1
|
Egawa R, Yawo H, Kuba H. Activity-dependent refinement of axonal projections forms one-to-one connection pattern in the developing chick ciliary ganglion. Front Cell Neurosci 2025; 19:1560402. [PMID: 40271539 PMCID: PMC12014593 DOI: 10.3389/fncel.2025.1560402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Although it is well established that initially overproduced synaptic connections are extensively remodeled through activity-dependent competition for postsynaptic innervation, the mechanisms determining the final number of postsynaptic targets per axon remain unclear. Here, we investigated the morphology of individual axonal projections during development and the influence of neural activity in the chick ciliary ganglion (CG), a traditional model system for synapse maturation. By single-axon tracing combining Brainbow labeling and tissue clearing, we revealed that by embryonic day 14 (E14), hundreds of preganglionic axons each establish a one-to-one synaptic connection with single CG neurons via a calyx-type presynaptic terminal enveloping the soma of its postsynaptic target. This homogeneous connection pattern emerged through presynaptic terminal maturation from bouton-like to calyx-like morphology and concurrent axonal branch pruning starting around E10. The calyx maturation was retarded by the presynaptic expression of genetically encoded tools for silencing neuronal activity, enhanced tetanus neurotoxin light chain (eTeNT) or Kir2.1, demonstrating the activity-dependence of this morphological refinement. These findings suggest that some presynaptic mechanisms as well as synaptic competition would operate to restrict the number of postsynaptic targets innervated by each axon in the CG. Together with the easy accessibility to single-axon tracing, our results highlight the potential of the chick CG as a model for investigating the presynaptic factors underlying circuit remodeling.
Collapse
Affiliation(s)
- Ryo Egawa
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hiromu Yawo
- Department of Developmental Biology and Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
2
|
Rekha A, Afzal M, Babu MA, Menon SV, Nathiya D, Supriya S, Mishra SB, Gupta S, Goyal K, Rana M, Ali H, Imran M. GSK-3β dysregulation in aging: Implications for tau pathology and Alzheimer's disease progression. Mol Cell Neurosci 2025; 133:104005. [PMID: 40120784 DOI: 10.1016/j.mcn.2025.104005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/05/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
The role of glycogen synthase kinase-3β (GSK-3β) in the pathogenesis of Alzheimer's disease (AD) is critical for linking amyloid-beta (Aβ) and Tau pathology. The activity of GSK-3β is dysregulated in the regulation of Tau hyperphosphorylation, formation of neurofibrillary tangles (NFTs), and production of Aβ by modulating amyloid precursor protein (APP) processing. This review discusses the mechanisms controlling GSK-3β dysregulation in aging and its influence on AD progression, focusing on the role of neuroinflammation, oxidative stress, and defective signaling pathways, including PI3K/Akt and Wnt. Critical analysis is presented for therapeutic strategies targeting GSK-3β using natural compounds (e.g., curcumin, geniposide) and emerging approaches such as TREM2 modulation and miRNA therapies. In preclinical models, these interventions promise to reduce Tau hyperphosphorylation and Aβ burden, along with associated neurodegeneration. Nevertheless, achieving selective GSK-3β inhibition and optimizing drug delivery are still critical barriers to clinical translation. This review underscores the central role of GSK-3β in AD pathogenesis to highlight its potential as a multifaceted therapeutic target of an innovative strategy for treating this complex neurodegenerative disease.
Collapse
Affiliation(s)
- A Rekha
- D.Y.Patil Medical College, Hospital and Research centre, Pimpri, Pune, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Nathiya
- NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - S Supriya
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Shakti Bedanta Mishra
- Department of Anaesthesiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar 73213, Saudi Arabia
| |
Collapse
|
3
|
Costa AC, Murillo BR, Bessa R, Ribeiro R, Ferreira da Silva T, Porfírio-Rodrigues P, Martins GG, Brites P, Kneussel M, Misgeld T, Brill MS, Sousa MM. Axon-specific microtubule regulation drives asymmetric regeneration of sensory neuron axons. eLife 2025; 13:RP104069. [PMID: 39992313 PMCID: PMC11850000 DOI: 10.7554/elife.104069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
Sensory dorsal root ganglion (DRG) neurons have a unique pseudo-unipolar morphology in which a stem axon bifurcates into a peripheral and a central axon, with different regenerative abilities. Whereas peripheral DRG axons regenerate, central axons are unable to regrow. Central axon regeneration can however be elicited by a prior conditioning lesion to the peripheral axon. How DRG axon asymmetry is established remains unknown. Here we developed a rodent in vitro system replicating DRG pseudo-unipolarization and asymmetric axon regeneration. Using this model, we observed that from early development, central DRG axons have a higher density of growing microtubules. This asymmetry was also present in vivo and was abolished by a conditioning lesion that decreased microtubule polymerization of central DRG axons. An axon-specific microtubule-associated protein (MAP) signature, including the severases spastin and katanin and the microtubule regulators CRMP5 and tau, was found and shown to adapt upon conditioning lesion. Supporting its significance, interfering with the DRG MAP signature either in vitro or in vivo readily abolished central-peripheral asymmetries in microtubule dynamics and regenerative ability. In summary, our data unveil that axon-specific microtubule regulation drives asymmetric regeneration of sensory neuron axons.
Collapse
Affiliation(s)
- Ana Catarina Costa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of PortoPortoPortugal
- Graduate Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar, University of PortoPortoPortugal
| | - Blanca R Murillo
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of PortoPortoPortugal
| | - Rita Bessa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of PortoPortoPortugal
| | - Ricardo Ribeiro
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of PortoPortoPortugal
| | - Tiago Ferreira da Silva
- Neurolipid Biology Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of PortoPortoPortugal
| | | | - Gabriel G Martins
- Advanced Imaging Unit, Instituto Gulbenkian de CiênciaLisboaPortugal
| | - Pedro Brites
- Neurolipid Biology Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of PortoPortoPortugal
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg, ZMNH, University Medical Centre Hamburg-EppendorfHamburgGermany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Monica M Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of PortoPortoPortugal
| |
Collapse
|
4
|
Imai T. Activity-dependent synaptic competition and dendrite pruning in developing mitral cells. Front Neural Circuits 2025; 19:1541926. [PMID: 40034992 PMCID: PMC11873734 DOI: 10.3389/fncir.2025.1541926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/10/2025] [Indexed: 03/05/2025] Open
Abstract
During the early postnatal period, neurons in sensory circuits dynamically remodel their connectivity to acquire discrete receptive fields. Neuronal activity is thought to play a central role in circuit remodeling during this period: Neuronal activity stabilizes some synaptic connections while eliminating others. Synaptic competition plays a central role in the binary choice between stabilization and elimination. While activity-dependent "punishment signals" propagating from winner to loser synapses have been hypothesized to drive synapse elimination, their exact nature has remained elusive. In this review, I summarize recent studies in mouse mitral cells that explain how only one dendrite is stabilized while others are eliminated, based on early postnatal spontaneous activity in the olfactory bulb. I discuss how the hypothetical punishment signals act on loser but not winner dendrites to establish only one primary dendrite per mitral cell, the anatomical basis for the odorant receptor-specific parallel information processing in the olfactory bulb.
Collapse
Affiliation(s)
- Takeshi Imai
- Department of Developmental Neurophysiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Liu S, Alexander KD, Francis MM. Neural Circuit Remodeling: Mechanistic Insights from Invertebrates. J Dev Biol 2024; 12:27. [PMID: 39449319 PMCID: PMC11503349 DOI: 10.3390/jdb12040027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
As nervous systems mature, neural circuit connections are reorganized to optimize the performance of specific functions in adults. This reorganization of connections is achieved through a remarkably conserved phase of developmental circuit remodeling that engages neuron-intrinsic and neuron-extrinsic molecular mechanisms to establish mature circuitry. Abnormalities in circuit remodeling and maturation are broadly linked with a variety of neurodevelopmental disorders, including autism spectrum disorders and schizophrenia. Here, we aim to provide an overview of recent advances in our understanding of the molecular processes that govern neural circuit remodeling and maturation. In particular, we focus on intriguing mechanistic insights gained from invertebrate systems, such as the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster. We discuss how transcriptional control mechanisms, synaptic activity, and glial engulfment shape specific aspects of circuit remodeling in worms and flies. Finally, we highlight mechanistic parallels across invertebrate and mammalian systems, and prospects for further advances in each.
Collapse
Affiliation(s)
- Samuel Liu
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Program in Neuroscience, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kellianne D. Alexander
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Program in Neuroscience, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Michael M. Francis
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Program in Neuroscience, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
6
|
Aiken J, Holzbaur ELF. Spastin locally amplifies microtubule dynamics to pattern the axon for presynaptic cargo delivery. Curr Biol 2024; 34:1687-1704.e8. [PMID: 38554708 PMCID: PMC11042977 DOI: 10.1016/j.cub.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/10/2024] [Accepted: 03/08/2024] [Indexed: 04/02/2024]
Abstract
Neurons rely on the long-range trafficking of synaptic components to form and maintain the complex neural networks that encode the human experience. With a single neuron capable of forming thousands of distinct en passant synapses along its axon, spatially precise delivery of the necessary synaptic components is paramount. How these synapses are patterned, as well as how the efficient delivery of synaptic components is regulated, remains largely unknown. Here, we reveal a novel role for the microtubule (MT)-severing enzyme spastin in locally enhancing MT polymerization to influence presynaptic cargo pausing and retention along the axon. In human neurons derived from induced pluripotent stem cells (iPSCs), we identify sites stably enriched for presynaptic components along the axon prior to the robust assembly of mature presynapses apposed by postsynaptic contacts. These sites are capable of cycling synaptic vesicles, are enriched with spastin, and are hotspots for new MT growth and synaptic vesicle precursor (SVP) pausing/retention. The disruption of neuronal spastin level or activity, by CRISPRi-mediated depletion, transient overexpression, or pharmacologic inhibition of enzymatic activity, interrupts the localized enrichment of dynamic MT plus ends and diminishes SVP accumulation. Using an innovative human heterologous synapse model, where microfluidically isolated human axons recognize and form presynaptic connections with neuroligin-expressing non-neuronal cells, we reveal that neurons deficient for spastin do not achieve the same level of presynaptic component accumulation as control neurons. We propose a model where spastin acts locally as an amplifier of MT polymerization to pattern specific regions of the axon for synaptogenesis and guide synaptic cargo delivery.
