, including altered gene expression and enhanced phagocytic activity to ensure prompt removal of damaged neurons. The molecular underpinnings of glial responses to trauma remain unclear. Here, we find that the evolutionarily conserved insulin-like signaling (ILS) pathway promotes glial phagocytic clearance of degenerating axons in adult Drosophila. We find that the insulin-like receptor (InR) and downstream effector Akt1 are acutely activated in local ensheathing glia after axotomy and are required for proper clearance of axonal debris. InR/Akt1 activity, it is also essential for injury-induced activation of STAT92E and its transcriptional target draper, which encodes a conserved receptor essential for glial engulfment of degenerating axons. Increasing Draper levels in adult glia partially rescues delayed clearance of severed axons in glial InR-inhibited flies. We propose that ILS functions as a key post-injury communication relay to activate glial responses, including phagocytic activity.
Collapse
Affiliation(s)
- Derek T Musashe
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Maria D Purice
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Sean D Speese
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Johnna Doherty
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 55 North Lake Avenue, Worcester, MA 01605, USA
| | - Mary A Logan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
35
|
Roberts L, Leise TL, Welsh DK, Holmes TC. Functional Contributions of Strong and Weak Cellular Oscillators to Synchrony and Light-shifted Phase Dynamics. J Biol Rhythms 2016; 31:337-51. [PMID: 27221103 DOI: 10.1177/0748730416649550] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Light is the primary signal that calibrates circadian neural circuits and thus coordinates daily physiological and behavioral rhythms with solar entrainment cues. Drosophila and mammalian circadian circuits consist of diverse populations of cellular oscillators that exhibit a wide range of dynamic light responses, periods, phases, and degrees of synchrony. How heterogeneous circadian circuits can generate robust physiological rhythms while remaining flexible enough to respond to synchronizing stimuli has long remained enigmatic. Cryptochrome is a short-wavelength photoreceptor that is endogenously expressed in approximately half of Drosophila circadian neurons. In a previous study, physiological light response was measured using real-time bioluminescence recordings in Drosophila whole-brain explants, which remain intrinsically light-sensitive. Here we apply analysis of real-time bioluminescence experimental data to show detailed dynamic ensemble representations of whole circadian circuit light entrainment at single neuron resolution. Organotypic whole-brain explants were either maintained in constant darkness (DD) for 6 days or exposed to a phase-advancing light pulse on the second day. We find that stronger circadian oscillators support robust overall circuit rhythmicity in DD, whereas weaker oscillators can be pushed toward transient desynchrony and damped amplitude to facilitate a new state of phase-shifted network synchrony. Additionally, we use mathematical modeling to examine how a network composed of distinct oscillator types can give rise to complex dynamic signatures in DD conditions and in response to simulated light pulses. Simulations suggest that complementary coupling mechanisms and a combination of strong and weak oscillators may enable a robust yet flexible circadian network that promotes both synchrony and entrainment. A more complete understanding of how the properties of oscillators and their signaling mechanisms facilitate their distinct roles in light entrainment may allow us to direct and augment the circadian system to speed recovery from jet lag, shift work, and seasonal affective disorder.
Collapse
Affiliation(s)
- Logan Roberts
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
| | - Tanya L Leise
- Department of Mathematics and Statistics, Amherst College, Amherst, MA
| | - David K Welsh
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, La Jolla, CA Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Todd C Holmes
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA
| |
Collapse
|
36
|
Exosomes Derived from Mesenchymal Stromal Cells Promote Axonal Growth of Cortical Neurons. Mol Neurobiol 2016; 54:2659-2673. [PMID: 26993303 DOI: 10.1007/s12035-016-9851-0] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/09/2016] [Indexed: 02/07/2023]
Abstract
Treatment of brain injury with exosomes derived from mesenchymal stromal cells (MSCs) enhances neurite growth. However, the direct effect of exosomes on axonal growth and molecular mechanisms underlying exosome-enhanced neurite growth are not known. Using primary cortical neurons cultured in a microfluidic device, we found that MSC-exosomes promoted axonal growth, whereas attenuation of argonaut 2 protein, one of the primary microRNA (miRNA) machinery proteins, in MSC-exosomes abolished their effect on axonal growth. Both neuronal cell bodies and axons internalized MSC-exosomes, which was blocked by botulinum neurotoxins (BoNTs) that cleave proteins of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex. Moreover, tailored MSC-exosomes carrying elevated miR-17-92 cluster further enhanced axonal growth compared to native MSC-exosomes. Quantitative RT-PCR and Western blot analysis showed that the tailored MSC-exosomes increased levels of individual members of this cluster and activated the PTEN/mTOR signaling pathway in recipient neurons, respectively. Together, our data demonstrate that native MSC-exosomes promote axonal growth while the tailored MSC-exosomes can further boost this effect and that tailored exosomes can deliver their selective cargo miRNAs into and activate their target signals in recipient neurons. Neuronal internalization of MSC-exosomes is mediated by the SNARE complex. This study reveals molecular mechanisms that contribute to MSC-exosome-promoted axonal growth, which provides a potential therapeutic strategy to enhance axonal growth.
