1
|
Leterrier C. Stress balls for the brain: How beading protects axons from mechanical damage. J Cell Biol 2024; 223:e202407024. [PMID: 38985206 PMCID: PMC11233989 DOI: 10.1083/jcb.202407024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024] Open
Abstract
The slender shape of axons makes them uniquely susceptible to mechanical stress. In this issue, Pan, Hu et al. (https://doi.org/10.1083/jcb.202206046) use a microfluidic axon-on-chip device to reveal how actomyosin protects axons from mild mechanical stress, by transiently adopting a beaded shape that helps limit the spread of damaging calcium waves.
Collapse
|
2
|
Pan X, Hu Y, Lei G, Wei Y, Li J, Luan T, Zhang Y, Chu Y, Feng Y, Zhan W, Zhao C, Meunier FA, Liu Y, Li Y, Wang T. Actomyosin-II protects axons from degeneration induced by mild mechanical stress. J Cell Biol 2024; 223:e202206046. [PMID: 38713825 PMCID: PMC11076810 DOI: 10.1083/jcb.202206046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 06/16/2023] [Accepted: 04/09/2024] [Indexed: 05/09/2024] Open
Abstract
Whether, to what extent, and how the axons in the central nervous system (CNS) can withstand sudden mechanical impacts remain unclear. By using a microfluidic device to apply controlled transverse mechanical stress to axons, we determined the stress levels that most axons can withstand and explored their instant responses at nanoscale resolution. We found mild stress triggers a highly reversible, rapid axon beading response, driven by actomyosin-II-dependent dynamic diameter modulations. This mechanism contributes to hindering the long-range spread of stress-induced Ca2+ elevations into non-stressed neuronal regions. Through pharmacological and molecular manipulations in vitro, we found that actomyosin-II inactivation diminishes the reversible beading process, fostering progressive Ca2+ spreading and thereby increasing acute axonal degeneration in stressed axons. Conversely, upregulating actomyosin-II activity prevents the progression of initial injury, protecting stressed axons from acute degeneration both in vitro and in vivo. Our study unveils the periodic actomyosin-II in axon shafts cortex as a novel protective mechanism, shielding neurons from detrimental effects caused by mechanical stress.
Collapse
Affiliation(s)
- Xiaorong Pan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yiqing Hu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Gaowei Lei
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences China, Shanghai, China
| | - Yaxuan Wei
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences China, Shanghai, China
| | - Jie Li
- Division of Chemistry and Physical Biology, School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| | - Tongshu Luan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yunfan Zhang
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences China, Shanghai, China
| | - Yuanyuan Chu
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yu Feng
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Wenrong Zhan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chunxia Zhao
- School of Chemical Engineering, The University of Adelaide, Adelaide, Australia
| | - Frédéric A. Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Yifan Liu
- Division of Chemistry and Physical Biology, School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, China
| | - Yi Li
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences China, Shanghai, China
| | - Tong Wang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
3
|
Kim JY, Yang JE, Mitchell JW, English LA, Yang SZ, Tenpas T, Dent EW, Wildonger J, Wright ER. Handling Difficult Cryo-ET Samples: A Study with Primary Neurons from Drosophila melanogaster. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:2127-2148. [PMID: 37966978 PMCID: PMC11168236 DOI: 10.1093/micmic/ozad125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/01/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023]
Abstract
Cellular neurobiology has benefited from recent advances in the field of cryo-electron tomography (cryo-ET). Numerous structural and ultrastructural insights have been obtained from plunge-frozen primary neurons cultured on electron microscopy grids. With most primary neurons having been derived from rodent sources, we sought to expand the breadth of sample availability by using primary neurons derived from 3rd instar Drosophila melanogaster larval brains. Ultrastructural abnormalities were encountered while establishing this model system for cryo-ET, which were exemplified by excessive membrane blebbing and cellular fragmentation. To optimize neuronal samples, we integrated substrate selection, micropatterning, montage data collection, and chemical fixation. Efforts to address difficulties in establishing Drosophila neurons for future cryo-ET studies in cellular neurobiology also provided insights that future practitioners can use when attempting to establish other cell-based model systems.
Collapse
Affiliation(s)
- Joseph Y. Kim
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jie E. Yang
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Cryo-Electron Microscopy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Josephine W. Mitchell
- Department of Chemistry and Biochemistry, Kalamazoo College, Kalamazoo, MI 49006, USA
| | - Lauren A. English
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sihui Z. Yang
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Tanner Tenpas
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Erik W. Dent
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jill Wildonger
- Departments of Pediatrics and Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Elizabeth R. Wright
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Cryo-Electron Microscopy Research Center, University of Wisconsin-Madison, Madison, WI 53706, USA
- Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53715, USA
| |
Collapse
|
4
|
Ma D, Sun C, Manne R, Guo T, Bosc C, Barry J, Magliery T, Andrieux A, Li H, Gu C. A cytoskeleton-membrane interaction conserved in fast-spiking neurons controls movement, emotion, and memory. Mol Psychiatry 2023; 28:3994-4010. [PMID: 37833406 PMCID: PMC10905646 DOI: 10.1038/s41380-023-02286-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/20/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023]
Abstract
The pathogenesis of schizophrenia is believed to involve combined dysfunctions of many proteins including microtubule-associated protein 6 (MAP6) and Kv3.1 voltage-gated K+ (Kv) channel, but their relationship and functions in behavioral regulation are often not known. Here we report that MAP6 stabilizes Kv3.1 channels in parvalbumin-positive (PV+ ) fast-spiking GABAergic interneurons, regulating behavior. MAP6-/- and Kv3.1-/- mice display similar hyperactivity and avoidance reduction. Their proteins colocalize in PV+ interneurons and MAP6 deletion markedly reduces Kv3.1 protein level. We further show that two microtubule-binding modules of MAP6 bind the Kv3.1 tetramerization domain with high affinity, maintaining the channel level in both neuronal soma and axons. MAP6 knockdown by AAV-shRNA in the amygdala or the hippocampus reduces avoidance or causes hyperactivity and recognition memory deficit, respectively, through elevating projection neuron activity. Finally, knocking down Kv3.1 or disrupting the MAP6-Kv3.1 binding in these brain regions causes avoidance reduction and hyperactivity, consistent with the effects of MAP6 knockdown. Thus, disrupting this conserved cytoskeleton-membrane interaction in fast-spiking neurons causes different degrees of functional vulnerability in various neural circuits.
Collapse
Affiliation(s)
- Di Ma
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Chao Sun
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
- MCDB graduate program, The Ohio State University, Columbus, OH, USA
| | - Rahul Manne
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Tianqi Guo
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Christophe Bosc
- Univ. Grenoble Alpes, Inserm, U1216, CEA, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Joshua Barry
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
- IDDRC, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas Magliery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
| | - Annie Andrieux
- Univ. Grenoble Alpes, Inserm, U1216, CEA, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Houzhi Li
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA
| | - Chen Gu
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH, USA.
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, 43210, USA.
- MCDB graduate program, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
5
|
Kim JY, Yang JE, Mitchell JW, English LA, Yang SZ, Tenpas T, Dent EW, Wildonger J, Wright ER. Handling difficult cryo-ET samples: A study with primary neurons from Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.10.548468. [PMID: 37502991 PMCID: PMC10369871 DOI: 10.1101/2023.07.10.548468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Cellular neurobiology has benefited from recent advances in the field of cryo-electron tomography (cryo-ET). Numerous structural and ultrastructural insights have been obtained from plunge-frozen primary neurons cultured on electron microscopy grids. With most primary neurons been derived from rodent sources, we sought to expand the breadth of sample availability by using primary neurons derived from 3rd instar Drosophila melanogaster larval brains. Ultrastructural abnormalities were encountered while establishing this model system for cryo-ET, which were exemplified by excessive membrane blebbing and cellular fragmentation. To optimize neuronal samples, we integrated substrate selection, micropatterning, montage data collection, and chemical fixation. Efforts to address difficulties in establishing Drosophila neurons for future cryo-ET studies in cellular neurobiology also provided insights that future practitioners can use when attempting to establish other cell-based model systems.