Collapse
Affiliation(s)
- Jayne Aiken
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
7
|
Brill MS, Fassier C, Song Y. Editorial: Cytoskeletal alterations in aging and disease. Front Cell Dev Biol 2024; 11:1359465. [PMID: 38299006 PMCID: PMC10828968 DOI: 10.3389/fcell.2023.1359465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 02/02/2024] Open
Affiliation(s)
- Monika S. Brill
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Coralie Fassier
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Yuyu Song
- Department of Neurology, Harvard Medical School, Boston, MA, United States
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
8
|
Ho KH, Candat A, Scarpetta V, Faucourt M, Weill S, Salio C, D'Este E, Meschkat M, Wurm CA, Kneussel M, Janke C, Magiera MM, Genovesio A, Meunier A, Sassoè-Pognetto M, Brill MS, Spassky N, Patrizi A. Choroid plexuses carry nodal-like cilia that undergo axoneme regression from early adult stage. Dev Cell 2023; 58:2641-2651.e6. [PMID: 37890489 DOI: 10.1016/j.devcel.2023.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/06/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023]
Abstract
Choroid plexuses (ChPs) produce cerebrospinal fluid and sense non-cell-autonomous stimuli to control the homeostasis of the central nervous system. They are mainly composed of epithelial multiciliated cells, whose development and function are still controversial. We have thus characterized the stepwise order of mammalian ChP epithelia cilia formation using a combination of super-resolution-microscopy approaches and mouse genetics. We show that ChP ciliated cells are built embryonically on a treadmill of spatiotemporally regulated events, starting with atypical centriole amplification and ending with the construction of nodal-like 9+0 cilia, characterized by both primary and motile features. ChP cilia undergo axoneme resorption at early postnatal stages through a microtubule destabilization process controlled by the microtubule-severing enzyme spastin and mitigated by polyglutamylation levels. Notably, this phenotype is preserved in humans, suggesting a conserved ciliary resorption mechanism in mammals.
Collapse
Affiliation(s)
- Kim Hoa Ho
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg 69120, Germany
| | - Adrien Candat
- Electron Microscopy Facility, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Valentina Scarpetta
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurosciences "Rita Levi Montalcini," University of Turin, Turin 10126, Italy
| | - Marion Faucourt
- Cilia biology and Neurogenesis Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Solene Weill
- Computational Bioimaging and Bioinformatics Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Chiara Salio
- Department of Veterinary Sciences, University of Turin, Grugliasco 10095, Italy
| | - Elisa D'Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | | | | | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR 3348, Orsay 91401, France; Université Paris-Saclay, CNRS UMR 3348, Orsay 91401, France
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR 3348, Orsay 91401, France; Université Paris-Saclay, CNRS UMR 3348, Orsay 91401, France
| | - Auguste Genovesio
- Computational Bioimaging and Bioinformatics Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Alice Meunier
- Cilia biology and Neurogenesis Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Marco Sassoè-Pognetto
- Department of Neurosciences "Rita Levi Montalcini," University of Turin, Turin 10126, Italy
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich 80802, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany
| | - Nathalie Spassky
- Cilia biology and Neurogenesis Team, Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, Paris 75005, France
| | - Annarita Patrizi
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Interdisciplinary Center for Neuroscience, Heidelberg University, Heidelberg 69120, Germany; Zentrum für Molekulare Biologie der Universität Heidelberg, DKFZ-ZMBH Alliance, Heidelberg 69120, Germany.
| |
Collapse
|
9
|
Dey S, Barkai O, Gokhman I, Suissa S, Haffner-Krausz R, Wigoda N, Feldmesser E, Ben-Dor S, Kovalenko A, Binshtok A, Yaron A. Kinesin family member 2A gates nociception. Cell Rep 2023; 42:113257. [PMID: 37851573 DOI: 10.1016/j.celrep.2023.113257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/23/2023] [Accepted: 09/27/2023] [Indexed: 10/20/2023] Open
Abstract
Nociceptive axons undergo remodeling as they innervate their targets during development and in response to environmental insults and pathological conditions. How is nociceptive morphogenesis regulated? Here, we show that the microtubule destabilizer kinesin family member 2A (Kif2a) is a key regulator of nociceptive terminal structures and pain sensitivity. Ablation of Kif2a in sensory neurons causes hyperinnervation and hypersensitivity to noxious stimuli in young adult mice, whereas touch sensitivity and proprioception remain unaffected. Computational modeling predicts that structural remodeling is sufficient to explain the phenotypes. Furthermore, Kif2a deficiency triggers a transcriptional response comprising sustained upregulation of injury-related genes and homeostatic downregulation of highly specific channels and receptors at the late stage. The latter effect can be predicted to relieve the hyperexcitability of nociceptive neurons, despite persisting morphological aberrations, and indeed correlates with the resolution of pain hypersensitivity. Overall, we reveal a critical control node defining nociceptive terminal structure, which is regulating nociception.
Collapse
Affiliation(s)
- Swagata Dey
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Omer Barkai
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem 91120, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Irena Gokhman
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sapir Suissa
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rebecca Haffner-Krausz
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Noa Wigoda
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ester Feldmesser
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Shifra Ben-Dor
- Bioinformatics Unit, Life Science Core Facilities, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Andrew Kovalenko
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah School of Medicine, Jerusalem 91120, Israel; The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avraham Yaron
- Department of Biomolecular Sciences and Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
10
|
Dan L, Zhang Z. Alzheimer's disease: an axonal injury disease? Front Aging Neurosci 2023; 15:1264448. [PMID: 37927337 PMCID: PMC10620718 DOI: 10.3389/fnagi.2023.1264448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/14/2023] [Indexed: 11/07/2023] Open
Abstract
Alzheimer's disease (AD) is the primary cause of dementia and is anticipated to impose a substantial economic burden in the future. Over a significant period, the widely accepted amyloid cascade hypothesis has guided research efforts, and the recent FDA approval of an anti- amyloid-beta (Aβ) protofibrils antibody, believed to decelerate AD progression, has further solidified its significance. However, the excessive emphasis placed on the amyloid cascade hypothesis has overshadowed the physiological nature of Aβ and tau proteins within axons. Axons, specialized neuronal structures, sustain damage during the early stages of AD, exerting a pivotal influence on disease progression. In this review, we present a comprehensive summary of the relationship between axonal damage and AD pathology, amalgamating the physiological roles of Aβ and tau proteins, along with the impact of AD risk genes such as APOE and TREM2. Furthermore, we underscore the exceptional significance of axonal damage in the context of AD.
Collapse
Affiliation(s)
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
11
|
Guan PP, Ge TQ, Wang P. As a Potential Therapeutic Target, C1q Induces Synapse Loss Via Inflammasome-activating Apoptotic and Mitochondria Impairment Mechanisms in Alzheimer's Disease. J Neuroimmune Pharmacol 2023; 18:267-284. [PMID: 37386257 DOI: 10.1007/s11481-023-10076-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 06/16/2023] [Indexed: 07/01/2023]
Abstract
C1q, the initiator of the classical pathway of the complement system, is activated during Alzheimer's disease (AD) development and progression and is especially associated with the production and deposition of β-amyloid protein (Aβ) and phosphorylated tau in β-amyloid plaques (APs) and neurofibrillary tangles (NFTs). Activation of C1q is responsible for induction of synapse loss, leading to neurodegeneration in AD. Mechanistically, C1q could activate glial cells, which results in the loss of synapses via regulation of synapse pruning and phagocytosis in AD. In addition, C1q induces neuroinflammation by inducing proinflammatory cytokine secretion, which is partially mediated by inflammasome activation. Activation of inflammasomes might mediate the effects of C1q on induction of synapse apoptosis. On the other hand, activation of C1q impairs mitochondria, which hinders the renovation and regeneration of synapses. All these actions of C1q contribute to the loss of synapses during neurodegeneration in AD. Therefore, pharmacological, or genetic interventions targeting C1q may provide potential therapeutic strategies for combating AD.
Collapse
Affiliation(s)
- Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China
| | - Tong-Qi Ge
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China.
| |
Collapse
|
12
|
Aiken J, Holzbaur ELF. Spastin locally amplifies microtubule dynamics to pattern the axon for presynaptic cargo delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.08.552320. [PMID: 37609249 PMCID: PMC10441300 DOI: 10.1101/2023.08.08.552320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Neurons rely on long-range trafficking of synaptic components to form and maintain the complex neural networks that encode the human experience. With a single neuron capable of forming thousands of distinct en passant synapses along its axon, spatially precise delivery of the necessary synaptic components is paramount. How these synapses are patterned, and how efficient delivery of synaptic components is regulated, remains largely unknown. Here, we reveal a novel role for the microtubule severing enzyme spastin in locally enhancing microtubule polymerization to influence presynaptic cargo pausing and retention along the axon. In human neurons derived from induced pluripotent stem cells (iPSCs), we identify sites stably enriched for presynaptic components, termed 'protosynapses', which are distributed along the axon prior to the robust assembly of mature presynapses apposed by postsynaptic contacts. These sites are capable of cycling synaptic vesicles, are enriched with spastin, and are hotspots for new microtubule growth and synaptic vesicle precursor (SVP) pausing/retention. Disruption of neuronal spastin, either by CRISPRi-mediated depletion or transient overexpression, interrupts the localized enrichment of dynamic microtubule plus ends and diminishes SVP accumulation. Using an innovative human heterologous synapse model, where microfluidically isolated human axons recognize and form presynaptic connections with neuroligin-expressing non-neuronal cells, we reveal that neurons deficient for spastin do not achieve the same level of presynaptic component accumulation as control neurons. We propose a model where spastin acts locally as an amplifier of microtubule polymerization to pattern specific regions of the axon for synaptogenesis and guide synaptic cargo delivery.
