1
|
Widner J, Faust PL, Louis ED, Fujita H. Axonal pathology differentially affects human Purkinje cell subtypes in the essential tremor cerebellum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.633063. [PMID: 39974874 PMCID: PMC11838201 DOI: 10.1101/2025.01.26.633063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The cerebellar cortex is organized into discrete regions populated by molecularly distinct Purkinje cells (PCs), the sole cortical output neurons. While studies in animal models have shown that PC subtypes differ in their vulnerability to disease, our understanding of human PC subtype and vulnerability remains limited. Here, we demonstrate that human cerebellar regions specialized for motor vs cognitive functions (lobule HV vs Crus I) contain distinct PC populations characterized by specific molecular and anatomical features, which show selective vulnerability in essential tremor (ET), a cerebellar degenerative disorder. Using a known PC subtype marker, neurofilament heavy chain (NEFH), we found that motor lobule HV contains PCs with high NEFH expression, while cognitive lobule Crus I contains PCs with low NEFH expression in post-mortem samples from healthy controls. In the same cerebella, PC axons in lobule HV were 2.2-fold thicker than those in Crus I. Across lobules, axon caliber positively correlated with NEFH expression. In ET cerebella, we identified motor lobule-specific PC axon pathology with a 1.5-fold reduction in caliber and increased axon variability in lobule HV, while Crus I axons were unaffected. Tremor severity and duration in ET correlated with axon diameter variability selectively in lobule HV PCs. Given that axonal caliber is a major determinant of neural signaling capacity, our results (1) suggest that disrupted cerebellar corticonuclear signaling is occurring in ET, (2) provide evidence of region-specific PC subtypes in the human cerebellum and offer insight into how selective PC vulnerability may contribute to the pathophysiology of cerebellar degeneration.
Collapse
Affiliation(s)
- James Widner
- Movement Disorder Section, Dept. of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Phyllis L. Faust
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center and the New York Presbyterian Hospital, New York, New York, USA
| | - Elan D. Louis
- Movement Disorder Section, Dept. of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hirofumi Fujita
- Movement Disorder Section, Dept. of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
2
|
Ching K, Sagasti A. Caliber of zebrafish touch-sensory axons is dynamic in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.04.626901. [PMID: 39713467 PMCID: PMC11661087 DOI: 10.1101/2024.12.04.626901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Cell shape is crucial to cell function, particularly in neurons. The cross-sectional diameter, also known as caliber, of axons and dendrites is an important parameter of neuron shape, best appreciated for its influence on the speed of action potential propagation. Many studies of axon caliber focus on cell-wide regulation and assume that caliber is static. Here, we have characterized local variation and dynamics of axon caliber in vivo using the peripheral axons of zebrafish touch-sensing neurons at embryonic stages, prior to sex determination. To obtain absolute measurements of caliber in vivo, we paired sparse membrane labeling with super-resolution microscopy of neurons in live fish. We observed that axon segments had varicose or "pearled" morphologies, and thus vary in caliber along their length, consistent with reports from mammalian systems. Sister axon segments originating from the most proximal branch point in the axon arbor had average calibers that were uncorrelated with each other. Axon caliber also tapered across the branch point. Varicosities and caliber, overall, were dynamic on the timescale of minutes, and dynamicity changed over the course of development. By measuring the caliber of axons adjacent to dividing epithelial cells, we found that skin cell division is one aspect of the cellular microenvironment that may drive local differences and dynamics in axon caliber. Our findings support the possibility that spatial and temporal variation in axon caliber could significantly influence neuronal physiology.