Collapse
|
37
|
Okray Z, de Esch CEF, Van Esch H, Devriendt K, Claeys A, Yan J, Verbeeck J, Froyen G, Willemsen R, de Vrij FMS, Hassan BA. A novel fragile X syndrome mutation reveals a conserved role for the carboxy-terminus in FMRP localization and function. EMBO Mol Med 2015; 7:423-37. [PMID: 25693964 PMCID: PMC4403044 DOI: 10.15252/emmm.201404576] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Loss of function of the FMR1 gene leads to fragile X syndrome (FXS), the most common form of intellectual disability. The loss of FMR1 function is usually caused by epigenetic silencing of the FMR1 promoter leading to expansion and subsequent methylation of a CGG repeat in the 5′ untranslated region. Very few coding sequence variations have been experimentally characterized and shown to be causal to the disease. Here, we describe a novel FMR1 mutation and reveal an unexpected nuclear export function for the C-terminus of FMRP. We screened a cohort of patients with typical FXS symptoms who tested negative for CGG repeat expansion in the FMR1 locus. In one patient, we identified a guanine insertion in FMR1 exon 15. This mutation alters the open reading frame creating a short novel C-terminal sequence, followed by a stop codon. We find that this novel peptide encodes a functional nuclear localization signal (NLS) targeting the patient FMRP to the nucleolus in human cells. We also reveal an evolutionarily conserved nuclear export function associated with the endogenous C-terminus of FMRP. In vivo analyses in Drosophila demonstrate that a patient-mimetic mutation alters the localization and function of Dfmrp in neurons, leading to neomorphic neuronal phenotypes.
Collapse
Affiliation(s)
- Zeynep Okray
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium Program in Molecular and Developmental Genetics, Doctoral School of Biomedical Sciences, University of Leuven, Leuven, Belgium
| | - Celine E F de Esch
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Hilde Van Esch
- Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium
| | - Koen Devriendt
- Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium
| | - Annelies Claeys
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium
| | - Jiekun Yan
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium
| | - Jelle Verbeeck
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium
| | - Guy Froyen
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Femke M S de Vrij
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bassem A Hassan
- VIB Center for the Biology of Disease, VIB, Leuven, Belgium Center for Human Genetics, University of Leuven School of Medicine and University Hospitals Leuven, Leuven, Belgium Program in Molecular and Developmental Genetics, Doctoral School of Biomedical Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Özel MN, Langen M, Hassan BA, Hiesinger PR. Filopodial dynamics and growth cone stabilization in Drosophila visual circuit development. eLife 2015; 4. [PMID: 26512889 PMCID: PMC4728134 DOI: 10.7554/elife.10721] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/26/2015] [Indexed: 01/04/2023] Open
Abstract
Filopodial dynamics are thought to control growth cone guidance, but the types and roles of growth cone dynamics underlying neural circuit assembly in a living brain are largely unknown. To address this issue, we have developed long-term, continuous, fast and high-resolution imaging of growth cone dynamics from axon growth to synapse formation in cultured Drosophila brains. Using R7 photoreceptor neurons as a model we show that >90% of the growth cone filopodia exhibit fast, stochastic dynamics that persist despite ongoing stepwise layer formation. Correspondingly, R7 growth cones stabilize early and change their final position by passive dislocation. N-Cadherin controls both fast filopodial dynamics and growth cone stabilization. Surprisingly, loss of N-Cadherin causes no primary targeting defects, but destabilizes R7 growth cones to jump between correct and incorrect layers. Hence, growth cone dynamics can influence wiring specificity without a direct role in target recognition and implement simple rules during circuit assembly. DOI:http://dx.doi.org/10.7554/eLife.10721.001 Genes encode complicated developmental processes, but it is clear that genetic information cannot encode each and every individual connection that forms between the nerve cells in a brain. Instead, the