Collapse
Affiliation(s)
- Jayne Aiken
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
14
|
Imanaka C, Shimada S, Ito S, Kamada M, Iguchi T, Konishi Y. A model for generating differences in microtubules between axonal branches depending on the distance from terminals. Brain Res 2023; 1799:148166. [PMID: 36402177 DOI: 10.1016/j.brainres.2022.148166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/11/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
In the remodeling of axonal arbor, the growth and retraction of branches are differentially regulated within a single axon. Although cell-autonomously generated differences in microtubule (MT) turnover are thought to be involved in selective branch regulation, the cellular system whereby neurons generate differences of MTs between axonal branches has not been clarified. Because MT turnover tends to be slower in longer branches compared with neighboring shorter branches, feedback regulation depending on branch length is thought to be involved. In the present study, we generated a model of MT lifetime in axonal terminal branches by adapting a length-dependent model in which parameters for MT dynamics were constant in the arbor. The model predicted that differences in MT lifetime between neighboring branches could be generated depending on the distance from terminals. In addition, the following points were predicted. Firstly, destabilization of MTs throughout the arbor decreased the differences in MT lifetime between branches. Secondly, differences of MT lifetime existed even before MTs entered the branch point. In axonal MTs in primary neurons, treatment with a low concentration of nocodazole significantly decreased the differences of detyrosination (deTyr) and tyrosination (Tyr) of tubulins, indicators of MT turnover. Expansion microscopy of the axonal shaft before the branch point revealed differences in deTyr/Tyr modification on MTs. Our model recapitulates the differences in MT turnover between branches and provides a feedback mechanism for MT regulation that depends on the axonal arbor geometry.
Collapse
Affiliation(s)
- Chiaki Imanaka
- Department of Applied Chemistry and Biotechnology, Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Satoshi Shimada
- Department of Human and Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Shino Ito
- Department of Applied Chemistry and Biotechnology, Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Marina Kamada
- Department of Applied Chemistry and Biotechnology, Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan
| | - Tokuichi Iguchi
- Department of Applied Chemistry and Biotechnology, Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan; Department of Nursing, Faculty of Health Science, Fukui Health Science University, Fukui 910-3190, Japan
| | - Yoshiyuki Konishi
- Department of Applied Chemistry and Biotechnology, Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui 910-8507, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-8507, Japan.
| |
Collapse
|
15
|
Peotter JL, Pustova I, Lettman MM, Shatadal S, Bradberry MM, Winter-Reed AD, Charan M, Sharkey EE, Alvin JR, Bren AM, Oie AK, Chapman ER, Salamat MS, Audhya A. TFG regulates secretory and endosomal sorting pathways in neurons to promote their activity and maintenance. Proc Natl Acad Sci U S A 2022; 119:e2210649119. [PMID: 36161950 PMCID: PMC9546632 DOI: 10.1073/pnas.2210649119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/18/2022] [Indexed: 02/01/2023] Open
Abstract
Molecular pathways that intrinsically regulate neuronal maintenance are poorly understood, but rare pathogenic mutations that underlie neurodegenerative disease can offer important insights into the mechanisms that facilitate lifelong neuronal function. Here, we leverage a rat model to demonstrate directly that the TFG p.R106C variant implicated previously in complicated forms of hereditary spastic paraplegia (HSP) underlies progressive spastic paraparesis with accompanying ventriculomegaly and thinning of the corpus callosum, consistent with disease phenotypes identified in adolescent patients. Analyses of primary cortical neurons obtained from CRISPR-Cas9-edited animals reveal a kinetic delay in biosynthetic secretory protein transport from the endoplasmic reticulum (ER), in agreement with prior induced pluripotent stem cell-based studies. Moreover, we identify an unexpected role for TFG in the trafficking of Rab4A-positive recycling endosomes specifically within axons and dendrites. Impaired TFG function compromises the transport of at least a subset of endosomal cargoes, which we show results in down-regulated inhibitory receptor signaling that may contribute to excitation-inhibition imbalances. In contrast, the morphology and trafficking of other organelles, including mitochondria and lysosomes, are unaffected by the TFG p.R106C mutation. Our findings demonstrate a multifaceted role for TFG in secretory and endosomal protein sorting that is unique to cells of the central nervous system and highlight the importance of these pathways to maintenance of corticospinal tract motor neurons.
Collapse
Affiliation(s)
- Jennifer L. Peotter
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Iryna Pustova
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Molly M. Lettman
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Shalini Shatadal
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Mazdak M. Bradberry
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Allison D. Winter-Reed
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Maya Charan
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Erin E. Sharkey
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - James R. Alvin
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Alyssa M. Bren
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Annika K. Oie
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Edwin R. Chapman
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
- HHMI, University of Wisconsin-Madison, Madison, WI 53705
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705
| | - M. Shahriar Salamat
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| | - Anjon Audhya
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705
| |
Collapse
|
16
|
Transport-dependent maturation of organelles in neurons. Curr Opin Cell Biol 2022; 78:102121. [PMID: 36030563 DOI: 10.1016/j.ceb.2022.102121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/15/2022] [Indexed: 01/31/2023]
Abstract
Some organelles show a spatial gradient of maturation along the neuronal process where more mature organelles are found closer to the cell body. This gradient is set up by progressive maturation steps that are aided by differential organelle distribution as well as transport. Autophagosomes and endosomes mature as they acquire lysosomal membrane proteins and decrease their luminal pH as they are retrogradely transported towards the cell body. The acquisition of lysosomal proteins along the neuronal processes likely occurs through fusion or membrane exchange events with Golgi-derived donor transport carriers that are transported anterogradely from the cell body. The mechanisms by which endosomes and autophagosomes mature might be applicable to other organelles that are transported along neuronal processes. Defects in axonal transport may also contribute to the accumulation of immature organelles in neurons. Such accumulations have been seen in neurons of neurodegenerative models.
Collapse
|
17
|
Costa AC, Sousa MM. The Role of Spastin in Axon Biology. Front Cell Dev Biol 2022; 10:934522. [PMID: 35865632 PMCID: PMC9294387 DOI: 10.3389/fcell.2022.934522] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/07/2022] [Indexed: 12/05/2022] Open
Abstract
Neurons are highly polarized cells with elaborate shapes that allow them to perform their function. In neurons, microtubule organization—length, density, and dynamics—are essential for the establishment of polarity, growth, and transport. A mounting body of evidence shows that modulation of the microtubule cytoskeleton by microtubule-associated proteins fine tunes key aspects of neuronal cell biology. In this respect, microtubule severing enzymes—spastin, katanin and fidgetin—a group of microtubule-associated proteins that bind to and generate internal breaks in the microtubule lattice, are emerging as key modulators of the microtubule cytoskeleton in different model systems. In this review, we provide an integrative view on the latest research demonstrating the key role of spastin in neurons, specifically in the context of axonal cell biology. We focus on the function of spastin in the regulation of microtubule organization, and axonal transport, that underlie its importance in the intricate control of axon growth, branching and regeneration.
Collapse
Affiliation(s)
- Ana Catarina Costa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
- Graduate Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- *Correspondence: Ana Catarina Costa, ; Monica Mendes Sousa,
| | - Monica Mendes Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação Em Saúde (i3S), University of Porto, Porto, Portugal
- *Correspondence: Ana Catarina Costa, ; Monica Mendes Sousa,
| |
Collapse
|
18
|
Herwerth M, Kenet S, Schifferer M, Winkler A, Weber M, Snaidero N, Wang M, Lohrberg M, Bennett JL, Stadelmann C, Hemmer B, Misgeld T. A new form of axonal pathology in a spinal model of neuromyelitis optica. Brain 2022; 145:1726-1742. [PMID: 35202467 PMCID: PMC9166560 DOI: 10.1093/brain/awac079] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 01/31/2022] [Accepted: 02/12/2022] [Indexed: 11/14/2022] Open
Abstract
Neuromyelitis optica is a chronic neuroinflammatory disease, which primarily targets astrocytes and often results in severe axon injury of unknown mechanism. Neuromyelitis optica patients harbour autoantibodies against the astrocytic water channel protein, aquaporin-4 (AQP4-IgG), which induce complement-mediated astrocyte lysis and subsequent axon damage. Using spinal in vivo imaging in a mouse model of such astrocytopathic lesions, we explored the mechanism underlying neuromyelitis optica-related axon injury. Many axons showed a swift and morphologically distinct 'pearls-on-string' transformation also readily detectable in human neuromyelitis optica lesions, which especially affected small calibre axons independently of myelination. Functional imaging revealed that calcium homeostasis was initially preserved in this 'acute axonal beading' state, ruling out disruption of the axonal membrane, which sets this form of axon injury apart from previously described forms of traumatic and inflammatory axon damage. Morphological, pharmacological and genetic analyses showed that AQP4-IgG-induced axon injury involved osmotic stress and ionic overload, but does not appear to use canonical pathways of Wallerian-like degeneration. Subcellular analysis demonstrated remodelling of the axonal cytoskeleton in beaded axons, especially local loss of microtubules. Treatment with the microtubule stabilizer epothilone, a putative therapy approach for traumatic and degenerative axonopathies, prevented axonal beading, while destabilizing microtubules sensitized axons for beading. Our results reveal a distinct form of immune-mediated axon pathology in neuromyelitis optica that mechanistically differs from known cascades of post-traumatic and inflammatory axon loss, and suggest a new strategy for neuroprotection in neuromyelitis optica and related diseases.
Collapse
Affiliation(s)
- Marina Herwerth
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Selin Kenet
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- Graduate School of Systemic Neurosciences, Ludwig-Maximilians University, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Anne Winkler
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Melanie Weber
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Nicolas Snaidero
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
| | - Melanie Lohrberg
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Jeffrey L. Bennett
- Departments of Neurology and Ophthalmology, Programs in Neuroscience and Immunology, University of Colorado School of Medicine, Aurora, USA
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
19
|
Meltzer H, Schuldiner O. Spatiotemporal Control of Neuronal Remodeling by Cell Adhesion Molecules: Insights From Drosophila. Front Neurosci 2022; 16:897706. [PMID: 35645712 PMCID: PMC9135462 DOI: 10.3389/fnins.2022.897706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/22/2022] [Indexed: 01/26/2023] Open
Abstract
Developmental neuronal remodeling is required for shaping the precise connectivity of the mature nervous system. Remodeling involves pruning of exuberant neural connections, often followed by regrowth of adult-specific ones, as a strategy to refine neural circuits. Errors in remodeling are associated with neurodevelopmental disorders such as schizophrenia and autism. Despite its fundamental nature, our understanding of the mechanisms governing neuronal remodeling is far from complete. Specifically, how precise spatiotemporal control of remodeling and rewiring is achieved is largely unknown. In recent years, cell adhesion molecules (CAMs), and other cell surface and secreted proteins of various families, have been implicated in processes of neurite pruning and wiring specificity during circuit reassembly. Here, we review some of the known as well as speculated roles of CAMs in these processes, highlighting recent advances in uncovering spatiotemporal aspects of regulation. Our focus is on the fruit fly Drosophila, which is emerging as a powerful model in the field, due to the extensive, well-characterized and stereotypic remodeling events occurring throughout its nervous system during metamorphosis, combined with the wide and constantly growing toolkit to identify CAM binding and resulting cellular interactions in vivo. We believe that its many advantages pose Drosophila as a leading candidate for future breakthroughs in the field of neuronal remodeling in general, and spatiotemporal control by CAMs specifically.