Collapse
Affiliation(s)
- Kaitlin Ching
- Department of Cell, Molecular, and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Alvaro Sagasti
- Department of Cell, Molecular, and Developmental Biology, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
3
|
Devarakonda SS, Basha S, Pithakumar A, L B T, Mukunda DC, Rodrigues J, K A, Biswas S, Pai AR, Belurkar S, Mahato KK. Molecular mechanisms of neurofilament alterations and its application in assessing neurodegenerative disorders. Ageing Res Rev 2024; 102:102566. [PMID: 39481763 DOI: 10.1016/j.arr.2024.102566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Neurofilaments are intermediate filaments present in neurons. These provide structural support and maintain the size and shape of the neurons. Dysregulation, mutation, and aggregation of neurofilaments raise the levels of these proteins in the blood and cerebrospinal fluid (CSF), which are characteristic features of axonal damage and certain rare neurological diseases, such as Giant Axonal Neuropathy and Charcot-Mare-Tooth disease. Understanding the structure, dynamics, and function of neurofilaments has been greatly enhanced by a diverse range of biochemical and preclinical investigations conducted over more than four decades. Recently, there has been a resurgence of interest in post-translational modifications of neurofilaments, such as phosphorylation, aggregation, mutation, oxidation, etc. Over the past twenty years, several rare disorders have been studied from structural alterations of neurofilaments. These disorders are monitored by fluid biomarkers such as neurofilament light chains. Currently, there are many tools, such as Enzyme-Linked Immunosorbent Assay, Electrochemiluminescence Assay, Single-Molecule Array, Western/immunoblotting, etc., in use to assess the neurofilament proteins in Blood and CSF. However, all these techniques utilize expensive, non-specific, or antibody-based methods, which make them unsuitable for routine screening of neurodegenerative disorders. This provides room to search for newer sensitive, cost-effective, point-of-care tools for rapid screening of the disease. For a long time, the molecular mechanisms of neurofilaments have been poorly understood due to insufficient research attempts, and a deeper understanding of them remains elusive. Therefore, this review aims to highlight the available literature on molecular mechanisms of neurofilaments and the function of neurofilaments in axonal transport, axonal conduction, axonal growth, and neurofilament aggregation, respectively. Further, this review discusses the role of neurofilaments as potential biomarkers for the identification of several neurodegenerative diseases in clinical laboratory practice.
Collapse
Affiliation(s)
| | - Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Anjana Pithakumar
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Thoshna L B
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | | | - Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Ameera K
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Shimul Biswas
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Sushma Belurkar
- Department of Pathology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India.
| |
Collapse
|
4
|
Adelman JW, Sukowaty AT, Partridge KJ, Gawrys JE, Terhune SS, Ebert AD. Stabilizing microtubules aids neurite structure and disrupts syncytia formation in human cytomegalovirus-infected human forebrain neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608340. [PMID: 39229072 PMCID: PMC11370344 DOI: 10.1101/2024.08.16.608340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Human cytomegalovirus (HCMV) is a prolific human herpesvirus that infects most individuals by adulthood. While typically asymptomatic in adults, congenital infection can induce serious neurological symptoms including hearing loss, visual deficits, cognitive impairment, and microcephaly in 10-15% of cases. HCMV has been shown to infect most neural cells with our group recently demonstrating this capacity in stem cell-derived forebrain neurons. Infection of neurons induces deleterious effects on calcium dynamics and electrophysiological function paired with gross restructuring of neuronal morphology. Here, we utilize an iPSC-derived model of the human forebrain to demonstrate how HCMV infection induces syncytia, drives neurite retraction, and remodels microtubule networks to promote viral production and release. We establish that HCMV downregulates microtubule associated proteins at 14 days postinfection while simultaneously sparing other cytoskeletal elements, and this includes HCMV-driven alterations to microtubule stability. Further, we pharmacologically modulate microtubule dynamics using paclitaxel (stabilize) and colchicine (destabilize) to examine the effects on neurite structure, syncytial morphology, assembly compartment formation, and viral release. With paclitaxel, we found improvement of neurite outgrowth with a corresponding disruption to HCMV-induced syncytia formation and Golgi network disruptions but with limited impact on viral titers. Together, these data suggest that HCMV infection-induced disruption of microtubules in human cortical neurons can be partially mitigated with microtubule stabilization, suggesting a potential avenue for future neuroprotective therapeutic exploration.