Collapse
Affiliation(s)
- Joseph Y. Kim
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jie E. Yang
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Cryo-Electron Microscopy Research Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Josephine W. Mitchell
- Department of Chemistry and Biochemistry, Kalamazoo College, Kalamazoo, MI, 49006, USA
| | - Lauren A. English
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Sihui Z. Yang
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
| | - Tanner Tenpas
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Erik W. Dent
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Jill Wildonger
- Departments of Pediatrics and Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Elizabeth R. Wright
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Cryo-Electron Microscopy Research Center, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI, 53715, USA
| |
Collapse
|
6
|
Ghose A, Pullarkat P. The role of mechanics in axonal stability and development. Semin Cell Dev Biol 2023; 140:22-34. [PMID: 35786351 PMCID: PMC7615100 DOI: 10.1016/j.semcdb.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/05/2022] [Accepted: 06/13/2022] [Indexed: 01/28/2023]
Abstract
Much of the focus of neuronal cell biology has been devoted to growth cone guidance, synaptogenesis, synaptic activity, plasticity, etc. The axonal shaft too has received much attention, mainly for its astounding ability to transmit action potentials and the transport of material over long distances. For these functions, the axonal cytoskeleton and membrane have been often assumed to play static structural roles. Recent experiments have changed this view by revealing an ultrastructure much richer in features than previously perceived and one that seems to be maintained at a dynamic steady state. The role of mechanics in this is only beginning to be broadly appreciated and appears to involve passive and active modes of coupling different biopolymer filaments, filament turnover dynamics and membrane biophysics. Axons, being unique cellular processes in terms of high aspect ratios and often extreme lengths, also exhibit unique passive mechanical properties that might have evolved to stabilize them under mechanical stress. In this review, we summarize the experiments that have exposed some of these features. It is our view that axonal mechanics deserves much more attention not only due to its significance in the development and maintenance of the nervous system but also due to the susceptibility of axons to injury and neurodegeneration.
Collapse
Affiliation(s)
- Aurnab Ghose
- Indian Institute of Science Education and Research, Pune 411 008, India.
| | - Pramod Pullarkat
- Raman Research Institute, C. V. Raman Avenue, Bengaluru 560 080, India.
| |
Collapse
|
7
|
Boal AM, McGrady NR, Chamling X, Kagitapalli BS, Zack DJ, Calkins DJ, Risner ML. Microfluidic Platforms Promote Polarization of Human-Derived Retinal Ganglion Cells That Model Axonopathy. Transl Vis Sci Technol 2023; 12:1. [PMID: 37010860 PMCID: PMC10080917 DOI: 10.1167/tvst.12.4.1] [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: 12/06/2022] [Accepted: 03/09/2023] [Indexed: 04/04/2023] Open
Abstract
Purpose Axons depend on long-range transport of proteins and organelles which increases susceptibility to metabolic stress in disease. The axon initial segment (AIS) is particularly vulnerable due to the high bioenergetic demand of action potential generation. Here, we prepared retinal ganglion cells derived from human embryonic stem cells (hRGCs) to probe how axonal stress alters AIS morphology. Methods hRGCs were cultured on coverslips or microfluidic platforms. We assayed AIS specification and morphology by immunolabeling against ankyrin G (ankG), an axon-specific protein, and postsynaptic density 95 (PSD-95), a dendrite-specific protein. Using microfluidic platforms that enable fluidic isolation, we added colchicine to the axon compartment to lesion axons. We verified axonopathy by measuring the anterograde axon transport of cholera toxin subunit B and immunolabeling against cleaved caspase 3 (CC3) and phosphorylated neurofilament H (SMI-34). We determined the influence of axon injury on AIS morphology by immunolabeling samples against ankG and measuring AIS distance from soma and length. Results Based on measurements of ankG and PSD-95 immunolabeling, microfluidic platforms promote the formation and separation of distinct somatic-dendritic versus axonal compartments in hRGCs compared to coverslip cultures. Chemical lesioning of axons by colchicine reduced hRGC anterograde axon transport, increased varicosity density, and enhanced expression of CC3 and SMI-34. Interestingly, we found that colchicine selectively affected hRGCs with axon-carrying dendrites by reducing AIS distance from somas and increasing length, thus suggesting reduced capacity to maintain excitability. Conclusions Thus, microfluidic platforms promote polarized hRGCs that enable modeling of axonopathy. Translational Relevance Microfluidic platforms may be used to assay compartmentalized degeneration that occurs during glaucoma.
Collapse
Affiliation(s)
- Andrew M. Boal
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nolan R. McGrady
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xitiz Chamling
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bhanu S. Kagitapalli
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Donald J. Zack
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J. Calkins
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael L. Risner
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
8
|
Zeng ML, Kong S, Chen TX, Peng BW. Transient Receptor Potential Vanilloid 4: a Double-Edged Sword in the Central Nervous System. Mol Neurobiol 2023; 60:1232-1249. [PMID: 36434370 DOI: 10.1007/s12035-022-03141-6] [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: 08/09/2022] [Accepted: 11/17/2022] [Indexed: 11/26/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel that can be activated by diverse stimuli, such as heat, mechanical force, hypo-osmolarity, and arachidonic acid metabolites. TRPV4 is widely expressed in the central nervous system (CNS) and participates in many significant physiological processes. However, accumulative evidence has suggested that deficiency, abnormal expression or distribution, and overactivation of TRPV4 are involved in pathological processes of multiple neurological diseases. Here, we review the latest studies concerning the known features of this channel, including its expression, structure, and its physiological and pathological roles in the CNS, proposing an emerging therapeutic strategy for CNS diseases.
Collapse
Affiliation(s)
- Meng-Liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Tao-Xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.
| |
Collapse
|
9
|
Karimy JK, Newville JC, Sadegh C, Morris JA, Monuki ES, Limbrick DD, McAllister Ii JP, Koschnitzky JE, Lehtinen MK, Jantzie LL. Outcomes of the 2019 hydrocephalus association workshop, "Driving common pathways: extending insights from posthemorrhagic hydrocephalus". Fluids Barriers CNS 2023; 20:4. [PMID: 36639792 PMCID: PMC9838022 DOI: 10.1186/s12987-023-00406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The Hydrocephalus Association (HA) workshop, Driving Common Pathways: Extending Insights from Posthemorrhagic Hydrocephalus, was held on November 4 and 5, 2019 at Washington University in St. Louis. The workshop brought together a diverse group of basic, translational, and clinical scientists conducting research on multiple hydrocephalus etiologies with select outside researchers. The main goals of the workshop were to explore areas of potential overlap between hydrocephalus etiologies and identify drug targets that could positively impact various forms of hydrocephalus. This report details the major themes of the workshop and the research presented on three cell types that are targets for new hydrocephalus interventions: choroid plexus epithelial cells, ventricular ependymal cells, and immune cells (macrophages and microglia).
Collapse
Affiliation(s)
- Jason K Karimy
- Department of Family Medicine, Mountain Area Health Education Center - Boone, North Carolina, 28607, USA
| | - Jessie C Newville
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Cameron Sadegh
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, MA, Boston, 02114, USA
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Jill A Morris
- National Institute of Neurological Disorders and Stroke, Neuroscience Center, National Institutes of Health, 6001 Executive Blvd, NSC Rm 2112, Bethesda, MD, 20892, USA
| | - Edwin S Monuki
- Departments of Pathology & Laboratory Medicine and Developmental & Cell Biology, University of California Irvine, Irvine, CA, 92697, USA
| | - David D Limbrick
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - James P McAllister Ii
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | | | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA.
| | - Lauren L Jantzie
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
- Kennedy Krieger Institute, Baltimore, MD, 21287, USA.
| |
Collapse
|
10
|
Pan X, Li J, Li W, Wang H, Durisic N, Li Z, Feng Y, Liu Y, Zhao CX, Wang T. Axons-on-a-chip for mimicking non-disruptive diffuse axonal injury underlying traumatic brain injury. LAB ON A CHIP 2022; 22:4541-4555. [PMID: 36318066 DOI: 10.1039/d2lc00730d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Diffuse axonal injury (DAI) is the most severe pathological feature of traumatic brain injury (TBI). However, how primary axonal injury is induced by transient mechanical impacts remains unknown, mainly due to the low temporal and spatial resolution of medical imaging approaches. Here we established an axon-on-a-chip (AoC) model for mimicking DAI and monitoring instant cellular responses. Integrating computational fluid dynamics and microfluidic techniques, DAI was induced by injecting a precisely controlled micro-flux in the transverse direction. The clear correlation between the flow speed of injecting flux and the severity of DAI was elucidated. We next used the AoC to investigate the instant intracellular responses underlying DAI and found that the dynamic formation of focal axonal swellings (FAS) accompanied by Ca2+ surge occurs during the flux. Surprisingly, periodic axonal cytoskeleton disruption also occurs rapidly after the flux. These instant injury responses are spatially restricted to the fluxed axon, not affecting the overall viability of the neuron in the acute stage. Compatible with high-resolution live microscopy, the AoC provides a versatile system to identify early mechanisms underlying DAI, offering a platform for screening effective treatments to alleviate TBI.