individual cells and nerve endings must make decisions during brain development. Up until now, few examples were known for how these nerve endings move and choose their paths and partners in a living, developing brain. The fruit fly Drosophila provides a useful model to explore the ‘wiring’ of nerve cells in the brain, partly because a fruit fly’s brain develops within a few days. However, most previous studies have relied on identifying mutant flies with disrupted brain wiring and studying them using still images. Now, Özel et al. have developed a new imaging method that has enough resolution and speed over sufficiently long periods to track the growing nerve endings in a developing fly brain. The method was applied to a model nerve cell in the fly’s visual system. This revealed that most of this nerve’s dynamic changes are short-lived and random, and appear to help to stabilize the developing nerve ending, rather than guide it to a target. Özel et al. also found that a protein called N-Cadherin, previously thought to be required for the targeting of developing nerve endings, actually plays a role in their stabilization. These findings uncover the roles of changes in nerve endings during long-term brain development; this was previously largely unknown for any organism. The next stage in this research will involve further analyses of both wild type and mutant flies to try and work out general principles about how the brain develops via the decoding of genetic information. DOI:http://dx.doi.org/10.7554/eLife.10721.002
Collapse
Affiliation(s)
- Mehmet Neset Özel
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, United States.,Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence, Charite Universitätsmedizin Berlin, Berlin, Germany
| | - Marion Langen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Bassem A Hassan
- Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, Leuven, Belgium
| | - P Robin Hiesinger
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, United States.,Division of Neurobiology, Institute for Biology, Freie Universität Berlin, Berlin, Germany.,NeuroCure Cluster of Excellence, Charite Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
39
|
Rabinovich D, Mayseless O, Schuldiner O. Long term ex vivo culturing of Drosophila brain as a method to live image pupal brains: insights into the cellular mechanisms of neuronal remodeling. Front Cell Neurosci 2015; 9:327. [PMID: 26379498 PMCID: PMC4547045 DOI: 10.3389/fncel.2015.00327] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/07/2015] [Indexed: 01/01/2023] Open
Abstract
Holometabolous insects, including Drosophila melanogaster, undergo complete metamorphosis that includes a pupal stage. During metamorphosis, the Drosophila nervous system undergoes massive remodeling and growth, that include cell death and large-scale axon and synapse elimination as well as neurogenesis, developmental axon regrowth, and formation of new connections. Neuronal remodeling is an essential step in the development of vertebrate and invertebrate nervous systems. Research on the stereotypic remodeling of Drosophila mushroom body (MB) γ neurons has contributed to our knowledge of the molecular mechanisms of remodeling but our knowledge of the cellular mechanisms remain poorly understood. A major hurdle in understanding various dynamic processes that occur during metamorphosis is the lack of time-lapse resolution. The pupal case and opaque fat bodies that enwrap the central nervous system (CNS) make live-imaging of the central brain in-vivo impossible. We have established an ex vivo long-term brain culture system that supports the development and neuronal remodeling of pupal brains. By optimizing culture conditions and dissection protocols, we have observed development in culture at kinetics similar to what occurs in vivo. Using this new method, we have obtained the first time-lapse sequence of MB γ neurons undergoing remodeling in up to a single cell resolution. We found that axon pruning is initiated by blebbing, followed by one-two nicks that seem to initiate a more widely spread axon fragmentation. As such, we have set up some of the tools and methodologies needed for further exploration of the cellular mechanisms of neuronal remodeling, not limited to the MB. The long-term ex vivo brain culture system that we report here could be used to study dynamic aspects of neurodevelopment of any Drosophila neuron.