Collapse
Affiliation(s)
- Jacob W Adelman
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Andrew T Sukowaty
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kaitlyn J Partridge
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jessica E. Gawrys
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott S. Terhune
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
- Marquette University and Medical College of Wisconsin Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
5
|
Kotaich F, Caillol D, Bomont P. Neurofilaments in health and Charcot-Marie-Tooth disease. Front Cell Dev Biol 2023; 11:1275155. [PMID: 38164457 PMCID: PMC10758125 DOI: 10.3389/fcell.2023.1275155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024] Open
Abstract
Neurofilaments (NFs) are the most abundant component of mature neurons, that interconnect with actin and microtubules to form the cytoskeleton. Specifically expressed in the nervous system, NFs present the particularity within the Intermediate Filament family of being formed by four subunits, the neurofilament light (NF-L), medium (NF-M), heavy (NF-H) proteins and α-internexin or peripherin. Here, we review the current knowledge on NF proteins and neurofilaments, from their domain structures and their model of assembly to the dynamics of their transport and degradation along the axon. The formation of the filament and its behaviour are regulated by various determinants, including post-transcriptional (miRNA and RBP proteins) and post-translational (phosphorylation and ubiquitination) modifiers. Altogether, the complex set of modifications enable the neuron to establish a stable but elastic NF array constituting the structural scaffold of the axon, while permitting the local expression of NF proteins and providing the dynamics necessary to fulfil local demands and respond to stimuli and injury. Thus, in addition to their roles in mechano-resistance, radial axonal outgrowth and nerve conduction, NFs control microtubule dynamics, organelle distribution and neurotransmission at the synapse. We discuss how the studies of neurodegenerative diseases with NF aggregation shed light on the biology of NFs. In particular, the NEFL and NEFH genes are mutated in Charcot-Marie-Tooth (CMT) disease, the most common inherited neurological disorder of the peripheral nervous system. The clinical features of the CMT forms (axonal CMT2E, CMT2CC; demyelinating CMT1F; intermediate I-CMT) with symptoms affecting the central nervous system (CNS) will allow us to further investigate the physiological roles of NFs in the brain. Thus, NF-CMT mouse models exhibit various degrees of sensory-motor deficits associated with CNS symptoms. Cellular systems brought findings regarding the dominant effect of NF-L mutants on NF aggregation and transport, although these have been recently challenged. Neurofilament detection without NF-L in recessive CMT is puzzling, calling for a re-examination of the current model in which NF-L is indispensable for NF assembly. Overall, we discuss how the fundamental and translational fields are feeding each-other to increase but also challenge our knowledge of NF biology, and to develop therapeutic avenues for CMT and neurodegenerative diseases with NF aggregation.
Collapse
Affiliation(s)
| | | | - Pascale Bomont
- ERC team, NeuroMyoGene Institute-Pathophysiology and Genetics of Neuron and Muscle, Inserm U1315, CNRS UMR5261, University of Lyon 1, Lyon, France
| |
Collapse
|
6
|
Schwann cell functions in peripheral nerve development and repair. Neurobiol Dis 2023; 176:105952. [PMID: 36493976 DOI: 10.1016/j.nbd.2022.105952] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
The glial cell of the peripheral nervous system (PNS), the Schwann cell (SC), counts among the most multifaceted cells of the body. During development, SCs secure neuronal survival and participate in axonal path finding. Simultaneously, they orchestrate the architectural set up of the developing nerves, including the blood vessels and the endo-, peri- and epineurial layers. Perinatally, in rodents, SCs radially sort and subsequently myelinate individual axons larger than 1 μm in diameter, while small calibre axons become organised in non-myelinating Remak bundles. SCs have a vital role in maintaining axonal health throughout life and several specialized SC types perform essential functions at specific locations, such as terminal SC at the neuromuscular junction (NMJ) or SC within cutaneous sensory end organs. In addition, neural crest derived satellite glia maintain a tight communication with the soma of sensory, sympathetic, and parasympathetic neurons and neural crest derivatives are furthermore an indispensable part of the enteric nervous system. The remarkable plasticity of SCs becomes evident in the context of a nerve injury, where SC transdifferentiate into intriguing repair cells, which orchestrate a regenerative response that promotes nerve repair. Indeed, the multiple adaptations of SCs are captivating, but remain often ill-resolved on the molecular level. Here, we summarize and discuss the knowns and unknowns of the vast array of functions that this single cell type can cover in peripheral nervous system development, maintenance, and repair.