Collapse
Affiliation(s)
- Xiaorong Pan
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Jie Li
- Division of Chemistry and Physical Biology, School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Haofei Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nela Durisic
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhenyu Li
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Yu Feng
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Yifan Liu
- Division of Chemistry and Physical Biology, School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, SA 5005, Australia.
| | - Tong Wang
- The Brain Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
11
|
Song H, Chen C, Kelley B, Tomasevich A, Lee H, Dolle JP, Cheng J, Garcia B, Meaney DF, Smith DH. Traumatic brain injury recapitulates developmental changes of axons. Prog Neurobiol 2022; 217:102332. [PMID: 35870679 PMCID: PMC9454890 DOI: 10.1016/j.pneurobio.2022.102332] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022]
Abstract
During development, half of brain white matter axons are maintained for growth, while the remainder undergo developmental axon degeneration. After traumatic brain injury (TBI), injured axons also appear to follow pathways leading to either degeneration or repair. These observations raise the intriguing, but unexamined possibility that TBI recapitulates developmental axonal programs. Here, we examined axonal changes in the developing brain in young rats and after TBI in adult rat. Multiple shared changes in axonal microtubule (MT) through tubulin post-translational modifications and MT associated proteins (MAPs), tau and MAP6, were found in both development and TBI. Specifically, degenerating axons in both development and TBI underwent phosphorylation of tau and excessive tubulin tyrosination, suggesting MT instability and depolyermization. Conversely, nearby axons without degenerating morphologies, had increased MAP6 expression and maintenance of tubulin acetylation, suggesting enhanced MT stabilization, thereby supporting survival or repair. Quantitative proteomics revealed similar signaling pathways of axon degeneration and growth/repair, including protein clusters and networks. This comparison approach demonstrates how focused evaluation of developmental processes may provide insight into pathways initiated by TBI. In particular, the data suggest that TBI may reawaken dormant axonal programs that direct axons towards either degeneration or growth/repair, supporting further study in this area.
Collapse
Affiliation(s)
- Hailong Song
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Chen Chen
- Department of Computer Sciences, University of Missouri, Columbia, MO 65211, United States
| | - Brian Kelley
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Alexandra Tomasevich
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Hyoungjoo Lee
- Department of Biochemistry and Biophysics, Quantitative Proteomics Resource Core, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jean-Pierre Dolle
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jianlin Cheng
- Department of Computer Sciences, University of Missouri, Columbia, MO 65211, United States
| | - Benjamin Garcia
- Department of Biochemistry and Biophysics, Quantitative Proteomics Resource Core, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - David F Meaney
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Douglas H Smith
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
12
|
Unraveling axonal mechanisms of traumatic brain injury. Acta Neuropathol Commun 2022; 10:140. [PMID: 36131329 PMCID: PMC9494812 DOI: 10.1186/s40478-022-01414-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Axonal swellings (AS) are one of the neuropathological hallmark of axonal injury in several disorders from trauma to neurodegeneration. Current evidence proposes a role of perturbed Ca2+ homeostasis in AS formation, involving impaired axonal transport and focal distension of the axons. Mechanisms of AS formation, in particular moments following injury, however, remain unknown. Here we show that AS form independently from intra-axonal Ca2+ changes, which are required primarily for the persistence of AS in time. We further show that the majority of axonal proteins undergoing de/phosphorylation immediately following injury belong to the cytoskeleton. This correlates with an increase in the distance of the actin/spectrin periodic rings and with microtubule tracks remodeling within AS. Observed cytoskeletal rearrangements support axonal transport without major interruptions. Our results demonstrate that the earliest axonal response to injury consists in physiological adaptations of axonal structure to preserve function rather than in immediate pathological events signaling axonal destruction.
Collapse
|
13
|
The Mechanical Microenvironment Regulates Axon Diameters Visualized by Cryo-Electron Tomography. Cells 2022; 11:cells11162533. [PMID: 36010609 PMCID: PMC9406316 DOI: 10.3390/cells11162533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 12/21/2022] Open
Abstract
Axonal varicosities or swellings are enlarged structures along axon shafts and profoundly affect action potential propagation and synaptic transmission. These structures, which are defined by morphology, are highly heterogeneous and often investigated concerning their roles in neuropathology, but why they are present in the normal brain remains unknown. Combining confocal microscopy and cryo-electron tomography (Cryo-ET) with in vivo and in vitro systems, we report that non-uniform mechanical interactions with the microenvironment can lead to 10-fold diameter differences within an axon of the central nervous system (CNS). In the brains of adult Thy1-YFP transgenic mice, individual axons in the cortex displayed significantly higher diameter variation than those in the corpus callosum. When being cultured on lacey carbon film-coated electron microscopy (EM) grids, CNS axons formed varicosities exclusively in holes and without microtubule (MT) breakage, and they contained mitochondria, multivesicular bodies (MVBs), and/or vesicles, similar to the axonal varicosities induced by mild fluid puffing. Moreover, enlarged axon branch points often contain MT free ends leading to the minor branch. When the axons were fasciculated by mimicking in vivo axonal bundles, their varicosity levels reduced. Taken together, our results have revealed the extrinsic regulation of the three-dimensional ultrastructures of central axons by the mechanical microenvironment under physiological conditions.
Collapse
|
14
|
Pozo Devoto VM, Onyango IG, Stokin GB. Mitochondrial behavior when things go wrong in the axon. Front Cell Neurosci 2022; 16:959598. [PMID: 35990893 PMCID: PMC9389222 DOI: 10.3389/fncel.2022.959598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Axonal homeostasis is maintained by processes that include cytoskeletal regulation, cargo transport, synaptic activity, ionic balance, and energy supply. Several of these processes involve mitochondria to varying degrees. As a transportable powerplant, the mitochondria deliver ATP and Ca2+-buffering capabilities and require fusion/fission to maintain proper functioning. Taking into consideration the long distances that need to be covered by mitochondria in the axons, their transport, distribution, fusion/fission, and health are of cardinal importance. However, axonal homeostasis is disrupted in several disorders of the nervous system, or by traumatic brain injury (TBI), where the external insult is translated into physical forces that damage nervous tissue including axons. The degree of damage varies and can disconnect the axon into two segments and/or generate axonal swellings in addition to cytoskeletal changes, membrane leakage, and changes in ionic composition. Cytoskeletal changes and increased intra-axonal Ca2+ levels are the main factors that challenge mitochondrial homeostasis. On the other hand, a proper function and distribution of mitochondria can determine the recovery or regeneration of the axonal physiological state. Here, we discuss the current knowledge regarding mitochondrial transport, fusion/fission, and Ca2+ regulation under axonal physiological or pathological conditions.