Collapse
Affiliation(s)
- Dana Rabinovich
- Department of Molecular Cell Biology, Weizmann Institute of Sciences Rehovot, Israel
| | - Oded Mayseless
- Department of Molecular Cell Biology, Weizmann Institute of Sciences Rehovot, Israel
| | - Oren Schuldiner
- Department of Molecular Cell Biology, Weizmann Institute of Sciences Rehovot, Israel
| |
Collapse
|
40
|
Marmor-Kollet N, Schuldiner O. Contrasting developmental axon regrowth and neurite sprouting of Drosophila mushroom body neurons reveals shared and unique molecular mechanisms. Dev Neurobiol 2015; 76:262-76. [PMID: 26037037 DOI: 10.1002/dneu.22312] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/14/2015] [Accepted: 05/28/2015] [Indexed: 11/06/2022]
Abstract
The molecular mechanisms regulating intrinsic axon growth potential during development or following injury remain largely unknown despite their vast importance. Here, we have established a neurite sprouting assay of primary cultured mushroom body (MB) neurons. We used the MARCM technique to both mark and manipulate MB neurons, enabling us to quantify the sprouting abilities of single WT and mutant neurons originating from flies at different developmental stages. Sprouting of dissociated MB neurons was dependent on wnd, the DLK ortholog, a conserved gene that is required for axon regeneration. Next, and as expected, we found that the sprouting ability of adult MB neurons was significantly decreased. In contrast, and to our surprise, we found that pupal-derived neurons exhibit increased sprouting compared with neurons derived from larvae, suggesting the existence of an elevated growth potential state. We then contrasted the molecular requirements of neurite sprouting to developmental axon regrowth of MB ɣ neurons, a process that we have previously shown requires the nuclear receptor UNF acting via the target of rapamycin (TOR) pathway. Strikingly, we found that while TOR was required for neurite sprouting, UNF was not. In contrast, we found that PTEN inhibits sprouting in adult neurons, suggesting that TOR is regulated by the PI3K/PTEN pathway during sprouting and by UNF during developmental regrowth. Interestingly, the PI3K pathway as well as Wnd were not required for developmental regrowth nor for initial axon outgrowth suggesting that axon growth during circuit formation, remodeling, and regeneration share some molecular components but differ in others.
Collapse
Affiliation(s)
- Neta Marmor-Kollet
- Department of Molecular Cell Biology, Weizmann Institute of Sciences, Rehovot, 76100, Israel
| | - Oren Schuldiner
- Department of Molecular Cell Biology, Weizmann Institute of Sciences, Rehovot, 76100, Israel
| |
Collapse
|
41
|
Medioni C, Ephrussi A, Besse F. Live imaging of axonal transport in Drosophila pupal brain explants. Nat Protoc 2015; 10:574-84. [PMID: 25763834 DOI: 10.1038/nprot.2015.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Axonal transport is essential for the initial growth, maintenance and synaptic plasticity of axons, and altered axonal transport has been observed in different models of neurodegenerative pathologies. Dissecting the mechanisms underlying axonal transport in developing or degenerating brains requires dynamic imaging of axonal cargo movement in living samples. Whereas methods exist to image axonal transport in Drosophila larval neurons, they are not suitable to follow this process during metamorphosis, when brains undergo extensive remodeling. Here we present a simple method that enables confocal imaging of both fast and slow axonal transport in Drosophila pupal brain explants. We describe how to prepare chambers adapted for live imaging, how to maintain brain explants under physiological conditions and how to monitor and quantitatively analyze the movement of fluorescently labeled cargoes. This protocol requires minimal equipment and is ideally suited for experiments that combine genetics, optogenetics and pharmacological approaches. The brains can be prepared for image acquisition in 1.5 h, and the protocol can be performed easily in any fly laboratory.