Collapse
Affiliation(s)
- Hagar Meltzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
- *Correspondence: Hagar Meltzer,
| | - Oren Schuldiner
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
- Oren Schuldiner,
| |
Collapse
|
20
|
Involvement of the Voltage-Gated Calcium Channels L- P/Q- and N-Types in Synapse Elimination During Neuromuscular Junction Development. Mol Neurobiol 2022; 59:4044-4064. [PMID: 35474562 PMCID: PMC9167222 DOI: 10.1007/s12035-022-02818-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 03/22/2022] [Indexed: 10/26/2022]
Abstract
During the nervous system development, synapses are initially overproduced. In the neuromuscular junction (NMJ) however, competition between several motor nerve terminals and the synapses they made ends with the maturation of only one axon. The competitive signaling between axons is mediated by the differential activity-dependent release of the neurotransmitter ACh, co-transmitters, and neurotrophic factors. A multiple metabotropic receptor-driven downstream balance between PKA and PKC isoforms modulates the phosphorylation of targets involved in transmitter release and nerve terminal stability. Previously, we observed in the weakest endings on the polyinnervated NMJ that M1 mAChR receptors reduce ACh release through the PKC pathway coupled to an excess of Ca2+ inflow through P/Q- N- and L-type voltage-gated calcium channels (VGCC). This signaling would contribute to the elimination of this nerve terminal. Here, we investigate the involvement of the P/Q-, N-, and L-subtype channels in transgenic B6.Cg-Tg (Thy1-YFP)16-Jrs/J mice during synapse elimination. Then, the axon number and postsynaptic receptor cluster morphologic maturation were evaluated. The results show that both L- and P/Q-type VGCC (but not the N-type) are equally involved in synapse elimination. Their normal function favors supernumerary axonal loss by jointly enhancing intracellular calcium [Ca2+]i. The block of these VGCCs or [Ca2+]i i sequestration results in the same delay of axonal loss as the cPKCβI and nPKCε isoform block or PKA activation. The specific block of the muscle cell's contraction with μ-conotoxin GIIIB also delays synapse maturation, and thus, a retrograde influence from the postsynaptic site regulating the presynaptic CaV1.3 may contribute to the synapse elimination.
Collapse
|
21
|
Tymanskyj SR, Curran BM, Ma L. Selective axonal transport through branch junctions is directed by growth cone signaling and mediated by KIF1/kinesin-3 motors. Cell Rep 2022; 39:110748. [PMID: 35476993 PMCID: PMC9097860 DOI: 10.1016/j.celrep.2022.110748] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/27/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Development and function of nerve cells rely on the orchestration of microtubule-based transport from the cell body into distal axonal terminals. Neurons often have highly elaborate branches innervating multiple targets, but how protein or membrane cargos navigate through branch junctions to specific branch targets is unknown. Here, we demonstrate that anterograde transport of membrane vesicles through axonal branch junctions is highly selective, which is influenced by branch length and more strongly by growth cone motility. Using an optogenetic tool, we demonstrate that signaling from the growth cone can rapidly direct transport through branch junctions. We further demonstrate that such transport selectivity is differentially regulated for different vesicles and mediated by the KIF1/kinesin-3 family motors. We propose that this transport regulation through branch junctions could broadly impact neuronal development, function, and regeneration.
Collapse
Affiliation(s)
- Stephen R Tymanskyj
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bridget M Curran
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Le Ma
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sydney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
22
|
Wang M, Misgeld T, Brill MS. Neural labeling and manipulation by neonatal intraventricular viral injection in mice. STAR Protoc 2022; 3:101081. [PMID: 35059654 PMCID: PMC8760487 DOI: 10.1016/j.xpro.2021.101081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This step-by-step protocol provides a fast and easy technique to label and/or genetically manipulate neural cells, achieved by intraventricular injection of viral vectors into neonatal mice under ultrasound guidance. Successful injection of adeno-associated viral vectors (AAV) induces neural transduction as fast as 3 days post injection (dpi) in both the central and peripheral nervous systems. Virally driven expression persists until early adulthood. The same setup enables injection of other viral vectors as well as intramuscular injection. For complete details on the use and execution of this protocol, please refer to Wang et al. (2021) and Brill et al. (2016).
Collapse
Affiliation(s)
- Mengzhe Wang
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, 81377 Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany
| |
Collapse
|
23
|
Piermarini E, Akarsu S, Connors T, Kneussel M, Lane MA, Morfini G, Karabay A, Baas PW, Qiang L. Modeling gain-of-function and loss-of-function components of SPAST-based hereditary spastic paraplegia using transgenic mice. Hum Mol Genet 2021; 31:1844-1859. [PMID: 34935948 PMCID: PMC9169457 DOI: 10.1093/hmg/ddab367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) is a disease in which dieback degeneration of corticospinal tracts, accompanied by axonal swellings, leads to gait deficiencies. SPG4-HSP, the most common form of the disease, results from mutations of human spastin gene (SPAST), which is the gene that encodes spastin, a microtubule-severing protein. The lack of a vertebrate model that recapitulates both the etiology and symptoms of SPG4-HSP has stymied the development of effective therapies for the disease. hSPAST-C448Y mice, which express human mutant spastin at the ROSA26 locus, display corticospinal dieback and gait deficiencies but not axonal swellings. On the other hand, mouse spastin gene (Spast)-knockout (KO) mice display axonal swellings but not corticospinal dieback or gait deficiencies. One possibility is that reduced spastin function, resulting in axonal swellings, is not the cause of the disease but exacerbates the toxic effects of the mutant protein. To explore this idea, Spast-KO and hSPAST-C448Y mice were crossbred, and the offspring were compared with the parental lines via histological and behavioral analyses. The crossbred animals displayed axonal swellings as well as earlier onset, worsened gait deficiencies and corticospinal dieback compared with the hSPAST-C448Y mouse. These results, together with observations on changes in histone deacetylases 6 and tubulin modifications in the axon, indicate that each of these three transgenic mouse lines is valuable for investigating a different component of the disease pathology. Moreover, the crossbred mice are the best vertebrate model to date for testing potential therapies for SPG4-HSP.
Collapse
Affiliation(s)
- Emanuela Piermarini
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Seyma Akarsu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA,Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul 34469, Turkey
| | - Theresa Connors
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Arzu Karabay
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul 34469, Turkey
| | - Peter W Baas
- To whom correspondence should be addressed at: Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA. Tel: +1 2159918311; Fax: +1 2158439082; ; Tel: +1 2159918298;
| | - Liang Qiang
- To whom correspondence should be addressed at: Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA. Tel: +1 2159918311; Fax: +1 2158439082; ; Tel: +1 2159918298;
| |
Collapse
|
24
|
Chen R, Du S, Yao Y, Zhang L, Luo J, Shen Y, Xu Z, Zeng X, Zhang L, Liu M, Yin C, Tang B, Tan J, Xu X, Liu JY. A Novel SPAST Mutation Results in Spastin Accumulation and Defects in Microtubule Dynamics. Mov Disord 2021; 37:598-607. [PMID: 34927746 PMCID: PMC9300132 DOI: 10.1002/mds.28885] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/24/2021] [Accepted: 11/27/2021] [Indexed: 11/25/2022] Open
Abstract
Background Haploinsufficiency is widely accepted as the pathogenic mechanism of spastic paraplegia type 4 (SPG4). However, there are some cases that cannot be explained by reduced function of the spastin protein encoded by SPAST. Objectives To identify the causative gene of autosomal dominant hereditary spastic paraplegia in three large Chinese families and explore the pathological mechanism of a spastin variant. Methods Three large Chinese hereditary spastic paraplegia families with a total of 247 individuals (67 patients) were investigated, of whom 59 members were recruited to the study. Genetic testing was performed to identify the causative gene. Western blotting and immunofluorescence were used to analyze the effects of the mutant proteins in vitro. Results In the three hereditary spastic paraplegia families, of whom three index cases were misdiagnosed as other types of neurological diseases, a novel c.985dupA (p.Met329Asnfs*3) variant in SPAST was identified and was shown to cosegregate with the phenotype in the three families. The c.985dupA mutation produced two truncated mutants (mutant M1 and M87 isoforms) that accumulated to a higher level than their wild‐type counterparts. Furthermore, the mutant M1 isoform heavily decorated the microtubules and rendered them resistant to depolymerization. In contrast, the mutant M87 isoform was diffusely localized in both the nucleus and the cytoplasm, could not decorate microtubules, and was not able to promote microtubule disassembly. Conclusions SPAST mutations leading to premature stop codons do not always act through haploinsufficiency. The truncated spastin may damage the corticospinal tracts through an isoform‐specific toxic effect.
Collapse
Affiliation(s)
- Rui Chen
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Shiyue Du
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Yanyi Yao
- Medical Genetics Center, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Lu Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Junyu Luo
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Yinhua Shen
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Zhenping Xu
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Xiaomei Zeng
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Luoying Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Mugen Liu
- College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Chuang Yin
- Department of Neurology, Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Jun Tan
- Department of Neurology, Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xuan Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jing Yu Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
25
|
Hakanen J, Parmentier N, Sommacal L, Garcia-Sanchez D, Aittaleb M, Vertommen D, Zhou L, Ruiz-Reig N, Tissir F. The Celsr3-Kif2a axis directs neuronal migration in the postnatal brain. Prog Neurobiol 2021; 208:102177. [PMID: 34582949 DOI: 10.1016/j.pneurobio.2021.102177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/12/2021] [Accepted: 09/20/2021] [Indexed: 12/27/2022]
Abstract
The tangential migration of immature neurons in the postnatal brain involves consecutive migration cycles and depends on constant remodeling of the cell cytoskeleton, particularly in the leading process (LP). Despite the identification of several proteins with permissive and empowering functions, the mechanisms that specify the direction of migration remain largely unknown. Here, we report that planar cell polarity protein Celsr3 orients neuroblasts migration from the subventricular zone (SVZ) to olfactory bulb (OB). In Celsr3-forebrain conditional knockout mice, neuroblasts loose directionality and few can reach the OB. Celsr3-deficient neuroblasts exhibit aberrant branching of LP, de novo LP formation, and decreased growth rate of microtubules (MT). Mechanistically, we show that Celsr3 interacts physically with Kif2a, a MT depolymerizing protein and that conditional inactivation of Kif2a in the forebrain recapitulates the Celsr3 knockout phenotype. Our findings provide evidence that Celsr3 and Kif2a cooperatively specify the directionality of neuroblasts tangential migration in the postnatal brain.