Collapse
Affiliation(s)
- Victorio M. Pozo Devoto
- Translational Neuroscience and Ageing Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czechia
| | - Isaac G. Onyango
- Translational Neuroscience and Ageing Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czechia
| | - Gorazd B. Stokin
- Translational Neuroscience and Ageing Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czechia
- Division of Neurology, University Medical Centre, Ljubljana, Slovenia
- Department of Neurosciences, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
15
|
Bagnell AM, Sumner CJ, McCray BA. TRPV4: A trigger of pathological RhoA activation in neurological disease. Bioessays 2022; 44:e2100288. [PMID: 35297520 PMCID: PMC9295809 DOI: 10.1002/bies.202100288] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/14/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4), a member of the TRP superfamily, is a broadly expressed, cell surface-localized cation channel that is activated by a variety of environmental stimuli. Importantly, TRPV4 has been increasingly implicated in the regulation of cellular morphology. Here we propose that TRPV4 and the cytoskeletal remodeling small GTPase RhoA together constitute an environmentally sensitive signaling complex that contributes to pathological cell cytoskeletal alterations during neurological injury and disease. Supporting this hypothesis is our recent work demonstrating direct physical and bidirectional functional interactions of TRPV4 with RhoA, which can lead to activation of RhoA and reorganization of the actin cytoskeleton. Furthermore, a confluence of evidence implicates TRPV4 and/or RhoA in pathological responses triggered by a range of acute neurological insults ranging from stroke to traumatic injury. While initiated by a variety of insults, TRPV4-RhoA signaling may represent a common pathway that disrupts axonal regeneration and blood-brain barrier integrity. These insights also suggest that TRPV4 inhibition may represent a safe, feasible, and precise therapeutic strategy for limiting pathological TRPV4-RhoA activation in a range of neurological diseases.
Collapse
Affiliation(s)
- Anna M. Bagnell
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charlotte J. Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brett A. McCray
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Radomski KL, Zi X, Lischka FW, Noble MD, Galdzicki Z, Armstrong RC. Acute axon damage and demyelination are mitigated by 4-aminopyridine (4-AP) therapy after experimental traumatic brain injury. Acta Neuropathol Commun 2022; 10:67. [PMID: 35501931 PMCID: PMC9059462 DOI: 10.1186/s40478-022-01366-z] [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] [Received: 02/04/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Damage to long axons in white matter tracts is a major pathology in closed head traumatic brain injury (TBI). Acute TBI treatments are needed that protect against axon damage and promote recovery of axon function to prevent long term symptoms and neurodegeneration. Our prior characterization of axon damage and demyelination after TBI led us to examine repurposing of 4-aminopyridine (4-AP), an FDA-approved inhibitor of voltage-gated potassium (Kv) channels. 4-AP is currently indicated to provide symptomatic relief for patients with chronic stage multiple sclerosis, which involves axon damage and demyelination. We tested clinically relevant dosage of 4-AP as an acute treatment for experimental TBI and found multiple benefits in corpus callosum axons. This randomized, controlled pre-clinical study focused on the first week after TBI, when axons are particularly vulnerable. 4-AP treatment initiated one day post-injury dramatically reduced axon damage detected by intra-axonal fluorescence accumulations in Thy1-YFP mice of both sexes. Detailed electron microscopy in C57BL/6 mice showed that 4-AP reduced pathological features of mitochondrial swelling, cytoskeletal disruption, and demyelination at 7 days post-injury. Furthermore, 4-AP improved the molecular organization of axon nodal regions by restoring disrupted paranode domains and reducing Kv1.2 channel dispersion. 4-AP treatment did not resolve deficits in action potential conduction across the corpus callosum, based on ex vivo electrophysiological recordings at 7 days post-TBI. Thus, this first study of 4-AP effects on axon damage in the acute period demonstrates a significant decrease in multiple pathological hallmarks of axon damage after experimental TBI.
Collapse
Affiliation(s)
- Kryslaine L. Radomski
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Xiaomei Zi
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Fritz W. Lischka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Mark D. Noble
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Ave, Box 633, Rochester, NY 14642 USA
| | - Zygmunt Galdzicki
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| | - Regina C. Armstrong
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 USA
| |
Collapse
|
17
|
Sun C, Qi L, Cheng Y, Zhao Y, Gu C. Immediate induction of varicosities by transverse compression but not uniaxial stretch in axon mechanosensation. Acta Neuropathol Commun 2022; 10:7. [PMID: 35074017 PMCID: PMC8785443 DOI: 10.1186/s40478-022-01309-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 01/05/2022] [Indexed: 01/12/2023] Open
Abstract
Uniaxial stretch is believed to drive diffuse axonal injury (DAI) in mild traumatic brain injury (mTBI). Axonal varicosities are enlarged structures along axonal shafts and represent a hallmark feature of DAI. Here we report that axonal varicosities initiate in vivo immediately after head impact and are mainly induced by transverse compression but not uniaxial stretch. Vertical and lateral impacts to the mouse head induced axonal varicosities in distinct brain regions before any changes of microglial markers. Varicosities preferentially formed along axons perpendicular to impact direction. In cultured neurons, whereas 50% uniaxial strain was needed to rapidly induce axonal varicosities in a nanowrinkled stretch assay, physiologically-relevant transverse compression effectively induced axonal varicosities in a fluid puffing assay and can generate large but nonuniform deformation simulated by finite element analysis. Therefore, impact strength and direction may determine the threshold and spatial pattern of axonal varicosity initiation, respectively, partially resulting from intrinsic properties of axon mechanosensation.
Collapse
|
18
|
Varier P, Raju G, Madhusudanan P, Jerard C, Shankarappa SA. A Brief Review of In Vitro Models for Injury and Regeneration in the Peripheral Nervous System. Int J Mol Sci 2022; 23:816. [PMID: 35055003 PMCID: PMC8775373 DOI: 10.3390/ijms23020816] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 02/06/2023] Open
Abstract
Nerve axonal injury and associated cellular mechanisms leading to peripheral nerve damage are important topics of research necessary for reducing disability and enhancing quality of life. Model systems that mimic the biological changes that occur during human nerve injury are crucial for the identification of cellular responses, screening of novel therapeutic molecules, and design of neural regeneration strategies. In addition to in vivo and mathematical models, in vitro axonal injury models provide a simple, robust, and reductionist platform to partially understand nerve injury pathogenesis and regeneration. In recent years, there have been several advances related to in vitro techniques that focus on the utilization of custom-fabricated cell culture chambers, microfluidic chamber systems, and injury techniques such as laser ablation and axonal stretching. These developments seem to reflect a gradual and natural progression towards understanding molecular and signaling events at an individual axon and neuronal-soma level. In this review, we attempt to categorize and discuss various in vitro models of injury relevant to the peripheral nervous system and highlight their strengths, weaknesses, and opportunities. Such models will help to recreate the post-injury microenvironment and aid in the development of therapeutic strategies that can accelerate nerve repair.
Collapse
Affiliation(s)
| | | | | | | | - Sahadev A. Shankarappa
- Centre for Nanosciences & Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi 682041, India; (P.V.); (G.R.); (P.M.); (C.J.)
| |
Collapse
|
19
|
Yong Y, Hunter-Chang S, Stepanova E, Deppmann C. Axonal spheroids in neurodegeneration. Mol Cell Neurosci 2021; 117:103679. [PMID: 34678457 PMCID: PMC8742877 DOI: 10.1016/j.mcn.2021.103679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 10/20/2022] Open
Abstract
Axonal spheroids are bubble-like biological features that form on most degenerating axons, yet little is known about their influence on degenerative processes. Their formation and growth has been observed in response to various degenerative triggers such as injury, oxidative stress, inflammatory factors, and neurotoxic molecules. They often contain cytoskeletal elements and organelles, and, depending on the pathological insult, can colocalize with disease-related proteins such as amyloid precursor protein (APP), ubiquitin, and motor proteins. Initial formation of axonal spheroids depends on the disruption of axonal and membrane tension governed by cytoskeleton structure and calcium levels. Shortly after spheroid formation, the engulfment signal phosphatidylserine (PS) is exposed on the outer leaflet of spheroid plasma membrane, suggesting an important role for axonal spheroids in phagocytosis and debris clearance during degeneration. Spheroids can grow until they rupture, allowing pro-degenerative factors to exit the axon into extracellular space and accelerating neurodegeneration. Though much remains to be discovered in this area, axonal spheroid research promises to lend insight into the etiologies of neurodegenerative disease, and may be an important target for therapeutic intervention. This review summarizes over 100 years of work, describing what is known about axonal spheroid structure, regulation and function.