Collapse
Affiliation(s)
- Caroline Medioni
- Institute of Biology Valrose, Centre National de la Recherche Scientifique (CNRS)-Unité Mixte de Recherche (UMR) 7277, Institut National de la Santé et de la Recherche Médicale (INSERM)-UMR1091, University of Nice-Sophia Antipolis, Nice, France
| | | | - Florence Besse
- Institute of Biology Valrose, Centre National de la Recherche Scientifique (CNRS)-Unité Mixte de Recherche (UMR) 7277, Institut National de la Santé et de la Recherche Médicale (INSERM)-UMR1091, University of Nice-Sophia Antipolis, Nice, France
| |
Collapse
|
42
|
Roberts L, Leise TL, Noguchi T, Galschiodt AM, Houl JH, Welsh DK, Holmes TC. Light evokes rapid circadian network oscillator desynchrony followed by gradual phase retuning of synchrony. Curr Biol 2015; 25:858-67. [PMID: 25754644 DOI: 10.1016/j.cub.2015.01.056] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/26/2014] [Accepted: 01/21/2015] [Indexed: 11/30/2022]
Abstract
Circadian neural circuits generate near 24-hr physiological rhythms that can be entrained by light to coordinate animal physiology with daily solar cycles. To examine how a circadian circuit reorganizes its activity in response to light, we imaged period (per) clock gene cycling for up to 6 days at single-neuron resolution in whole-brain explant cultures prepared from per-luciferase transgenic flies. We compared cultures subjected to a phase-advancing light pulse (LP) to cultures maintained in darkness (DD). In DD, individual neuronal oscillators in all circadian subgroups are initially well synchronized but then show monotonic decrease in oscillator rhythm amplitude and synchrony with time. The small ventral lateral neurons (s-LNvs) and dorsal lateral neurons (LNds) exhibit this decrease at a slower relative rate. In contrast, the LP evokes a rapid loss of oscillator synchrony between and within most circadian neuronal subgroups, followed by gradual phase retuning of whole-circuit oscillator synchrony. The LNds maintain high rhythmic amplitude and synchrony following the LP along with the most rapid coherent phase advance. Immunocytochemical analysis of PER shows that these dynamics in DD and LP are recapitulated in vivo. Anatomically distinct circadian neuronal subgroups vary in their response to the LP, showing differences in the degree and kinetics of their loss, recovery and/or strengthening of synchrony, and rhythmicity. Transient desynchrony appears to be an integral feature of light response of the Drosophila multicellular circadian clock. Individual oscillators in different neuronal subgroups of the circadian circuit show distinct kinetic signatures of light response and phase retuning.
Collapse
Affiliation(s)
- Logan Roberts
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Tanya L Leise
- Department of Mathematics and Statistics, Amherst College, Amherst, MA 01002, USA
| | - Takako Noguchi
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexis M Galschiodt
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Jerry H Houl
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - David K Welsh
- Department of Psychiatry and Center for Circadian Biology, University of California, San Diego, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| | - Todd C Holmes
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
43
|
Soares L, Parisi M, Bonini NM. Axon injury and regeneration in the adult Drosophila. Sci Rep 2014; 4:6199. [PMID: 25160612 PMCID: PMC4145289 DOI: 10.1038/srep06199] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/04/2014] [Indexed: 01/09/2023] Open
Abstract
Neural regeneration is a fascinating process with profound impact on human health, such that defining biological and genetic pathways is of interest. Here we describe an in vivo preparation for neuronal regeneration in the adult Drosophila. The nerve along the anterior margin of the wing is comprised of ~225 neurons that send projections into the central neuropil (thorax). Precise ablation can be induced with a pulsed laser to sever the entire axonal tract. The animal can be recovered, and response to injury assessed over time. Upon ablation, there is local loss of axons near the injury site, scar formation, a rapid impact on the cytoskeleton, and stimulation of hemocytes. By 7d, ~50% of animals show nerve regrowth, with axons from the nerve cells extending down towards the injury or re-routing. Inhibition of JNK signaling promotes regrowth through the injury site, enabling regeneration of the axonal tract.