Collapse
|
7
|
Metzner K, Darawsha O, Wang M, Gaur N, Cheng Y, Rödiger A, Frahm C, Witte OW, Perocchi F, Axer H, Grosskreutz J, Brill MS. Age-dependent increase of cytoskeletal components in sensory axons in human skin. Front Cell Dev Biol 2022; 10:965382. [PMID: 36393849 PMCID: PMC9664158 DOI: 10.3389/fcell.2022.965382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/12/2022] [Indexed: 01/24/2023] Open
Abstract
Aging is a complex process characterized by several molecular and cellular imbalances. The composition and stability of the neuronal cytoskeleton is essential for the maintenance of homeostasis, especially in long neurites. Using human skin biopsies containing sensory axons from a cohort of healthy individuals, we investigate alterations in cytoskeletal content and sensory axon caliber during aging via quantitative immunostainings. Cytoskeletal components show an increase with aging in both sexes, while elevation in axon diameter is only evident in males. Transcriptomic data from aging males illustrate various patterns in gene expression during aging. Together, the data suggest gender-specific changes during aging in peripheral sensory axons, possibly influencing cytoskeletal functionality and axonal caliber. These changes may cumulatively increase susceptibility of aged individuals to neurodegenerative diseases.
Collapse
Affiliation(s)
- Klara Metzner
- Department of Neurology, Jena University Hospital, Jena, Germany,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Omar Darawsha
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Mengzhe Wang
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Nayana Gaur
- Department of Neurology, Jena University Hospital, Jena, Germany,Laboratory Animal Centre, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Yiming Cheng
- Helmholtz Diabetes Center (HDC), Helmholtz Center Munich, Institute for Diabetes and Obesity, Munich, Germany
| | | | - Christiane Frahm
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W. Witte
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Fabiana Perocchi
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany,Helmholtz Diabetes Center (HDC), Helmholtz Center Munich, Institute for Diabetes and Obesity, Munich, Germany,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Hubertus Axer
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Julian Grosskreutz
- Precision Neurology of the University of Lübeck, Lübeck, Germany,PMI Cluster, University of Lübeck, Lübeck, Germany
| | - Monika S. Brill
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany,Munich Cluster of Systems Neurology (SyNergy), Munich, Germany,*Correspondence: Monika S. Brill,
| |
Collapse
|
8
|
Serratos IN, Hernández-Pérez E, Campos C, Aschner M, Santamaría A. An Update on the Critical Role of α-Synuclein in Parkinson's Disease and Other Synucleinopathies: from Tissue to Cellular and Molecular Levels. Mol Neurobiol 2021; 59:620-642. [PMID: 34750787 DOI: 10.1007/s12035-021-02596-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022]
Abstract
The aggregation of alpha-synuclein (α-Syn) plays a critical role in the development of Parkinson's disease (PD) and other synucleinopathies. α-Syn, which is encoded by the SNCA gene, is a lysine-rich soluble amphipathic protein normally expressed in neurons. Located in the cytosolic domain, this protein has the ability to remodel itself in plasma membranes, where it assumes an alpha-helix conformation. However, the protein can also adopt another conformation rich in cross-beta sheets, undergoing mutations and post-translational modifications, then leading the protein to an unusual aggregation in the form of Lewy bodies (LB), which are cytoplasmic inclusions constituted predominantly by α-Syn. Pathogenic mechanisms affecting the structural and functional stability of α-Syn - such as endoplasmic reticulum stress, Golgi complex fragmentation, disfunctional protein degradation systems, aberrant interactions with mitochondrial membranes and nuclear DNA, altered cytoskeleton dynamics, disrupted neuronal plasmatic membrane, dysfunctional vesicular transport, and formation of extracellular toxic aggregates - contribute all to the pathogenic progression of PD and synucleinopathies. In this review, we describe the collective knowledge on this topic and provide an update on the critical role of α-Syn aggregates, both at the cellular and molecular levels, in the deregulation of organelles affecting the cellular homeostasis and leading to neuronal cell death in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Iris N Serratos
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Mexico City, Mexico
| | - Elizabeth Hernández-Pérez
- Departamento de Ciencias de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Mexico City, Mexico
| | - Carolina Campos
- Departamento de Ciencias de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Mexico City, Mexico.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, SSA, 14269, Mexico City, Mexico.