Collapse
Affiliation(s)
- Yu Yong
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Sarah Hunter-Chang
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Ekaterina Stepanova
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Christopher Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
20
|
Wu YH, Rosset S, Lee TR, Dragunow M, Park T, Shim V. In Vitro Models of Traumatic Brain Injury: A Systematic Review. J Neurotrauma 2021; 38:2336-2372. [PMID: 33563092 DOI: 10.1089/neu.2020.7402] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health challenge that is also the third leading cause of death worldwide. It is also the leading cause of long-term disability in children and young adults worldwide. Despite a large body of research using predominantly in vivo and in vitro rodent models of brain injury, there is no medication that can reduce brain damage or promote brain repair mainly due to our lack of understanding in the mechanisms and pathophysiology of the TBI. The aim of this review is to examine in vitro TBI studies conducted from 2008-2018 to better understand the TBI in vitro model available in the literature. Specifically, our focus was to perform a detailed analysis of the in vitro experimental protocols used and their subsequent biological findings. Our review showed that the uniaxial stretch is the most frequently used way of load application, accounting for more than two-thirds of the studies reviewed. The rate and magnitude of the loading were varied significantly from study to study but can generally be categorized into mild, moderate, and severe injuries. The in vitro studies reviewed here examined key processes in TBI pathophysiology such as membrane disruptions leading to ionic dysregulation, inflammation, and the subsequent damages to the microtubules and axons, as well as cell death. Overall, the studies examined in this review contributed to the betterment of our understanding of TBI as a disease process. Yet, our review also revealed the areas where more work needs to be done such as: 1) diversification of load application methods that will include complex loading that mimics in vivo head impacts; 2) more widespread use of human brain cells, especially patient-matched human cells in the experimental set-up; and 3) need for building a more high-throughput system to be able to discover effective therapeutic targets for TBI.
Collapse
Affiliation(s)
- Yi-Han Wu
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Samuel Rosset
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Tae-Rin Lee
- Advanced Institute of Convergence Technology, Seoul National University, Seoul, Korea
| | - Mike Dragunow
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Thomas Park
- Center for Brain Research, The University of Auckland, Auckland, New Zealand
- Department of Pharmacology, The University of Auckland, Auckland, New Zealand
| | - Vickie Shim
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
21
|
Pan X, Zhou Y, Hotulainen P, Meunier FA, Wang T. The axonal radial contractility: Structural basis underlying a new form of neural plasticity. Bioessays 2021; 43:e2100033. [PMID: 34145916 DOI: 10.1002/bies.202100033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/25/2022]
Abstract
Axons are the longest cellular structure reaching over a meter in the case of human motor axons. They have a relatively small diameter and contain several cytoskeletal elements that mediate both material and information exchange within neurons. Recently, a novel type of axonal plasticity, termed axonal radial contractility, has been unveiled. It is represented by dynamic and transient diameter changes of the axon shaft to accommodate the passages of large organelles. Mechanisms underpinning this plasticity are not fully understood. Here, we first summarised recent evidence of the functional relevance for axon radial contractility, then discussed the underlying structural basis, reviewing nanoscopic evidence of the subtle changes. Two models are proposed to explain how actomyosin rings are organised. Possible roles of non-muscle myosin II (NM-II) in axon degeneration are discussed. Finally, we discuss the concept of periodic functional nanodomains, which could sense extracellular cues and coordinate the axonal responses. Also see the video abstract here: https://youtu.be/ojCnrJ8RCRc.
Collapse
Affiliation(s)
- Xiaorong Pan
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Yimin Zhou
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Pirta Hotulainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Tong Wang
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China
| |
Collapse
|
22
|
Bradshaw DV, Knutsen AK, Korotcov A, Sullivan GM, Radomski KL, Dardzinski BJ, Zi X, McDaniel DP, Armstrong RC. Genetic inactivation of SARM1 axon degeneration pathway improves outcome trajectory after experimental traumatic brain injury based on pathological, radiological, and functional measures. Acta Neuropathol Commun 2021; 9:89. [PMID: 34001261 PMCID: PMC8130449 DOI: 10.1186/s40478-021-01193-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) causes chronic symptoms and increased risk of neurodegeneration. Axons in white matter tracts, such as the corpus callosum (CC), are critical components of neural circuits and particularly vulnerable to TBI. Treatments are needed to protect axons from traumatic injury and mitigate post-traumatic neurodegeneration. SARM1 protein is a central driver of axon degeneration through a conserved molecular pathway. Sarm1−/− mice with knockout (KO) of the Sarm1 gene enable genetic proof-of-concept testing of the SARM1 pathway as a therapeutic target. We evaluated Sarm1 deletion effects after TBI using a concussive model that causes traumatic axonal injury and progresses to CC atrophy at 10 weeks, indicating post-traumatic neurodegeneration. Sarm1 wild-type (WT) mice developed significant CC atrophy that was reduced in Sarm1 KO mice. Ultrastructural classification of pathology of individual axons, using electron microscopy, demonstrated that Sarm1 KO preserved more intact axons and reduced damaged or demyelinated axons. Longitudinal MRI studies in live mice identified significantly reduced CC volume after TBI in Sarm1 WT mice that was attenuated in Sarm1 KO mice. MR diffusion tensor imaging detected reduced fractional anisotropy in both genotypes while axial diffusivity remained higher in Sarm1 KO mice. Immunohistochemistry revealed significant attenuation of CC atrophy, myelin loss, and neuroinflammation in Sarm1 KO mice after TBI. Functionally, Sarm1 KO mice exhibited beneficial effects in motor learning and sleep behavior. Based on these findings, Sarm1 inactivation can protect axons and white matter tracts to improve translational outcomes associated with CC atrophy and post-traumatic neurodegeneration.
Collapse
|
23
|
Cuveillier C, Boulan B, Ravanello C, Denarier E, Deloulme JC, Gory-Fauré S, Delphin C, Bosc C, Arnal I, Andrieux A. Beyond Neuronal Microtubule Stabilization: MAP6 and CRMPS, Two Converging Stories. Front Mol Neurosci 2021; 14:665693. [PMID: 34025352 PMCID: PMC8131560 DOI: 10.3389/fnmol.2021.665693] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022] Open
Abstract
The development and function of the central nervous system rely on the microtubule (MT) and actin cytoskeletons and their respective effectors. Although the structural role of the cytoskeleton has long been acknowledged in neuronal morphology and activity, it was recently recognized to play the role of a signaling platform. Following this recognition, research into Microtubule Associated Proteins (MAPs) diversified. Indeed, historically, structural MAPs—including MAP1B, MAP2, Tau, and MAP6 (also known as STOP);—were identified and described as MT-binding and -stabilizing proteins. Extensive data obtained over the last 20 years indicated that these structural MAPs could also contribute to a variety of other molecular roles. Among multi-role MAPs, MAP6 provides a striking example illustrating the diverse molecular and cellular properties of MAPs and showing how their functional versatility contributes to the central nervous system. In this review, in addition to MAP6’s effect on microtubules, we describe its impact on the actin cytoskeleton, on neuroreceptor homeostasis, and its involvement in signaling pathways governing neuron development and maturation. We also discuss its roles in synaptic plasticity, brain connectivity, and cognitive abilities, as well as the potential relationships between the integrated brain functions of MAP6 and its molecular activities. In parallel, the Collapsin Response Mediator Proteins (CRMPs) are presented as examples of how other proteins, not initially identified as MAPs, fall into the broader MAP family. These proteins bind MTs as well as exhibiting molecular and cellular properties very similar to MAP6. Finally, we briefly summarize the multiple similarities between other classical structural MAPs and MAP6 or CRMPs.In summary, this review revisits the molecular properties and the cellular and neuronal roles of the classical MAPs, broadening our definition of what constitutes a MAP.