Collapse
Affiliation(s)
- Lorena Soares
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael Parisi
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
44
|
Gorostiza EA, Depetris-Chauvin A, Frenkel L, Pírez N, Ceriani MF. Circadian pacemaker neurons change synaptic contacts across the day. Curr Biol 2014; 24:2161-2167. [PMID: 25155512 DOI: 10.1016/j.cub.2014.07.063] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 06/14/2014] [Accepted: 07/23/2014] [Indexed: 12/19/2022]
Abstract
Daily cycles of rest and activity are a common example of circadian control of physiology. In Drosophila, rhythmic locomotor cycles rely on the activity of 150-200 neurons grouped in seven clusters [1, 2]. Work from many laboratories points to the small ventral lateral neurons (sLNvs) as essential for circadian control of locomotor rhythmicity [3-7]. sLNv neurons undergo circadian remodeling of their axonal projections, opening the possibility for a circadian control of connectivity of these relevant circadian pacemakers [8]. Here we show that circadian plasticity of the sLNv axonal projections has further implications than mere structural changes. First, we found that the degree of daily structural plasticity exceeds that originally described [8], underscoring that changes in the degree of fasciculation as well as extension or pruning of axonal terminals could be involved. Interestingly, the quantity of active zones changes along the day, lending support to the attractive hypothesis that new synapses are formed while others are dismantled between late night and the following morning. More remarkably, taking full advantage of the GFP reconstitution across synaptic partners (GRASP) technique [9], we showed that, in addition to new synapses being added or removed, sLNv neurons contact different synaptic partners at different times along the day. These results lead us to propose that the circadian network, and in particular the sLNv neurons, orchestrates some of the physiological and behavioral differences between day and night by changing the path through which information travels.
Collapse
Affiliation(s)
- E Axel Gorostiza
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Avenida Patricias Argentinas 435, 1405-BWE Buenos Aires, Argentina
| | - Ana Depetris-Chauvin
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Avenida Patricias Argentinas 435, 1405-BWE Buenos Aires, Argentina
| | - Lia Frenkel
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Avenida Patricias Argentinas 435, 1405-BWE Buenos Aires, Argentina
| | - Nicolás Pírez
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Avenida Patricias Argentinas 435, 1405-BWE Buenos Aires, Argentina
| | - María Fernanda Ceriani
- Laboratorio de Genética del Comportamiento, Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas-Buenos Aires (IIB-BA, CONICET), Avenida Patricias Argentinas 435, 1405-BWE Buenos Aires, Argentina.
| |
Collapse
|
45
|
Copf T. Developmental shaping of dendritic arbors in Drosophila relies on tightly regulated intra-neuronal activity of protein kinase A (PKA). Dev Biol 2014; 393:282-297. [PMID: 25017992 DOI: 10.1016/j.ydbio.2014.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 06/30/2014] [Accepted: 07/03/2014] [Indexed: 11/24/2022]
Abstract
Dendrites develop morphologies characterized by multiple levels of complexity that involve neuron type specific dendritic length and particular spatial distribution. How this is developmentally regulated and in particular which signaling molecules are crucial in the process is still not understood. Using Drosophila class IV dendritic arborization (da) neurons we test in vivo the effects of cell-autonomous dose-dependent changes in the activity levels of the cAMP-dependent Protein Kinase A (PKA) on the formation of complex dendritic arbors. We find that genetic manipulations of the PKA activity levels affect profoundly the arbor complexity with strongest impact on distal branches. Both decreasing and increasing PKA activity result in a reduced complexity of the arbors, as reflected in decreased dendritic length and number of branching points, suggesting an inverted U-shape response to PKA. The phenotypes are accompanied by changes in organelle distribution: Golgi outposts and early endosomes in distal dendritic branches are reduced in PKA mutants. By using Rab5 dominant negative we find that PKA interacts genetically with the early endosomal pathway. We test if the possible relationship between PKA and organelles may be the result of phosphorylation of the microtubule motor dynein components or Rab5. We find that Drosophila cytoplasmic dynein components are direct PKA phosphorylation targets in vitro, but not in vivo, thus pointing to a different putative in vivo target. Our data argue that tightly controlled dose-dependent intra-neuronal PKA activity levels are critical in determining the dendritic arbor complexity, one of the possible ways being through the regulation of organelle distribution.