Collapse
Affiliation(s)
- Janne Hakanen
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Nicolas Parmentier
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Leonie Sommacal
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Dario Garcia-Sanchez
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Mohamed Aittaleb
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Didier Vertommen
- Université catholique de Louvain, de Duve Institute, Massprot Platform, Brussels, Belgium
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, PR China
| | - Nuria Ruiz-Reig
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium
| | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, Developmental Neurobiology, Brussels, Belgium; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
26
|
Bodakuntla S, Nedozralova H, Basnet N, Mizuno N. Cytoskeleton and Membrane Organization at Axon Branches. Front Cell Dev Biol 2021; 9:707486. [PMID: 34540830 PMCID: PMC8440873 DOI: 10.3389/fcell.2021.707486] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Axon branching is a critical process ensuring a high degree of interconnectivity for neural network formation. As branching occurs at sites distant from the soma, it is necessary that axons have a local system to dynamically control and regulate axonal growth. This machinery depends on the orchestration of cellular functions such as cytoskeleton, subcellular transport, energy production, protein- and membrane synthesis that are adapted for branch formation. Compared to the axon shaft, branching sites show a distinct and dynamic arrangement of cytoskeleton components, endoplasmic reticulum and mitochondria. This review discusses the regulation of axon branching in the context of cytoskeleton and membrane remodeling.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Hana Nedozralova
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Nirakar Basnet
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States.,National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
27
|
Bu S, Yong WL, Lim BJW, Kondo S, Yu F. A systematic analysis of microtubule-destabilizing factors during dendrite pruning in Drosophila. EMBO Rep 2021; 22:e52679. [PMID: 34338441 DOI: 10.15252/embr.202152679] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 11/09/2022] Open
Abstract
It has long been thought that microtubule disassembly, one of the earliest cellular events, contributes to neuronal pruning and neurodegeneration in development and disease. However, how microtubule disassembly drives neuronal pruning remains poorly understood. Here, we conduct a systematic investigation of various microtubule-destabilizing factors and identify exchange factor for Arf6 (Efa6) and Stathmin (Stai) as new regulators of dendrite pruning in ddaC sensory neurons during Drosophila metamorphosis. We show that Efa6 is both necessary and sufficient to regulate dendrite pruning. Interestingly, Efa6 and Stai facilitate microtubule turnover and disassembly prior to dendrite pruning without compromising the minus-end-out microtubule orientation in dendrites. Moreover, our pharmacological and genetic manipulations strongly support a key role of microtubule disassembly in promoting dendrite pruning. Thus, this systematic study highlights the importance of two selective microtubule destabilizers in dendrite pruning and substantiates a causal link between microtubule disassembly and neuronal pruning.
Collapse
Affiliation(s)
- Shufeng Bu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Wei Lin Yong
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore
| | - Bryan Jian Wei Lim
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Shu Kondo
- Invertebrate Genetics Laboratory, National Institute of Genetics, Shizuoka, Japan
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
28
|
Aihara S, Fujimoto S, Sakaguchi R, Imai T. BMPR-2 gates activity-dependent stabilization of primary dendrites during mitral cell remodeling. Cell Rep 2021; 35:109276. [PMID: 34161760 DOI: 10.1016/j.celrep.2021.109276] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 04/28/2021] [Accepted: 05/28/2021] [Indexed: 11/29/2022] Open
Abstract
Developing neurons initially form excessive neurites and then remodel them based on molecular cues and neuronal activity. Developing mitral cells in the olfactory bulb initially extend multiple primary dendrites. They then stabilize single primary dendrites while eliminating others. However, the mechanisms underlying selective dendrite remodeling remain elusive. Using CRISPR-Cas9-based knockout screening combined with in utero electroporation, we identify BMPR-2 as a key regulator for selective dendrite stabilization. Bmpr2 knockout and its rescue experiments show that BMPR-2 inhibits LIMK without ligands and thereby permits dendrite destabilization. In contrast, the overexpression of antagonists and agonists indicates that ligand-bound BMPR-2 stabilizes dendrites, most likely by releasing LIMK. Using genetic and FRET imaging experiments, we demonstrate that free LIMK is activated by NMDARs via Rac1, facilitating dendrite stabilization through F-actin formation. Thus, the selective stabilization of primary dendrites is ensured by concomitant inputs of BMP ligands and neuronal activity.
Collapse
Affiliation(s)
- Shuhei Aihara
- Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Laboratory for Sensory Circuit Formation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Satoshi Fujimoto
- Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Laboratory for Sensory Circuit Formation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Richi Sakaguchi
- Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Laboratory for Sensory Circuit Formation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Takeshi Imai
- Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Laboratory for Sensory Circuit Formation, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
29
|
Morphological Heterogeneity of the Endoplasmic Reticulum within Neurons and Its Implications in Neurodegeneration. Cells 2021; 10:cells10050970. [PMID: 33919188 PMCID: PMC8143122 DOI: 10.3390/cells10050970] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
The endoplasmic reticulum (ER) is a multipurpose organelle comprising dynamic structural subdomains, such as ER sheets and tubules, serving to maintain protein, calcium, and lipid homeostasis. In neurons, the single ER is compartmentalized with a careful segregation of the structural subdomains in somatic and neurite (axodendritic) regions. The distribution and arrangement of these ER subdomains varies between different neuronal types. Mutations in ER membrane shaping proteins and morphological changes in the ER are associated with various neurodegenerative diseases implying significance of ER morphology in maintaining neuronal integrity. Specific neurons, such as the highly arborized dopaminergic neurons, are prone to stress and neurodegeneration. Differences in morphology and functionality of ER between the neurons may account for their varied sensitivity to stress and neurodegenerative changes. In this review, we explore the neuronal ER and discuss its distinct morphological attributes and specific functions. We hypothesize that morphological heterogeneity of the ER in neurons is an important factor that accounts for their selective susceptibility to neurodegeneration.
Collapse
|
30
|
Wang M, Kleele T, Xiao Y, Plucinska G, Avramopoulos P, Engelhardt S, Schwab MH, Kneussel M, Czopka T, Sherman DL, Brophy PJ, Misgeld T, Brill MS. Completion of neuronal remodeling prompts myelination along developing motor axon branches. J Cell Biol 2021; 220:e201911114. [PMID: 33538762 PMCID: PMC7868780 DOI: 10.1083/jcb.201911114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 11/20/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022] Open
Abstract
Neuronal remodeling and myelination are two fundamental processes during neurodevelopment. How they influence each other remains largely unknown, even though their coordinated execution is critical for circuit function and often disrupted in neuropsychiatric disorders. It is unclear whether myelination stabilizes axon branches during remodeling or whether ongoing remodeling delays myelination. By modulating synaptic transmission, cytoskeletal dynamics, and axonal transport in mouse motor axons, we show that local axon remodeling delays myelination onset and node formation. Conversely, glial differentiation does not determine the outcome of axon remodeling. Delayed myelination is not due to a limited supply of structural components of the axon-glial unit but rather is triggered by increased transport of signaling factors that initiate myelination, such as neuregulin. Further, transport of promyelinating signals is regulated via local cytoskeletal maturation related to activity-dependent competition. Our study reveals an axon branch-specific fine-tuning mechanism that locally coordinates axon remodeling and myelination.
Collapse
Affiliation(s)
- Mengzhe Wang
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| | - Tatjana Kleele
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| | - Yan Xiao
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| | - Gabriela Plucinska
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| | - Petros Avramopoulos
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Markus H. Schwab
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Matthias Kneussel
- University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), Institute for Molecular Neurogenetics, Hamburg, Germany
| | - Tim Czopka
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Diane L. Sherman
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Peter J. Brophy
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Monika S. Brill
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
31
|
Haddix SG, Rasband MN. Lose it to use it. J Cell Biol 2021; 220:e202102030. [PMID: 33734302 PMCID: PMC7980256 DOI: 10.1083/jcb.202102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this issue, Wang et al. (2021. J. Cell Biol.https://doi.org/10.1083/jcb.201911114) describe a phenomenon in which neuromuscular junction synapse elimination triggers myelination of terminal motor axon branches. They propose a mechanism initiated by synaptic pruning that depends on synaptic activity, cytoskeletal maturation, and the associated anterograde transport of trophic factors including Neuregulin 1-III.
Collapse
|
32
|
Bodakuntla S, Janke C, Magiera MM. Tubulin polyglutamylation, a regulator of microtubule functions, can cause neurodegeneration. Neurosci Lett 2021; 746:135656. [PMID: 33482309 DOI: 10.1016/j.neulet.2021.135656] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases lead to a progressive demise of neuronal functions that ultimately results in neuronal death. Besides a large variety of molecular pathways that have been linked to the degeneration of neurons, dysfunctions of the microtubule cytoskeleton are common features of many human neurodegenerative disorders. Yet, it is unclear whether microtubule dysfunctions are causative, or mere bystanders in the disease progression. A so-far little explored regulatory mechanism of the microtubule cytoskeleton, the posttranslational modifications of tubulin, emerge as candidate mechanisms involved in neuronal dysfunction, and thus, degeneration. Here we review the role of tubulin polyglutamylation, a prominent modification of neuronal microtubules. We discuss the current understanding of how polyglutamylation controls microtubule functions in healthy neurons, and how deregulation of this modification leads to neurodegeneration in mice and humans.