Collapse
|
24
|
Bradshaw DV, Kim Y, Fu A, Marion CM, Radomski KL, McCabe JT, Armstrong RC. Repetitive Blast Exposure Produces White Matter Axon Damage without Subsequent Myelin Remodeling: In Vivo Analysis of Brain Injury Using Fluorescent Reporter Mice. Neurotrauma Rep 2021; 2:180-192. [PMID: 34013219 PMCID: PMC8127063 DOI: 10.1089/neur.2020.0058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The potential effects of blast exposure on the brain health of military personnel have raised concerns and led to increased surveillance of blast exposures. Neuroimaging studies have reported white matter abnormalities in brains of service members with a history of blast exposure. However, blast effects on white matter microstructure remain poorly understood. As a novel approach to screen for white matter effects, transgenic mice that express fluorescent reporters to sensitively detect axon damage and myelin remodeling were exposed to simulated repetitive blasts (once/day on 5 consecutive days). Axons were visualized using Thy1-YFP-16 reporter mice that express yellow fluorescent protein (YFP) in a broad spectrum of neurons. Swelling along damaged axons forms varicosities that fill with YFP. The frequency and size of axonal varicosities were significantly increased in the corpus callosum (CC) and cingulum at 3 days after the final blast exposure, versus in sham procedures. CC immunolabeling for reactive astrocyte and microglial markers was also significantly increased. NG2CreER;mTmG mice were given tamoxifen (TMX) on days 2 and 3 after the final blast to induce fluorescent labeling of newly synthesized myelin membranes, indicating plasticity and/or repair. Myelin synthesis was not altered in the CC over the intervening 4 or 8 weeks after repetitive blast exposure. These experiments show the advantages of transgenic reporter mice for analysis of white matter injury that detects subtle, diffuse axon damage and the dynamic nature of myelin sheaths. These results show that repetitive low-level blast exposures produce infrequent but significant axon damage along with neuroinflammation in white matter.
Collapse
Affiliation(s)
- Donald V Bradshaw
- Graduate Program in Neuroscience, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Yeonho Kim
- Center for Neuroscience and Regenerative Medicine, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Amanda Fu
- Center for Neuroscience and Regenerative Medicine, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Christina M Marion
- Graduate Program in Neuroscience, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, Ohio, USA
| | - Kryslaine L Radomski
- Center for Neuroscience and Regenerative Medicine, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Joseph T McCabe
- Graduate Program in Neuroscience, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Center for Neuroscience and Regenerative Medicine, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Regina C Armstrong
- Graduate Program in Neuroscience, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Center for Neuroscience and Regenerative Medicine, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
25
|
Gu C. Rapid and Reversible Development of Axonal Varicosities: A New Form of Neural Plasticity. Front Mol Neurosci 2021; 14:610857. [PMID: 33613192 PMCID: PMC7886671 DOI: 10.3389/fnmol.2021.610857] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Axonal varicosities are enlarged, heterogeneous structures along axonal shafts, profoundly affecting axonal conduction and synaptic transmission. They represent a key pathological feature believed to develop via slow accumulation of axonal damage that occurs during irreversible degeneration, for example in mild traumatic brain injury (mTBI), Alzheimer's and Parkinson's diseases, and multiple sclerosis. Here this review first discusses recent in vitro results showing that axonal varicosities can be rapidly and reversibly induced by mechanical stress in cultured primary neurons from the central nervous system (CNS). This notion is further supported by in vivo studies revealing the induction of axonal varicosities across various brain regions in different mTBI mouse models, as a prominent feature of axonal pathology. Limited progress in understanding intrinsic and extrinsic regulatory mechanisms of axonal varicosity induction and development is further highlighted. Rapid and reversible formation of axonal varicosities likely plays a key role in CNS neuron mechanosensation and is a new form of neural plasticity. Future investigation in this emerging research field may reveal how to reverse axonal injury, contributing to the development of new strategies for treating brain injuries and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
26
|
Abstract
Several hypotheses have been put forth over time to explain how consciousness can be so rapidly lost, and then spontaneously regained, following mechanical head trauma. The knockout punch in boxing is a relatively homogenous form of traumatic brain injury and can thus be used to test the predictions of these hypotheses. While none of the hypotheses put forth can be considered fully verified, pore formation following stretching of the axonal cell membrane, mechanoporation, is a strong contender. We here argue that the theoretical foundation of mechanoporation can be strengthened by a comparison with the experimental method electroporation.
Collapse
Affiliation(s)
- Anders Hånell
- Section of Neurosurgery, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Elham Rostami
- Section of Neurosurgery, Department of Neuroscience, Uppsala University, Uppsala, Sweden
- Department of Neuroscience, Karolinska Institutet (KI), Stockholm, Sweden
| |
Collapse
|
27
|
Pernici CD, Rowe RK, Doughty PT, Madadi M, Lifshitz J, Murray TA. Longitudinal optical imaging technique to visualize progressive axonal damage after brain injury in mice reveals responses to different minocycline treatments. Sci Rep 2020; 10:7815. [PMID: 32385407 PMCID: PMC7210987 DOI: 10.1038/s41598-020-64783-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/17/2020] [Indexed: 12/14/2022] Open
Abstract
A high-resolution, three-dimensional, optical imaging technique for the murine brain was developed to identify the effects of different therapeutic windows for preclinical brain research. This technique tracks the same cells over several weeks. We conducted a pilot study of a promising drug to treat diffuse axonal injury (DAI) caused by traumatic brain injury, using two different therapeutic windows, as a means to demonstrate the utility of this novel longitudinal imaging technique. DAI causes immediate, sporadic axon damage followed by progressive secondary axon damage. We administered minocycline for three days commencing one hour after injury in one treatment group and beginning 72 hours after injury in another group to demonstrate the method’s ability to show how and when the therapeutic drug exerts protective and/or healing effects. Fewer varicosities developed in acutely treated mice while more varicosities resolved in mice with delayed treatment. For both treatments, the drug arrested development of new axonal damage by 30 days. In addition to evaluation of therapeutics for traumatic brain injury, this hybrid microlens imaging method should be useful to study other types of brain injury and neurodegeneration and cellular responses to treatment.
Collapse
Affiliation(s)
- Chelsea D Pernici
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, LA, USA
| | - Rachel K Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ, USA
| | - P Timothy Doughty
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, LA, USA
| | - Mahboubeh Madadi
- Department of Marketing and Business Analytics, Lucas College of Business, San Jose State University, San Jose, CA, USA
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA.,Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.,Phoenix Veterans Affairs Health Care System, Phoenix, AZ, USA
| | - Teresa A Murray
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, LA, USA.
| |
Collapse
|
28
|
Wang T, Li W, Martin S, Papadopulos A, Joensuu M, Liu C, Jiang A, Shamsollahi G, Amor R, Lanoue V, Padmanabhan P, Meunier FA. Radial contractility of actomyosin rings facilitates axonal trafficking and structural stability. J Cell Biol 2020; 219:e201902001. [PMID: 32182623 PMCID: PMC7199852 DOI: 10.1083/jcb.201902001] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 09/17/2019] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
Most mammalian neurons have a narrow axon, which constrains the passage of large cargoes such as autophagosomes that can be larger than the axon diameter. Radial axonal expansion must therefore occur to ensure efficient axonal trafficking. In this study, we reveal that the speed of various large cargoes undergoing axonal transport is significantly slower than that of small ones and that the transit of diverse-sized cargoes causes an acute, albeit transient, axonal radial expansion, which is immediately restored by constitutive axonal contractility. Using live super-resolution microscopy, we demonstrate that actomyosin-II controls axonal radial contractility and local expansion, and that NM-II filaments associate with periodic F-actin rings via their head domains. Pharmacological inhibition of NM-II activity significantly increases axon diameter by detaching the NM-II from F-actin and impacts the trafficking speed, directionality, and overall efficiency of long-range retrograde trafficking. Consequently, prolonged NM-II inactivation leads to disruption of periodic actin rings and formation of focal axonal swellings, a hallmark of axonal degeneration.
Collapse
Affiliation(s)
- Tong Wang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Wei Li
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Sally Martin
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
- The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Andreas Papadopulos
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Chunxia Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Anmin Jiang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Golnoosh Shamsollahi
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Rumelo Amor
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Vanessa Lanoue
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Frédéric A. Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
29
|
Rice J, Coutellier L, Weiner JL, Gu C. Region-specific interneuron demyelination and heightened anxiety-like behavior induced by adolescent binge alcohol treatment. Acta Neuropathol Commun 2019; 7:173. [PMID: 31703603 DOI: 10.1186/s40478-019-0829-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/13/2019] [Indexed: 12/12/2022] Open
Abstract
Adolescent binge drinking represents a major public health challenge and can lead to persistent neurological and mental conditions, but the underlying pathogenic mechanisms remain poorly understood. Using a mouse model of adolescent binge ethanol treatment (ABET), we found that this treatment induced behavioral changes associated with demyelination in different brain regions. After ABET, adolescent mice exhibited anxiogenic behaviors with no change in locomotion on the elevated plus maze, and impaired spatial memory indicated by a significant reduction in spontaneous alternation in the Y maze test. Both effects persisted into adulthood. Anatomical studies further showed that ABET induced a significant reduction of parvalbumin-positive (PV+) GABAergic interneurons and myelin density in the hippocampus and medial prefrontal cortex (mPFC). While these deficits in PV+ interneurons and myelin persisted into early adulthood in the hippocampus, the myelin density recovered in the mPFC. Moreover, whereas ABET mainly damaged myelin of PV+ axons in the hippocampus, it primarily damaged myelin of PV-negative axons in the mPFC. Thus, our findings reveal that an adolescent binge alcohol treatment regimen disrupts spatial working memory, increases anxiety-like behaviors, and exerts unique temporal and spatial patterns of gray matter demyelination in the hippocampus and mPFC.