| |
Collapse
|
9
|
Studying Neuronal Biology Using Spinning Disc Confocal Microscopy. Methods Mol Biol 2021. [PMID: 34028722 DOI: 10.1007/978-1-0716-1402-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Cytoskeletal integrity is essential for neuronal complexity and functionality. Certain inherited neurological diseases are associated with mutated genes that directly or indirectly compromise cytoskeletal stability. While the large size and complexity of the neurons grown in culture poses certain challenges for imaging, live-cell imaging is an excellent approach to determine the morphological consequences of such mutants. This protocol details the use of spinning disk confocal microscopy and image analysis tools to evaluate branching and neurite length of healthy iPSC-derived glutamatergic neurons that express specific fluorescent proteins. The protocols can be adapted to neuronal cell lines of choice by the investigator.
Collapse
|
10
|
The emerging role of the sympathetic nervous system in skeletal muscle motor innervation and sarcopenia. Ageing Res Rev 2021; 67:101305. [PMID: 33610815 DOI: 10.1016/j.arr.2021.101305] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/06/2021] [Accepted: 02/15/2021] [Indexed: 12/30/2022]
Abstract
Examining neural etiologic factors'role in the decline of neuromuscular function with aging is essential to our understanding of the mechanisms underlying sarcopenia, the age-dependent decline in muscle mass, force and power. Innervation of the skeletal muscle by both motor and sympathetic axons has been established, igniting interest in determining how the sympathetic nervous system (SNS) affect skeletal muscle composition and function throughout the lifetime. Selective expression of the heart and neural crest derivative 2 gene in peripheral SNs increases muscle mass and force regulating skeletal muscle sympathetic and motor innervation; improving acetylcholine receptor stability and NMJ transmission; preventing inflammation and myofibrillar protein degradation; increasing autophagy; and probably enhancing protein synthesis. Elucidating the role of central SNs will help to define the coordinated response of the visceral and neuromuscular system to physiological and pathological challenges across ages. This review discusses the following questions: (1) Does the SNS regulate skeletal muscle motor innervation? (2) Does the SNS regulate presynaptic and postsynaptic neuromuscular junction (NMJ) structure and function? (3) Does sympathetic neuron (SN) regulation of NMJ transmission decline with aging? (4) Does maintenance of SNs attenuate aging sarcopenia? and (5) Do central SN group relays influence sympathetic and motor muscle innervation?
Collapse
|
11
|
Ren N, Ito S, Hafizi H, Beggs JM, Stevenson IH. Model-based detection of putative synaptic connections from spike recordings with latency and type constraints. J Neurophysiol 2020; 124:1588-1604. [DOI: 10.1152/jn.00066.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Detecting synaptic connections using large-scale extracellular spike recordings is a difficult statistical problem. Here, we develop an extension of a generalized linear model that explicitly separates fast synaptic effects and slow background fluctuations in cross-correlograms between pairs of neurons while incorporating circuit properties learned from the whole network. This model outperforms two previously developed synapse detection methods in the simulated networks and recovers plausible connections from hundreds of neurons in in vitro multielectrode array data.
Collapse
Affiliation(s)
- Naixin Ren
- Department of Psychological Sciences, University of Connecticut, Storrs, Connecticut
| | - Shinya Ito
- Santa Cruz Institute for Particle Physics, University of California, Santa Cruz, California
| | - Hadi Hafizi
- Department of Physics, Indiana University, Bloomington, Indiana
| | - John M. Beggs
- Department of Physics, Indiana University, Bloomington, Indiana
| | - Ian H. Stevenson
- Department of Psychological Sciences, University of Connecticut, Storrs, Connecticut
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
12
|
Hernández-Jasso I, Domínguez-Del-Toro E, Delgado-García JM, Quintanar JL. Recovery of sciatic nerve with complete transection in rats treated with leuprolide acetate: A gonadotropin-releasing hormone agonist. Neurosci Lett 2020; 739:135439. [PMID: 33132176 DOI: 10.1016/j.neulet.2020.135439] [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: 04/28/2020] [Revised: 09/28/2020] [Accepted: 10/02/2020] [Indexed: 11/16/2022]
Abstract
It has been reported that the Gonadotropin-releasing hormone (GnRH) and its agonist leuprolide acetate (LA) can act as promoters of nerve regeneration. The aim of this study is to evaluate the effect of LA in a complete transection model. Sciatic nerve injury (SNI) was performed using a complete nerve transection and immediately repaired by epineural sutures. Rats were divided into three groups: SHAM, SNI treated with LA (SNI + LA) or saline solution (SNI + SS) for 5 weeks. Sciatic nerve regeneration was evaluated by kinematic gait analyzes, electrophysiological, morphological and biochemical tests. SNI + LA group had a functional recovery in kinematic gait, an increase in ankle angle value and a faster walking speed, compound muscle action potential amplitude, nerve conduction velocity (NCV). Furthermore, the number of myelinated axons and microtubule-associated protein 2 (MAP-2) expression were also higher compared to SS group. In conclusion, LA treatment improves of gait, walking speed, NCV, axons morphometry and MAP-2 expression in rats with sciatic nerve complete transection. These results suggest that LA can be a potential treatment for peripheral nerve injuries.