Collapse
Affiliation(s)
- Satish Bodakuntla
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France
| | - Carsten Janke
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| | - Maria M Magiera
- Institut Curie, PSL Research University, CNRS UMR3348, F-91401 Orsay, France; Université Paris-Saclay, CNRS UMR3348, F-91401 Orsay, France.
| |
Collapse
|
33
|
Chemoradiation impairs myofiber hypertrophic growth in a pediatric tumor model. Sci Rep 2020; 10:19501. [PMID: 33177579 PMCID: PMC7659015 DOI: 10.1038/s41598-020-75913-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/19/2020] [Indexed: 01/05/2023] Open
Abstract
Pediatric cancer treatment often involves chemotherapy and radiation, where off-target effects can include skeletal muscle decline. The effect of such treatments on juvenile skeletal muscle growth has yet to be investigated. We employed a small animal irradiator to administer fractionated hindlimb irradiation to juvenile mice bearing implanted rhabdomyosarcoma (RMS) tumors. Hindlimb-targeted irradiation (3 × 8.2 Gy) of 4-week-old mice successfully eliminated RMS tumors implanted one week prior. After establishment of this preclinical model, a cohort of tumor-bearing mice were injected with the chemotherapeutic drug, vincristine, alone or in combination with fractionated irradiation (5 × 4.8 Gy). Single myofiber analysis of fast-contracting extensor digitorum longus (EDL) and slow-contracting soleus (SOL) muscles was conducted 3 weeks post-treatment. Although a reduction in myofiber size was apparent, EDL and SOL myonuclear number were differentially affected by juvenile irradiation and/or vincristine treatment. In contrast, a decrease in myonuclear domain (myofiber volume/myonucleus) was observed regardless of muscle or treatment. Thus, inhibition of myofiber hypertrophic growth is a consistent feature of pediatric cancer treatment.
Collapse
|
34
|
Kuo YW, Howard J. Cutting, Amplifying, and Aligning Microtubules with Severing Enzymes. Trends Cell Biol 2020; 31:50-61. [PMID: 33183955 PMCID: PMC7749064 DOI: 10.1016/j.tcb.2020.10.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
Microtubule-severing enzymes - katanin, spastin, fidgetin - are related AAA-ATPases that cut microtubules into shorter filaments. These proteins, also called severases, are involved in a wide range of cellular processes including cell division, neuronal development, and tissue morphogenesis. Paradoxically, severases can amplify the microtubule cytoskeleton and not just destroy it. Recent work on spastin and katanin has partially resolved this paradox by showing that these enzymes are strong promoters of microtubule growth. Here, we review recent structural and biophysical advances in understanding the molecular mechanisms of severing and growth promotion that provide insight into how severing enzymes shape microtubule networks.
Collapse
Affiliation(s)
- Yin-Wei Kuo
- Department of Chemistry, Yale University, New Haven, CT 06511, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
35
|
Jiang T, Cai Z, Ji Z, Zou J, Liang Z, Zhang G, Liang Y, Lin H, Tan M. The lncRNA MALAT1/miR-30/Spastin Axis Regulates Hippocampal Neurite Outgrowth. Front Cell Neurosci 2020; 14:555747. [PMID: 33192306 PMCID: PMC7606917 DOI: 10.3389/fncel.2020.555747] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 09/23/2020] [Indexed: 11/13/2022] Open
Abstract
Spastin, a microtubule-severing enzyme, is important for neurite outgrowth. However, the mechanisms underlying the post-transcriptional regulation of spastin during microtubule-related processes are largely unknown. We demonstrated that the spastin expression level is controlled by a long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1)/microRNA-30 (miR-30) axis during neurite outgrowth. The miR-30 expression level decreased in hippocampal neurons with increasing days in culture, and miR-30 overexpression suppressed while miR-30 inhibition promoted neurite outgrowth in hippocampal neurons. Spastin was validated as a target gene of miR-30 using the luciferase reporter assay. The protein expression, microtubule severing activity, and neurite promoting effect of spastin were suppressed by the overexpression of miR-30 mimics and increased by miR-30 inhibitors. MALAT1 expression increased during neurite outgrowth and MALAT1 silencing impaired neurite outgrowth. miR-30 was a sponge target of MALAT1 and MALAT1/miR-30 altered neurite outgrowth in hippocampal neurons. MALAT1 overexpression reversed the inhibitory effect of miR-30 on the activity of a luciferase reporter construct containing spastin, as well as spastin mRNA and protein expression, indicating that spastin was a downstream effector of MALAT1/miR-30. The MALAT1/miR-30 cascade also modulated spastin-induced microtubule severing, and the MALAT1/miR-30/spastin axis regulated neurite outgrowth in hippocampal neurons. This study suggests a new mechanism governing neurite outgrowth in hippocampal neurons involving MALAT1/miR-30-regulated spastin expression.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Department of Orthopaedics, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhenbin Cai
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhisheng Ji
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jianyu Zou
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhi Liang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guowei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yaozhong Liang
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hongsheng Lin
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Minghui Tan
- Department of Orthopaedics, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
36
|
Photoswitchable paclitaxel-based microtubule stabilisers allow optical control over the microtubule cytoskeleton. Nat Commun 2020; 11:4640. [PMID: 32934232 PMCID: PMC7493900 DOI: 10.1038/s41467-020-18389-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 08/19/2020] [Indexed: 02/06/2023] Open
Abstract
Small molecule inhibitors are prime reagents for studies in microtubule cytoskeleton research, being applicable across a range of biological models and not requiring genetic engineering. However, traditional chemical inhibitors cannot be experimentally applied with spatiotemporal precision suiting the length and time scales inherent to microtubule-dependent cellular processes. We have synthesised photoswitchable paclitaxel-based microtubule stabilisers, whose binding is induced by photoisomerisation to their metastable state. Photoisomerising these reagents in living cells allows optical control over microtubule network integrity and dynamics, cell division and survival, with biological response on the timescale of seconds and spatial precision to the level of individual cells within a population. In primary neurons, they enable regulation of microtubule dynamics resolved to subcellular regions within individual neurites. These azobenzene-based microtubule stabilisers thus enable non-invasive, spatiotemporally precise modulation of the microtubule cytoskeleton in living cells, and promise new possibilities for studying intracellular transport, cell motility, and neuronal physiology. Light-based modulation of the microtubule (MT) cytoskeleton is an attractive goal for spatiotemporally-resolved MT studies. Here the authors develop a first generation photoswitchable small molecule MT stabiliser based on paclitaxel, allowing optical control over cellular MT dynamics.
Collapse
|
37
|
Fernandez-Valenzuela JJ, Sanchez-Varo R, Muñoz-Castro C, De Castro V, Sanchez-Mejias E, Navarro V, Jimenez S, Nuñez-Diaz C, Gomez-Arboledas A, Moreno-Gonzalez I, Vizuete M, Davila JC, Vitorica J, Gutierrez A. Enhancing microtubule stabilization rescues cognitive deficits and ameliorates pathological phenotype in an amyloidogenic Alzheimer's disease model. Sci Rep 2020; 10:14776. [PMID: 32901091 PMCID: PMC7479116 DOI: 10.1038/s41598-020-71767-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 08/18/2020] [Indexed: 01/10/2023] Open
Abstract
In Alzheimer's disease (AD), and other tauopathies, microtubule destabilization compromises axonal and synaptic integrity contributing to neurodegeneration. These diseases are characterized by the intracellular accumulation of hyperphosphorylated tau leading to neurofibrillary pathology. AD brains also accumulate amyloid-beta (Aβ) deposits. However, the effect of microtubule stabilizing agents on Aβ pathology has not been assessed so far. Here we have evaluated the impact of the brain-penetrant microtubule-stabilizing agent Epothilone D (EpoD) in an amyloidogenic model of AD. Three-month-old APP/PS1 mice, before the pathology onset, were weekly injected with EpoD for 3 months. Treated mice showed significant decrease in the phospho-tau levels and, more interesting, in the intracellular and extracellular hippocampal Aβ accumulation, including the soluble oligomeric forms. Moreover, a significant cognitive improvement and amelioration of the synaptic and neuritic pathology was found. Remarkably, EpoD exerted a neuroprotective effect on SOM-interneurons, a highly AD-vulnerable GABAergic subpopulation. Therefore, our results suggested that EpoD improved microtubule dynamics and axonal transport in an AD-like context, reducing tau and Aβ levels and promoting neuronal and cognitive protection. These results underline the existence of a crosstalk between cytoskeleton pathology and the two major AD protein lesions. Therefore, microtubule stabilizers could be considered therapeutic agents to slow the progression of both tau and Aβ pathology.
Collapse
Affiliation(s)
- Juan Jose Fernandez-Valenzuela
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Raquel Sanchez-Varo
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Clara Muñoz-Castro
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, C/Prof. Garcia Gonzalez 2, 41012, Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Vanessa De Castro
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain
| | - Elisabeth Sanchez-Mejias
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Victoria Navarro
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, C/Prof. Garcia Gonzalez 2, 41012, Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Sebastian Jimenez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, C/Prof. Garcia Gonzalez 2, 41012, Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Cristina Nuñez-Diaz
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Angela Gomez-Arboledas
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ines Moreno-Gonzalez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Marisa Vizuete
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, C/Prof. Garcia Gonzalez 2, 41012, Sevilla, Spain.,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC, Universidad de Sevilla, Sevilla, Spain
| | - Jose Carlos Davila
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Vitorica
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain. .,Dpto. Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, C/Prof. Garcia Gonzalez 2, 41012, Sevilla, Spain. .,Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocio/CSIC, Universidad de Sevilla, Sevilla, Spain.
| | - Antonia Gutierrez
- Dpto. Biología Celular, Genética y Fisiología, Instituto de Investigación Biomédica de Málaga-IBIMA, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, 29071, Málaga, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
38
|
Lopes AT, Hausrat TJ, Heisler FF, Gromova KV, Lombino FL, Fischer T, Ruschkies L, Breiden P, Thies E, Hermans-Borgmeyer I, Schweizer M, Schwarz JR, Lohr C, Kneussel M. Spastin depletion increases tubulin polyglutamylation and impairs kinesin-mediated neuronal transport, leading to working and associative memory deficits. PLoS Biol 2020; 18:e3000820. [PMID: 32866173 PMCID: PMC7485986 DOI: 10.1371/journal.pbio.3000820] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/11/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Mutations in the gene encoding the microtubule-severing protein spastin (spastic paraplegia 4 [SPG4]) cause hereditary spastic paraplegia (HSP), associated with neurodegeneration, spasticity, and motor impairment. Complicated forms (complicated HSP [cHSP]) further include cognitive deficits and dementia; however, the etiology and dysfunctional mechanisms of cHSP have remained unknown. Here, we report specific working and associative memory deficits upon spastin depletion in mice. Loss of spastin-mediated severing leads to reduced synapse numbers, accompanied by lower miniature excitatory postsynaptic current (mEPSC) frequencies. At the subcellular level, mutant neurons are characterized by longer microtubules with increased tubulin polyglutamylation levels. Notably, these conditions reduce kinesin-microtubule binding, impair the processivity of kinesin family protein (KIF) 5, and reduce the delivery of presynaptic vesicles and postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Rescue experiments confirm the specificity of these results by showing that wild-type spastin, but not the severing-deficient and disease-associated K388R mutant, normalizes the effects at the synaptic, microtubule, and transport levels. In addition, short hairpin RNA (shRNA)-mediated reduction of tubulin polyglutamylation on spastin knockout background normalizes KIF5 transport deficits and attenuates the loss of excitatory synapses. Our data provide a mechanism that connects spastin dysfunction with the regulation of kinesin-mediated cargo transport, synapse integrity, and cognition. This study identifies deficits in working and associative memory in spastin knockout mice, resembling the cognitive deficits described in humans with severe forms of SPG4-type hereditary spastic paraplegia. Mechanistically, the findings suggest that impaired microtubule growth, kinesin motility and cargo delivery of synaptic AMPA receptors may contribute to the etiology of the disease.