Collapse
|
30
|
Marion CM, McDaniel DP, Armstrong RC. Sarm1 deletion reduces axon damage, demyelination, and white matter atrophy after experimental traumatic brain injury. Exp Neurol 2019; 321:113040. [PMID: 31445042 DOI: 10.1016/j.expneurol.2019.113040] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) often damages axons in white matter tracts and causes corpus callosum (CC) atrophy in chronic TBI patients. Injured axons encounter irreversible damage if transected, or alternatively may maintain continuity and subsequently either recover or degenerate. Secondary mechanisms can cause further axon damage, myelin pathology, and neuroinflammation. Molecular mechanisms regulating the progression of white matter pathology indicate potential therapeutic targets. SARM1 is essential for execution of the conserved axon death pathway. We examined white matter pathology following mild TBI with CC traumatic axonal injury in mice with Sarm1 gene deletion (Sarm1-/-). High resolution ultrastructural analysis at 3 days post-TBI revealed dramatically reduced axon damage in Sarm1-/- mice, as compared to Sarm1+/+ wild-type controls. Sarm1 deletion produced larger axons with thinner myelin, and attenuated TBI induced demyelination, i.e. myelin loss along apparently intact axons. At 6 weeks post-TBI, Sarm1-/- mice had less demyelination and thinner myelin than Sarm1+/+ mice, but axonal protection was no longer observed. We next used Thy1-YFP crosses to assess Sarm1 involvement in white matter neurodegeneration and neuroinflammation at 8 weeks post-TBI, when significant CC atrophy indicates chronic pathology. Thy1-YFP expression demonstrated continued CC axon damage yet absence of overt cortical pathology. Importantly, significant CC atrophy in Thy1-YFP/Sarm1+/+ mice was associated with reduced neurofilament immunolabeling of axons. Both effects were attenuated in Thy1-YFP/Sarm1-/- mice. Surprisingly, Thy1-YFP/Sarm1-/- mice had increased CC astrogliosis. This study demonstrates that Sarm1 inactivation reduces demyelination, and white matter atrophy after TBI, while the post-injury stage impacts when axon protection is effective.
Collapse
Affiliation(s)
- Christina M Marion
- Center for Neuroscience and Regenerative Medicine, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Program in Neuroscience, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Dennis P McDaniel
- Biomedical Instrumentation Center, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Microbiology and Immunology, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Regina C Armstrong
- Center for Neuroscience and Regenerative Medicine, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology and Genetics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Program in Neuroscience, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| |
Collapse
|
31
|
Datar A, Ameeramja J, Bhat A, Srivastava R, Mishra A, Bernal R, Prost J, Callan-Jones A, Pullarkat PA. The Roles of Microtubules and Membrane Tension in Axonal Beading, Retraction, and Atrophy. Biophys J 2019; 117:880-891. [PMID: 31427070 DOI: 10.1016/j.bpj.2019.07.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/23/2019] [Accepted: 07/26/2019] [Indexed: 02/02/2023] Open
Abstract
Axonal beading, or the formation of a series of swellings along the axon, and retraction are commonly observed shape transformations that precede axonal atrophy in Alzheimer's disease, Parkinson's disease, and other neurodegenerative conditions. The mechanisms driving these morphological transformations are poorly understood. Here, we report controlled experiments that can induce either beading or retraction and follow the time evolution of these responses. By making quantitative analysis of the shape modes under different conditions, measurement of membrane tension, and using theoretical considerations, we argue that membrane tension is the main driving force that pushes cytosol out of the axon when microtubules are degraded, causing axonal thinning. Under pharmacological perturbation, atrophy is always retrograde, and this is set by a gradient in the microtubule stability. The nature of microtubule depolymerization dictates the type of shape transformation, vis-à-vis beading or retraction. Elucidating the mechanisms of these shape transformations may facilitate development of strategies to prevent or arrest axonal atrophy due to neurodegenerative conditions.
Collapse
Affiliation(s)
| | | | - Alka Bhat
- Raman Research Institute, Bengaluru, India
| | | | | | - Roberto Bernal
- Departamento de Física, SMAT-C, Universidad de Santiago de Chile, Santiago, Chile
| | - Jacques Prost
- Laboratoire Physico Chimie Curie, Institut Curie, 10 PSL Research University, CNRS UMR168, Paris, France; Mechanobiology Institute, National University of Singapore, Singapore
| | - Andrew Callan-Jones
- Laboratoire Matière et Systèmes Complexes, Université Paris Diderot, Paris, France.
| | | |
Collapse
|
32
|
Nickel M, Eid F, Jukkola P, Gu C. Copper chelation and autoimmunity differentially impact myelin in the hippocampal-prefrontal circuit. J Neuroimmunol 2019; 334:576998. [PMID: 31254928 DOI: 10.1016/j.jneuroim.2019.576998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 01/19/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. About 50% of MS patients develop deficits in learning, memory and executive function, which are accompanied by demyelinating lesions in the hippocampus and/or prefrontal cortex (PFC). Why demyelination in these regions occurs in some patients but not in others and what is the underlying mechanism remain unclear. Here we report that myelin density in the hippocampus and PFC is markedly reduced in the cuprizone model, but not in the chronic experimental autoimmune encephalomyelitis. These two models can be used for studying different neuropathophysiological aspects of demyelinating diseases.
Collapse
Affiliation(s)
- Mara Nickel
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Farida Eid
- College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Peter Jukkola
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Chen Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA; Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
33
|
Pernici CD, Kemp BS, Murray TA. Time course images of cellular injury and recovery in murine brain with high-resolution GRIN lens system. Sci Rep 2019; 9:7946. [PMID: 31138885 PMCID: PMC6538613 DOI: 10.1038/s41598-019-44174-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/10/2019] [Indexed: 12/20/2022] Open
Abstract
Time course, in vivo imaging of brain cells is crucial to fully understand the progression of secondary cellular damage and recovery in murine models of injury. We have combined high-resolution gradient index lens technology with a model of diffuse axonal injury in rodents to enable repeated visualization of fine features of individual cells in three-dimensional space over several weeks. For example, we recorded changes in morphology in the same axons in the external capsule numerous times over 30 to 60 days, before and after induced traumatic brain injury. We observed the expansion of secondary injury and limited recovery of individual axons in this subcortical white matter tract over time. In another application, changes in microglial activation state were visualized in the penumbra region of mice before and after ischemia induced by middle carotid artery occlusion. The ability to collect a series of high-resolution images of cellular features of the same cells pre- and post-injury enables a unique opportunity to study the progression of damage, spontaneous healing, and effects of therapeutics in mouse models of neurodegenerative disease and brain injury.