Collapse
Affiliation(s)
- Tijana Copf
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, 630 W. 168th St. P&S 12-403, NY 10032, USA; Institute of Molecular Biology and Biotechnology, Nikolaou Plastira 100, P.O Box 1385, GR-70013 Heraklion, Crete, Greece.
| |
Collapse
|
46
|
Pathak GK, Aranda-Espinoza H, Shah SB. Mouse hippocampal explant culture system to study isolated axons. J Neurosci Methods 2014; 232:157-64. [PMID: 24861423 DOI: 10.1016/j.jneumeth.2014.05.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/10/2014] [Accepted: 05/14/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Studies of neuronal regeneration require examination of axons independently of their cell bodies. Several effective strategies have been deployed to compartmentalize long axons of the peripheral nervous system (PNS). However, current strategies to compartmentalize axons of the central nervous system (CNS) may be limited by physical damage to cells during tissue dissociation or slicing, perturbation of three-dimensional tissue architecture, or insufficient axonal tissue for biological analysis. NEW METHODS We developed a novel mouse neonate whole-hippocampus explant culture system, to probe neuronal regeneration in the central nervous system. This system enables imaging, biological, and biophysical analysis of isolated axons. RESULTS We validated this model by isolating pure axonal populations. Additionally, cells within the explant were viable and amenable to transfection. We implemented the explant system to characterize axonal outgrowth following crush injury to the explant at the time of harvest, and also a secondary axonal transection injury 2 days post-culture. The initial crush injury delayed axonal outgrowth; however, axotomy did not alter rates of outgrowth up to 1h post-injury, with or without initial tissue crush injury. COMPARISON WITH EXISTING METHODS Our explant system addresses shortcomings of other strategies developed to compartmentalize CNS axons. It provides a simple method to examine axonal activity and function without requiring additional equipment to slice tissue or segregate axons. CONCLUSION Our hippocampal explant model may be used to study axonal response to injury. We have demonstrated the feasibility of probing axonal biology, biochemistry, and outgrowth free from confounding effects of neuronal cell bodies.
Collapse
Affiliation(s)
- Gunja K Pathak
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Helim Aranda-Espinoza
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Sameer B Shah
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States; Departments of Orthopaedic Surgery and Bioengineering, University of California, San Diego, La Jolla, CA, United States.
| |
Collapse
|
47
|
Zschätzsch M, Oliva C, Langen M, De Geest N, Ozel MN, Williamson WR, Lemon WC, Soldano A, Munck S, Hiesinger PR, Sanchez-Soriano N, Hassan BA. Regulation of branching dynamics by axon-intrinsic asymmetries in Tyrosine Kinase Receptor signaling. eLife 2014; 3:e01699. [PMID: 24755286 PMCID: PMC3990184 DOI: 10.7554/elife.01699] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Axonal branching allows a neuron to connect to several targets, increasing neuronal circuit complexity. While axonal branching is well described, the mechanisms that control it remain largely unknown. We find that in the Drosophila CNS branches develop through a process of excessive growth followed by pruning. In vivo high-resolution live imaging of developing brains as well as loss and gain of function experiments show that activation of Epidermal Growth Factor Receptor (EGFR) is necessary for branch dynamics and the final branching pattern. Live imaging also reveals that intrinsic asymmetry in EGFR localization regulates the balance between dynamic and static filopodia. Elimination of signaling asymmetry by either loss or gain of EGFR function results in reduced dynamics leading to excessive branch formation. In summary, we propose that the dynamic process of axon branch development is mediated by differential local distribution of signaling receptors. DOI:http://dx.doi.org/10.7554/eLife.01699.001 In the human brain, 100 billion neurons form 100 trillion connections. Each neuron consists of a cell body with numerous small branch-like projections known as dendrites (from the Greek word for ‘tree’), plus a long cable-like structure called the axon. Neurons receive electrical inputs from neighboring cells via their dendrites, and then relay these signals onto other cells in their network via their axons. The development of the brain relies on new neurons integrating successfully into existing networks. Axon branching helps with this by enabling a single neuron to establish connections with several cells, but it is unclear how individual neurons decide when and where to form branches. Now, Zschätzsch et al. have revealed the mechanism behind this process in the fruit fly, Drosophila. Mutant flies that lack a protein called EGFR produce abnormal numbers of axon branches, suggesting that this molecule regulates branch formation. Indeed