Collapse
Affiliation(s)
- Irma Hernández-Jasso
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes, Mexico
| | | | | | - J Luis Quintanar
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Basic Science Center, Autonomous University of Aguascalientes, Aguascalientes, Mexico.
| |
Collapse
|
13
|
Belrose JL, Prasad A, Sammons MA, Gibbs KM, Szaro BG. Comparative gene expression profiling between optic nerve and spinal cord injury in Xenopus laevis reveals a core set of genes inherent in successful regeneration of vertebrate central nervous system axons. BMC Genomics 2020; 21:540. [PMID: 32758133 PMCID: PMC7430912 DOI: 10.1186/s12864-020-06954-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The South African claw-toed frog, Xenopus laevis, is uniquely suited for studying differences between regenerative and non-regenerative responses to CNS injury within the same organism, because some CNS neurons (e.g., retinal ganglion cells after optic nerve crush (ONC)) regenerate axons throughout life, whereas others (e.g., hindbrain neurons after spinal cord injury (SCI)) lose this capacity as tadpoles metamorphose into frogs. Tissues from these CNS regions (frog ONC eye, tadpole SCI hindbrain, frog SCI hindbrain) were used in a three-way RNA-seq study of axotomized CNS axons to identify potential core gene expression programs for successful CNS axon regeneration. RESULTS Despite tissue-specific changes in expression dominating the injury responses of each tissue, injury-induced changes in gene expression were nonetheless shared between the two axon-regenerative CNS regions that were not shared with the non-regenerative region. These included similar temporal patterns of gene expression and over 300 injury-responsive genes. Many of these genes and their associated cellular functions had previously been associated with injury responses of multiple tissues, both neural and non-neural, from different species, thereby demonstrating deep phylogenetically conserved commonalities between successful CNS axon regeneration and tissue regeneration in general. Further analyses implicated the KEGG adipocytokine signaling pathway, which links leptin with metabolic and gene regulatory pathways, and a novel gene regulatory network with genes regulating chromatin accessibility at its core, as important hubs in the larger network of injury response genes involved in successful CNS axon regeneration. CONCLUSIONS This study identifies deep, phylogenetically conserved commonalities between CNS axon regeneration and other examples of successful tissue regeneration and provides new targets for studying the molecular underpinnings of successful CNS axon regeneration, as well as a guide for distinguishing pro-regenerative injury-induced changes in gene expression from detrimental ones in mammals.
Collapse
Affiliation(s)
- Jamie L Belrose
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Aparna Prasad
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA
| | - Kurt M Gibbs
- Department of Biology and Chemistry, Morehead State University, Morehead, KY, 40351, USA
| | - Ben G Szaro
- Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
- Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY, 12222, USA.