Collapse
Affiliation(s)
- André T. Lopes
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Torben J. Hausrat
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank F. Heisler
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kira V. Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franco L. Lombino
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Fischer
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Laura Ruschkies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Breiden
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Animal Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Morphology Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jürgen R. Schwarz
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
39
|
Rolls MM, Thyagarajan P, Feng C. Microtubule dynamics in healthy and injured neurons. Dev Neurobiol 2020; 81:321-332. [PMID: 32291942 DOI: 10.1002/dneu.22746] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/22/2020] [Accepted: 04/08/2020] [Indexed: 12/28/2022]
Abstract
Most neurons must last a lifetime and their microtubule cytoskeleton is an important contributor to their longevity. Neurons have some of the most stable microtubules of all cells, but the tip of every microtubule remains dynamic and, although requiring constant GTP consumption, microtubules are always being rebuilt. While some ongoing level of rebuilding always occurs, overall microtubule stability can be modulated in response to injury and stress as well as the normal developmental process of pruning. Specific microtubule severing proteins act in different contexts to increase microtubule dynamicity and promote degeneration and pruning. After axon injury, complex changes in dynamics occur and these are important for both neuroprotection induced by injury and subsequent outgrowth of a new axon. Understanding how microtubule dynamics is modulated in different scenarios, as well as the impact of the changes in stability, is an important avenue to explore for development of strategies to promote neuroprotection and regeneration.
Collapse
Affiliation(s)
- Melissa M Rolls
- Biochemistry and Molecular Biology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Pankajam Thyagarajan
- Biochemistry and Molecular Biology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Chengye Feng
- Biochemistry and Molecular Biology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
40
|
Tadepalle N, Robers L, Veronese M, Zentis P, Babatz F, Brodesser S, Gruszczyk AV, Schauss A, Höning S, Rugarli EI. Microtubule-dependent and independent roles of spastin in lipid droplet dispersion and biogenesis. Life Sci Alliance 2020; 3:3/6/e202000715. [PMID: 32321733 PMCID: PMC7184029 DOI: 10.26508/lsa.202000715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 12/21/2022] Open
Abstract
Lipid droplets (LDs) are metabolic organelles that store neutral lipids and dynamically respond to changes in energy availability by accumulating or mobilizing triacylglycerols (TAGs). How the plastic behavior of LDs is regulated is poorly understood. Hereditary spastic paraplegia is a central motor axonopathy predominantly caused by mutations in SPAST, encoding the microtubule-severing protein spastin. The spastin-M1 isoform localizes to nascent LDs in mammalian cells; however, the mechanistic significance of this targeting is not fully explained. Here, we show that tightly controlled levels of spastin-M1 are required to inhibit LD biogenesis and TAG accumulation. Spastin-M1 maintains the morphogenesis of the ER when TAG synthesis is prevented, independent from microtubule binding. Moreover, spastin plays a microtubule-dependent role in mediating the dispersion of LDs from the ER upon glucose starvation. Our results reveal a dual role of spastin to shape ER tubules and to regulate LD movement along microtubules, opening new perspectives for the pathogenesis of hereditary spastic paraplegia.
Collapse
Affiliation(s)
- Nimesha Tadepalle
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Lennart Robers
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Matteo Veronese
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Peter Zentis
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Felix Babatz
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Susanne Brodesser
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Anja V Gruszczyk
- Institute for Genetics, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Astrid Schauss
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Stefan Höning
- Institute for Biochemistry I, University of Cologne, Cologne, Germany
| | - Elena I Rugarli
- Institute for Genetics, University of Cologne, Cologne, Germany .,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| |
Collapse
|
41
|
Abstract
The intracellular transport system in neurons is specialized to an extraordinary degree, enabling the delivery of critical cargo to sites in axons or dendrites that are far removed from the cell center. Vesicles formed in the cell body are actively transported by kinesin motors along axonal microtubules to presynaptic sites that can be located more than a meter away. Both growth factors and degradative vesicles carrying aged organelles or aggregated proteins take the opposite route, driven by dynein motors. Distance is not the only challenge; precise delivery of cargos to sites of need must also be accomplished. For example, localized delivery of presynaptic components to hundreds of thousands of "en passant" synapses distributed along the length of a single axon in some neuronal subtypes provides a layer of complexity that must be successfully navigated to maintain synaptic transmission. We review recent advances in the field of axonal transport, with a focus on conceptual developments, and highlight our growing quantitative understanding of neuronal trafficking and its role in maintaining the synaptic function that underlies higher cognitive processes such as learning and memory.
Collapse
Affiliation(s)
- Pedro Guedes-Dias
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Institute of Neuronal Cell Biology, Technische Universität München, 80802 Munich, Germany
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
42
|
Fu XQ, Peng J, Wang AH, Luo ZG. Tumor necrosis factor alpha mediates neuromuscular synapse elimination. Cell Discov 2020; 6:9. [PMID: 32140252 PMCID: PMC7051980 DOI: 10.1038/s41421-020-0143-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/03/2020] [Indexed: 12/17/2022] Open
Abstract
During the development of mammalian neuromuscular junction (NMJ), the original supernumerary axon inputs are gradually eliminated, finally leaving each muscle fiber innervated by a single axon terminal. However, the molecular cues that mediate the elimination of redundant axon inputs remain unclear. Here we show that tumor necrosis factor-α (TNFα) expressed in postsynaptic muscle cells plays an important role in presynaptic axonal elimination at the NMJ. We found that intramuscular injection of TNFα into the levator auris longus (LAL) muscles caused disassociation of presynaptic nerve terminals from the postsynaptic acetylcholine receptor (AChR) clusters. By contrast, genetic ablation of TNFα globally or specifically in skeletal muscle cells, but not in motoneurons or Schwann cells, delayed the synaptic elimination. Moreover, ablation of TNFα in muscle cells attenuated the tendency of activity-dependent competition in a motoneuron-muscle coculture system. These results suggest a role of postsynaptic TNFα in the elimination of redundant synaptic inputs.
Collapse
Affiliation(s)
- Xiu-Qing Fu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210 China
| | - Jian Peng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210 China
- State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ai-Hua Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210 China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210 China
| |
Collapse
|
43
|
MMP-mediated modulation of ECM environment during axonal growth and NMJ development. Neurosci Lett 2020; 724:134822. [PMID: 32061716 DOI: 10.1016/j.neulet.2020.134822] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 12/19/2022]
Abstract
Motor neurons, skeletal muscles, and perisynaptic Schwann cells interact with extracellular matrix (ECM) to form the tetrapartite synapse in the peripheral nervous system. Dynamic remodeling of ECM composition is essential to diversify its functions for distinct cellular processes during neuromuscular junction (NMJ) development. In this review, we give an overview of the proteolytic regulation of ECM proteins, particularly by secreted and membrane-type matrix metalloproteinases (MMPs), in axonal growth and NMJ development. It is suggested that MMP-2, MMP-9, and membrane type 1-MMP (MT1-MMP) promote axonal outgrowth and regeneration upon injury by clearing the glial scars at the lesion site. In addition, these MMPs also play roles in neuromuscular synaptogenesis, ranging from spontaneous formation of synaptic specializations to activity-dependent synaptic elimination, via proteolytic cleavage or degradation of growth factors, neurotrophic factors, and ECM molecules. For instance, secreted MMP-3 has been known to cleave agrin, the main postsynaptic differentiation inducer, further highlighting the importance of MMPs in NMJ formation and maintenance. Furthermore, the increased level of several MMPs in myasthenia gravis (MG) patient suggest the pathophysiological mechanisms of MMP-mediated proteolytic degradation in MG pathogenesis. Finally, we speculate on potential major future directions for studying the regulatory functions of MMP-mediated ECM remodeling in axonal growth and NMJ development.
Collapse
|
44
|
Clark JA, Chuckowree JA, Dyer MS, Dickson TC, Blizzard CA. Epothilone D alters normal growth, viability and microtubule dependent intracellular functions of cortical neurons in vitro. Sci Rep 2020; 10:918. [PMID: 31969604 PMCID: PMC6976590 DOI: 10.1038/s41598-020-57718-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/18/2019] [Indexed: 01/09/2023] Open
Abstract
Brain penetrant microtubule stabilising agents (MSAs) are being increasingly validated as potential therapeutic strategies for neurodegenerative diseases and traumatic injuries of the nervous system. MSAs are historically used to treat malignancies to great effect. However, this treatment strategy can also cause adverse off-target impacts, such as the generation of debilitating neuropathy and axonal loss. Understanding of the effects that individual MSAs have on neurons of the central nervous system is still incomplete. Previous research has revealed that aberrant microtubule stabilisation can perturb many neuronal functions, such as neuronal polarity, neurite outgrowth, microtubule dependant transport and overall neuronal viability. In the current study, we evaluate the dose dependant impact of epothilone D, a brain penetrant MSA, on both immature and relatively mature mouse cortical neurons in vitro. We show that epothilone D reduces the viability, growth and complexity of immature cortical neurons in a dose dependant manner. Furthermore, in relatively mature cortical neurons, we demonstrate that while cellularly lethal doses of epothilone D cause cellular demise, low sub lethal doses can also affect mitochondrial transport over time. Our results reveal an underappreciated mitochondrial disruption over a wide range of epothilone D doses and reiterate the importance of understanding the dosage, timing and intended outcome of MSAs, with particular emphasis on brain penetrant MSAs being considered to target neurons in disease and trauma.