Collapse
Affiliation(s)
- Chelsea D Pernici
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Benjamin S Kemp
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Teresa A Murray
- Center for Biomedical Engineering and Rehabilitation Sciences, Louisiana Tech University, Ruston, Louisiana, USA.
| |
Collapse
|
34
|
Shenton FC, Pyner S. Transient receptor potential vanilloid type 4 is expressed in vasopressinergic neurons within the magnocellular subdivision of the rat paraventricular nucleus of the hypothalamus. J Comp Neurol 2018; 526:3035-3044. [PMID: 30078222 PMCID: PMC6492187 DOI: 10.1002/cne.24514] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 07/19/2018] [Accepted: 07/19/2018] [Indexed: 01/28/2023]
Abstract
Changes in plasma osmolality can drive changes in the output from brain centres known to control cardiovascular homeostasis, such as the paraventricular nucleus of the hypothalamus (PVN). Within the PVN hypotonicity reduces the firing rate of parvocellular neurons, a neuronal pool known to be involved in modulating sympathetic vasomotor tone. Also present in the PVN is the transient receptor potential vanilloid type 4 (TRPV4) ion channel. Activation of TRPV4 within the PVN mimics the reduction in firing rate of the parvocellular neurons but it is unknown if these neurons express the channel. We used neuronal tracing and immunohistochemistry to investigate which neurons expressed the TRPV4 ion channel protein and its relationship with neurons known to play a role in plasma volume regulation. Spinally projecting preautonomic neurons within the PVN were labelled after spinal cord injection of FluoroGold (FG). This was followed by immunolabelling with anti‐TRPV4 antibody in combination with either anti‐oxytocin (OXT) or anti‐vasopressin (AVP). The TRPV4 ion channel was expressed on 63% of the vasopressinergic magnocellular neurosecretory cells found predominantly within the posterior magnocellular division of the PVN. Oxytocinergic neurons and FG labelled preautonomic neurons were present in the same location, but were distinct from the TRPV4/vasopressin expressing neurons. Vasopressinergic neurons within the supraoptic nucleus (SON) were also found to express TRPV4 and the fibres extending between the SON and PVN. In conclusion within the PVN, TRPV4 is well placed to respond to changes in osmolality by regulating vasopressin secretion, which in turn influences sympathetic output via preautonomic neurons.
Collapse
Affiliation(s)
- F C Shenton
- Department of Biosciences, Durham University, Durham, UK
| | - S Pyner
- Department of Biosciences, Durham University, Durham, UK
| |
Collapse
|
35
|
Experimental Traumatic Brain Injury Identifies Distinct Early and Late Phase Axonal Conduction Deficits of White Matter Pathophysiology, and Reveals Intervening Recovery. J Neurosci 2018; 38:8723-8736. [PMID: 30143572 PMCID: PMC6181309 DOI: 10.1523/jneurosci.0819-18.2018] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/15/2018] [Accepted: 07/10/2018] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injury (TBI) patients often exhibit slowed information processing speed that can underlie diverse symptoms. Processing speed depends on neural circuit function at synapses, in the soma, and along axons. Long axons in white matter (WM) tracts are particularly vulnerable to TBI. We hypothesized that disrupted axon–myelin interactions that slow or block action potential conduction in WM tracts may contribute to slowed processing speed after TBI. Concussive TBI in male/female mice was used to produce traumatic axonal injury in the corpus callosum (CC), similar to WM pathology in human TBI cases. Compound action potential velocity was slowed along myelinated axons at 3 d after TBI with partial recovery by 2 weeks, suggesting early demyelination followed by remyelination. Ultrastructurally, dispersed demyelinated axons and disorganized myelin attachment to axons at paranodes were apparent within CC regions exhibiting traumatic axonal injury. Action potential conduction is exquisitely sensitive to paranode abnormalities. Molecular identification of paranodes and nodes of Ranvier detected asymmetrical paranode pairs and abnormal heminodes after TBI. Fluorescent labeling of oligodendrocyte progenitors in NG2CreER;mTmG mice showed increased synthesis of new membranes extended along axons to paranodes, indicating remyelination after TBI. At later times after TBI, an overall loss of conducting axons was observed at 6 weeks followed by CC atrophy at 8 weeks. These studies identify a progression of both myelinated axon conduction deficits and axon–myelin pathology in the CC, implicating WM injury in impaired information processing at early and late phases after TBI. Furthermore, the intervening recovery reveals a potential therapeutic window. SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is a major global health concern. Across the spectrum of TBI severities, impaired information processing can contribute to diverse functional deficits that underlie persistent symptoms. We used experimental TBI to exploit technical advantages in mice while modeling traumatic axonal injury in white matter tracts, which is a key pathological feature of human TBI. A combination of approaches revealed slowed and failed signal conduction along with damage to the structure and molecular composition of myelinated axons in the white matter after TBI. An early regenerative response was not sustained yet reveals a potential time window for intervention. These insights into white matter abnormalities underlying axon conduction deficits can inform strategies to improve treatment options for TBI patients.
Collapse
|
36
|
Servello D, Gu Y, Gu C. A Microbiomechanical System for Studying Varicosity Formation and Recovery in Central Neuron Axons. J Vis Exp 2018. [PMID: 29757278 DOI: 10.3791/57202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Axonal varicosities are enlarged structures along the shafts of axons with a high degree of heterogeneity. They are present not only in brains with neurodegenerative diseases or injuries, but also in the normal brain. Here, we describe a newly-established micromechanical system to rapidly, reliably, and reversibly induce axonal varicosities, allowing us to understand the mechanisms governing varicosity formation and heterogeneous protein composition. This system represents a novel means to evaluate the effects of compression and shear stress on different subcellular compartments of neurons, different from other in vitro systems that mainly focus on the effect of stretching. Importantly, owing to the unique features of our system, we recently made a novel discovery showing that the application of pressurized fluid can rapidly and reversibly induce axonal varicosities through the activation of a transient receptor potential channel. Our biomechanical system can be utilized conveniently in combination with drug perfusion, live cell imaging, calcium imaging, and patch clamp recording. Therefore, this method can be adopted for studying mechanosensitive ion channels, axonal transport regulation, axonal cytoskeleton dynamics, calcium signaling, and morphological changes related to traumatic brain injury.
Collapse
Affiliation(s)
- Dustin Servello
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University
| | - Yuanzheng Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University; Biogen
| | - Chen Gu
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University; Department of Biological Chemistry and Pharmacology, The Ohio State University;
| |
Collapse
|
37
|
Primary Traumatic Axonopathy in Mice Subjected to Impact Acceleration: A Reappraisal of Pathology and Mechanisms with High-Resolution Anatomical Methods. J Neurosci 2018; 38:4031-4047. [PMID: 29567804 DOI: 10.1523/jneurosci.2343-17.2018] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 02/21/2018] [Accepted: 03/15/2018] [Indexed: 11/21/2022] Open
Abstract
Traumatic axonal injury (TAI) is a common neuropathology in traumatic brain injury and is featured by primary injury to axons. Here, we generated TAI with impact acceleration of the head in male Thy1-eYFP-H transgenic mice in which specific populations of neurons and their axons are labeled with yellow fluorescent protein. This model results in axonal lesions in multiple axonal tracts along with blood-brain barrier disruption and neuroinflammation. The corticospinal tract, a prototypical long tract, is severely affected and is the focus of this study. Using optimized CLARITY at single-axon resolution, we visualized the entire corticospinal tract volume from the pons to the cervical spinal cord in 3D and counted the total number of axonal lesions and their progression over time. Our results divulged the presence of progressive traumatic axonopathy that was maximal at the pyramidal decussation. The perikarya of injured corticospinal neurons atrophied, but there was no evidence of neuronal cell death. We also used CLARITY at single-axon resolution to explore the role of the NMNAT2-SARM1 axonal self-destruction pathway in traumatic axonopathy. When we interfered with this pathway by genetically ablating SARM1 or by pharmacological strategies designed to increase levels of Nicotinamide (Nam), a feedback inhibitor of SARM1, we found a significant reduction in the number of axonal lesions early after injury. Our findings show that high-resolution neuroanatomical strategies reveal important features of TAI with biological implications, especially the progressive axonopathic nature of TAI and the role of the NMNAT2-SARM1 pathway in the early stages of axonopathy.SIGNIFICANCE STATEMENT In the first systematic application of novel high-resolution neuroanatomical tools in neuropathology, we combined CLARITY with 2-photon microscopy, optimized for detection of single axonal lesions, to reconstruct the injured mouse brainstem in a model of traumatic axonal injury (TAI) that is a common pathology associated with traumatic brain injury. The 3D reconstruction of the corticospinal tract at single-axon resolution allowed for a more advanced level of qualitative and quantitative understanding of TAI. Using this model, we showed that TAI is an axonopathy with a prominent role of the NMNAT2-SARM1 molecular pathway, that is also implicated in peripheral neuropathy. Our results indicate that high-resolution anatomical models of TAI afford a level of detail and sensitivity that is ideal for testing novel molecular and biomechanical hypotheses.
Collapse
|