| |
Collapse
|
14
|
Andersson E, Janelidze S, Lampinen B, Nilsson M, Leuzy A, Stomrud E, Blennow K, Zetterberg H, Hansson O. Blood and cerebrospinal fluid neurofilament light differentially detect neurodegeneration in early Alzheimer's disease. Neurobiol Aging 2020; 95:143-153. [PMID: 32810755 PMCID: PMC7649343 DOI: 10.1016/j.neurobiolaging.2020.07.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 01/08/2023]
Abstract
Cerebrospinal fluid (CSF) neurofilament light (NfL) concentration has reproducibly been shown to reflect neurodegeneration in brain disorders, including Alzheimer's disease (AD). NfL concentration in blood correlates with the corresponding CSF levels, but few studies have directly compared the reliability of these 2 markers in sporadic AD. Herein, we measured plasma and CSF concentrations of NfL in 478 cognitively unimpaired (CU) subjects, 227 patients with mild cognitive impairment, and 113 patients with AD dementia. We found that the concentration of NfL in CSF, but not in plasma, was increased in response to Aβ pathology in CU subjects. Both CSF and plasma NfL concentrations were increased in patients with mild cognitive impairment and AD dementia. Furthermore, only NfL in CSF was associated with reduced white matter microstructure in CU subjects. Finally, in a transgenic mouse model of AD, CSF NfL increased before serum NfL in response to the development of Aβ pathology. In conclusion, NfL in CSF may be a more reliable biomarker of neurodegeneration than NfL in blood in preclinical sporadic AD.
Collapse
Affiliation(s)
- Emelie Andersson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden.
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Björn Lampinen
- Clinical Sciences Lund, Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Markus Nilsson
- Clinical Sciences Lund, Department of Radiology, Lund University, Lund, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
15
|
Prokop A. Cytoskeletal organization of axons in vertebrates and invertebrates. J Cell Biol 2020; 219:e201912081. [PMID: 32369543 PMCID: PMC7337489 DOI: 10.1083/jcb.201912081] [Citation(s) in RCA: 44] [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: 12/18/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
The maintenance of axons for the lifetime of an organism requires an axonal cytoskeleton that is robust but also flexible to adapt to mechanical challenges and to support plastic changes of axon morphology. Furthermore, cytoskeletal organization has to adapt to axons of dramatically different dimensions, and to their compartment-specific requirements in the axon initial segment, in the axon shaft, at synapses or in growth cones. To understand how the cytoskeleton caters to these different demands, this review summarizes five decades of electron microscopic studies. It focuses on the organization of microtubules and neurofilaments in axon shafts in both vertebrate and invertebrate neurons, as well as the axon initial segments of vertebrate motor- and interneurons. Findings from these ultrastructural studies are being interpreted here on the basis of our contemporary molecular understanding. They strongly suggest that axon architecture in animals as diverse as arthropods and vertebrates is dependent on loosely cross-linked bundles of microtubules running all along axons, with only minor roles played by neurofilaments.
Collapse
Affiliation(s)
- Andreas Prokop
- School of Biology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
16
|
|
17
|
Gordon BA. Neurofilaments in disease: what do we know? Curr Opin Neurobiol 2020; 61:105-115. [PMID: 32151970 PMCID: PMC7198337 DOI: 10.1016/j.conb.2020.02.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/25/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022]
Abstract
Neurofilaments are proteins selectively expressed in the cytoskeleton of neurons, and increased levels are a marker of damage. Elevated neurofilament levels can serve as a marker of ongoing disease activity as well as a tool to measure response to therapeutic intervention. The potential utility of neurofilaments has drastically increased as recent advances have made it possible to measure levels in both the cerebrospinal fluid and blood. There is mounting evidence that neurofilament light chain (NfL) and phosphorylated neurofilament heavy chain (NfH) are abnormal in a host of neurodegenerative diseases. In this review we examine how both of these proteins behave across diseases and what we know about how these biomarkers relate to in vivo white matter pathology and each other.
Collapse
Affiliation(s)
- Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, MO, USA; Psychological & Brain Sciences, Washington University in St. Louis, MO, USA.
| |
Collapse
|
18
|
Much More Than a Scaffold: Cytoskeletal Proteins in Neurological Disorders. Cells 2020; 9:cells9020358. [PMID: 32033020 PMCID: PMC7072452 DOI: 10.3390/cells9020358] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 02/08/2023] Open
Abstract
Recent observations related to the structure of the cytoskeleton in neurons and novel cytoskeletal abnormalities involved in the pathophysiology of some neurological diseases are changing our view on the function of the cytoskeletal proteins in the nervous system. These efforts allow a better understanding of the molecular mechanisms underlying neurological diseases and allow us to see beyond our current knowledge for the development of new treatments. The neuronal cytoskeleton can be described as an organelle formed by the three-dimensional lattice of the three main families of filaments: actin filaments, microtubules, and neurofilaments. This organelle organizes well-defined structures within neurons (cell bodies and axons), which allow their proper development and function through life. Here, we will provide an overview of both the basic and novel concepts related to those cytoskeletal proteins, which are emerging as potential targets in the study of the pathophysiological mechanisms underlying neurological disorders.