Collapse
Affiliation(s)
- J A Clark
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - J A Chuckowree
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - M S Dyer
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - T C Dickson
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia
| | - C A Blizzard
- Menzies Institute for Medical Research, University of Tasmania 17 Liverpool Street Hobart, Tasmania, 7000, Australia.
| |
Collapse
|
45
|
Ziak J, Weissova R, Jeřábková K, Janikova M, Maimon R, Petrasek T, Pukajova B, Kleisnerova M, Wang M, Brill MS, Kasparek P, Zhou X, Alvarez-Bolado G, Sedlacek R, Misgeld T, Stuchlik A, Perlson E, Balastik M. CRMP2 mediates Sema3F-dependent axon pruning and dendritic spine remodeling. EMBO Rep 2020; 21:e48512. [PMID: 31919978 DOI: 10.15252/embr.201948512] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 11/09/2022] Open
Abstract
Regulation of axon guidance and pruning of inappropriate synapses by class 3 semaphorins are key to the development of neural circuits. Collapsin response mediator protein 2 (CRMP2) has been shown to regulate axon guidance by mediating semaphorin 3A (Sema3A) signaling; however, nothing is known about its role in synapse pruning. Here, using newly generated crmp2-/- mice we demonstrate that CRMP2 has a moderate effect on Sema3A-dependent axon guidance in vivo, and its deficiency leads to a mild defect in axon guidance in peripheral nerves and the corpus callosum. Surprisingly, crmp2-/- mice display prominent defects in stereotyped axon pruning in hippocampus and visual cortex and altered dendritic spine remodeling, which is consistent with impaired Sema3F signaling and with models of autism spectrum disorder (ASD). We demonstrate that CRMP2 mediates Sema3F signaling in primary neurons and that crmp2-/- mice display ASD-related social behavior changes in the early postnatal period as well as in adults. Together, we demonstrate that CRMP2 mediates Sema3F-dependent synapse pruning and its dysfunction shares histological and behavioral features of ASD.
Collapse
Affiliation(s)
- Jakub Ziak
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Romana Weissova
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Kateřina Jeřábková
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Janikova
- Department of Neurophysiology of the Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Roy Maimon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Tomas Petrasek
- Department of Neurophysiology of the Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Barbora Pukajova
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Marie Kleisnerova
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Petr Kasparek
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Xunlei Zhou
- Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | | | - Radislav Sedlacek
- Department of Transgenic Models of Diseases and Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.,German Center for Neurodegenerative Diseases and Munich Cluster for Systems Neurology, Munich, Germany
| | - Ales Stuchlik
- Department of Neurophysiology of the Memory, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Martin Balastik
- Department of Molecular Neurobiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
46
|
Congenic expression of poly-GA but not poly-PR in mice triggers selective neuron loss and interferon responses found in C9orf72 ALS. Acta Neuropathol 2020; 140:121-142. [PMID: 32562018 PMCID: PMC7360660 DOI: 10.1007/s00401-020-02176-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
Expansion of a (G4C2)n repeat in C9orf72 causes amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), but the link of the five repeat-encoded dipeptide repeat (DPR) proteins to neuroinflammation, TDP-43 pathology, and neurodegeneration is unclear. Poly-PR is most toxic in vitro, but poly-GA is far more abundant in patients. To directly compare these in vivo, we created congenic poly-GA and poly-PR mice. 40% of poly-PR mice were affected with ataxia and seizures, requiring euthanasia by 6 weeks of age. The remaining poly-PR mice were asymptomatic at 14 months of age, likely due to an 80% reduction of the transgene mRNA in this subgroup. In contrast, all poly-GA mice showed selective neuron loss, inflammation, as well as muscle denervation and wasting requiring euthanasia before 7 weeks of age. In-depth analysis of peripheral organs and blood samples suggests that peripheral organ failure does not drive these phenotypes. Although transgene mRNA levels were similar between poly-GA and affected poly-PR mice, poly-GA aggregated far more abundantly than poly-PR in the CNS and was also found in skeletal muscle. In addition, TDP-43 and other disease-linked RNA-binding proteins co-aggregated in rare nuclear inclusions in the hippocampus and frontal cortex only in poly-GA mice. Transcriptome analysis revealed activation of an interferon-responsive pro-inflammatory microglial signature in end-stage poly-GA but not poly-PR mice. This signature was also found in all ALS patients and enriched in C9orf72 cases. In summary, our rigorous comparison of poly-GA and poly-PR toxicity in vivo indicates that poly-GA, but not poly-PR at the same mRNA expression level, promotes interferon responses in C9orf72 disease and contributes to TDP-43 abnormalities and neuron loss selectively in disease-relevant regions.
Collapse
|
47
|
Lee YI. Developmental neuromuscular synapse elimination: Activity-dependence and potential downstream effector mechanisms. Neurosci Lett 2019; 718:134724. [PMID: 31877335 DOI: 10.1016/j.neulet.2019.134724] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
Synaptic connections initially formed during nervous system development undergo a significant transformation during nervous system maturation. Such maturation is essential for the proper architecture and function of the nervous system. Developmental synaptic transformation includes "synapse elimination," a process in which multiple immature presynaptic inputs converge at and compete for control of a common postsynaptic target. This developmental synaptic remodeling is best understood at mammalian neuromuscular junctions. It is well established that neuromuscular activity provides the impetus for the pruning of redundant motor axon inputs. Despite the dominant influence neuromuscular activity exerts on developmental synapse elimination, however, the downstream mechanisms of neuromuscular activity that affect synapse elimination remain poorly understood. Conversely, although several cellular and molecular effector mechanisms are known to impact synapse elimination, it is unclear whether they are modulated by neuromuscular activity. This review discusses how the motor neurons, synaptic glia and muscle fibers each contributes to the developmental phenomenon, and speculates how neuromuscular activity may modulate these contributions.
Collapse
Affiliation(s)
- Young Il Lee
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
48
|
Darabid H, St-Pierre-See A, Robitaille R. Purinergic-Dependent Glial Regulation of Synaptic Plasticity of Competing Terminals and Synapse Elimination at the Neuromuscular Junction. Cell Rep 2019; 25:2070-2082.e6. [PMID: 30463006 DOI: 10.1016/j.celrep.2018.10.075] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 07/23/2018] [Accepted: 10/19/2018] [Indexed: 01/20/2023] Open
Abstract
The precise wiring of synaptic connections requires the elimination of supernumerary inputs competing for innervation of the same target cell. This competition is activity-dependent, strengthening some inputs whereas others are eliminated. Although glial cells are required for the elimination and clearance of terminals, their involvement in activity-dependent synaptic competition remains ill-defined. Here, we used the developing neuromuscular junctions of mice to show that perisynaptic glial cells, through 2Y1 purinergic receptors (P2Y1Rs), decode synaptic efficacy of competing terminals in a Ca2+-dependent manner. This glial activity induces long-lasting synaptic potentiation of strong but not weak terminals via presynaptic adenosine 2A receptors. Blockade of glial activity by intracellular Ca2+ chelation or blockade of P2Y1Rs prevents this plasticity. In addition, blockade of P2Y1Rs delays synapse elimination in vivo. Hence, P2Y1Rs drive glial cell regulation of strong synaptic inputs and influence synapse competition and elimination.
Collapse
Affiliation(s)
- Houssam Darabid
- Département de Neurosciences, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada; Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Alexandre St-Pierre-See
- Département de Neurosciences, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada; Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Richard Robitaille
- Département de Neurosciences, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada; Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, PO Box 6128, Station Centre-ville, Montréal, QC H3C 3J7, Canada.
| |
Collapse
|
49
|
The proteasome regulator PI31 is required for protein homeostasis, synapse maintenance, and neuronal survival in mice. Proc Natl Acad Sci U S A 2019; 116:24639-24650. [PMID: 31754024 PMCID: PMC6900516 DOI: 10.1073/pnas.1911921116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The conserved proteasome-binding protein PI31 serves as an adapter to couple proteasomes with cellular motors to mediate their transport to distal tips of neurons where protein breakdown occurs. We generated global and conditional PI31 knockout mouse strains and show that this protein is required for protein homeostasis, and that its conditional inactivation in neurons disrupts synaptic structures and long-term survival. This work establishes a critical role for PI31 and local protein degradation in the maintenance of neuronal architecture, circuitry, and function. Because mutations in the PI31 pathway cause neurodegenerative diseases in humans, reduced PI31 activity may contribute to the etiology of these diseases. Proteasome-mediated degradation of intracellular proteins is essential for cell function and survival. The proteasome-binding protein PI31 (Proteasomal Inhibitor of 31kD) promotes 26S assembly and functions as an adapter for proteasome transport in axons. As localized protein synthesis and degradation is especially critical in neurons, we generated a conditional loss of PI31 in spinal motor neurons (MNs) and cerebellar Purkinje cells (PCs). A cKO of PI31 in these neurons caused axon degeneration, neuronal loss, and progressive spinal and cerebellar neurological dysfunction. For both MNs and PCs, markers of proteotoxic stress preceded axonal degeneration and motor dysfunction, indicating a critical role for PI31 in neuronal homeostasis. The time course of the loss of MN and PC function in developing mouse central nervous system suggests a key role for PI31 in human neurodegenerative diseases.
Collapse
|
50
|
Fecher C, Trovò L, Müller SA, Snaidero N, Wettmarshausen J, Heink S, Ortiz O, Wagner I, Kühn R, Hartmann J, Karl RM, Konnerth A, Korn T, Wurst W, Merkler D, Lichtenthaler SF, Perocchi F, Misgeld T. Cell-type-specific profiling of brain mitochondria reveals functional and molecular diversity. Nat Neurosci 2019; 22:1731-1742. [DOI: 10.1038/s41593-019-0479-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 07/25/2019] [Indexed: 12/21/2022]
|