in fruit flies, just as in mammals, the developing brain initially produces excessive numbers of branches, which are subsequently pruned to leave only those that have formed appropriate connections. In Drosophila, an uneven distribution of EGFR between branches belonging to the same axon acts as a signal to regulate this pruning process. To examine this mechanism in more detail, high-resolution four-dimensional imaging was used to study brains that had been removed from Drosophila pupae and kept alive in special culture chambers. Axon branching and loss could now be followed in real time, and were found to occur more slowly in brains that lacked EGFR. The receptor controlled the branching of axons by influencing the distribution of another protein called actin, which is a key component of the internal skeleton that gives cells their structure. In addition to providing new insights into a fundamental aspect of brain development, the work of Zschätzsch et al. also highlights the importance of stochastic events in shaping the network of connections within the developing brain. These findings may well be relevant to ongoing efforts to map the human brain ‘connectome’. DOI:http://dx.doi.org/10.7554/eLife.01699.002
Collapse
Affiliation(s)
- Marlen Zschätzsch
- Center for the Biology of Disease, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Fang Y, Soares L, Bonini NM. Design and implementation of in vivo imaging of neural injury responses in the adult Drosophila wing. Nat Protoc 2013; 8:810-9. [PMID: 23589940 DOI: 10.1038/nprot.2013.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Live-imaging technology has markedly advanced in the field of neural injury and axon degeneration; however, studies are still predominantly performed in in vitro settings such as cultured neuronal cells or in model organisms such as Caenorhabditis elegans in which axons lack glial wrappings. We recently developed a new in vivo model for adult-stage neural injury in Drosophila melanogaster, using the highly accessible wing of the animal. Because the Drosophila wing is translucent and dispensable for survival, it allows clear and direct visualization of injury-induced progressive responses of axons and glia highlighted by fluorescent protein (FP) markers in live animals over time. Moreover, unlike previous Drosophila models of neural injury, this procedure does not require dissection of the CNS. Thus, the key preparation steps for in vivo imaging of the neural injury response described in this protocol can be completed within 30 min.
Collapse
Affiliation(s)
- Yanshan Fang
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
49
|
Lakes-Harlan R. Lesion-induced insights in the plasticity of the insect auditory system. Front Physiol 2013; 4:48. [PMID: 23986709 PMCID: PMC3750944 DOI: 10.3389/fphys.2013.00048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 02/28/2013] [Indexed: 12/04/2022] Open
Abstract
The auditory networks of Orthoptera offer a model system uniquely suited to the study of neuronal connectivity and lesion-dependent neural plasticity. Monaural animals, following the permanent removal of one ear in nymphs or adults, adjust their auditory pathways by collateral sprouting of afferents and deafferented interneurons which connect to neurons on the contralateral side. Transient lesion of the auditory nerve allows us to study regeneration as well as plasticity processes. After crushing the peripheral auditory nerve, the lesioned afferents regrow and re-establish new synaptic connections which are relevant for auditory behavior. During this process collateral sprouting occurs in the central nervous networks, too. Interestingly, after regeneration a changed neuronal network will be maintained. These paradigms are now been used to analyze molecular mechanism in neuronal plasticity on the level of single neurons and small networks.
Collapse
Affiliation(s)
- Reinhard Lakes-Harlan
- AG Integrative Sensory Physiology, Institute for Animal Physiology, Justus-Liebig-University Gießen Gießen, Germany
| |
Collapse
|
50
|
Saijilafu, Zhang BY, Zhou FQ. Signaling pathways that regulate axon regeneration. Neurosci Bull 2013; 29:411-20. [PMID: 23846598 DOI: 10.1007/s12264-013-1357-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/25/2013] [Indexed: 10/26/2022] Open
Abstract
Neurons in the mammalian central nervous system (CNS) cannot regenerate axons after injury. in contrast, neurons in the mammalian peripheral nervous system and in some non-mammalian models, such as C. elegans and Drosophila, are able to regrow axons. Understanding the molecular mechanisms by which these neurons support axon regeneration will help us find ways to enhance mammalian CNS axon regeneration. Here, recent studies in which signaling pathways regulating naturally-occurring axon regeneration that have been identified are reviewed, focusing on how these pathways control gene expression and growth-cone function during axon regeneration.
Collapse
Affiliation(s)
- Saijilafu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | | |
Collapse
|