Collapse
|
19
|
Jiménez-Romero F, Bis-Humbert C, García-Fuster MJ. Adolescent morphine induces emotional signs of withdrawal paired with neurotoxicity selectively in male rats: Female resilience. Neurosci Lett 2020; 715:134625. [DOI: 10.1016/j.neulet.2019.134625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/08/2019] [Accepted: 11/09/2019] [Indexed: 10/25/2022]
|
20
|
Piao F, Chen Y, Yu L, Shi X, Liu X, Jiang L, Yang G, Wang N, Gao B, Zhang C. 2,5-hexanedione-induced deregulation of axon-related microRNA expression in rat nerve tissues. Toxicol Lett 2019; 320:95-102. [PMID: 31760062 DOI: 10.1016/j.toxlet.2019.11.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/18/2019] [Accepted: 11/18/2019] [Indexed: 12/31/2022]
Abstract
Exposure to organic solvent in industry, including n-hexane is correlated with central-peripheral axonopathy, which is mediated by its active metabolite, 2,5-hexanedione (HD). However, the underlying mechanism is still largely unknown. Recently identified microRNAs (miRNAs) may play important roles in toxicant exposure and in the process of toxicant-induced neuropathys. To examine the role of miRNAs in HD-induced toxicity, neuropathic animal model was successfully built. miRNA microarray analysis revealed 105 differentially expressed miRNAs after HD exposure. Bioinformatics analysis showed that "Axon" and "Neurotrophin Signaling Pathway" was the top significant GO term and pathway, respectively. 7 miRNAs both related to "Axon" and "Neurotrophin Signaling Pathway" were screened out and further confirmed by Real-Time PCR. Correspondingly, the deregulation expression levels of proteins of four target genes (GSK3β, Map3k1, BDNF and MAP1B) were further confirmed via western blot, verifying the results of gene target analysis. Taken together, our results showed that the axon-related miRNAs to be associated with MAP1B or neurotrophin signal pathways changed in nerve tissues following HD exposure. These miRNAs may play important roles in HD-induced neurotoxicity.
Collapse
Affiliation(s)
- Fengyuan Piao
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian 116044, PR China; Comprehensive Laboratory, Affiliated Zhongshan Hospital of Dalian University, Dalian 116044, PR China
| | - Yang Chen
- Department of Biotechnology, Basic Medical College, Dalian Medical University, Dalian 116044, PR China
| | - Li Yu
- College of Laboratory Medicine, Dalian Medical University, Dalian 116044, PR China
| | - Xiaoxia Shi
- Department of Occupational and Environmental Health, Dalian Medical University, Dalian 116044, PR China
| | - Xiaofang Liu
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian 116044, PR China
| | - Liping Jiang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian 116044, PR China
| | - Guang Yang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian 116044, PR China
| | - Ningning Wang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian 116044, PR China
| | - Bihu Gao
- Department of Nephrology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116044, PR China.
| | - Cong Zhang
- Department of Food Nutrition and Safety, Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
21
|
Hahn I, Voelzmann A, Liew YT, Costa-Gomes B, Prokop A. The model of local axon homeostasis - explaining the role and regulation of microtubule bundles in axon maintenance and pathology. Neural Dev 2019; 14:11. [PMID: 31706327 PMCID: PMC6842214 DOI: 10.1186/s13064-019-0134-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Axons are the slender, cable-like, up to meter-long projections of neurons that electrically wire our brains and bodies. In spite of their challenging morphology, they usually need to be maintained for an organism's lifetime. This makes them key lesion sites in pathological processes of ageing, injury and neurodegeneration. The morphology and physiology of axons crucially depends on the parallel bundles of microtubules (MTs), running all along to serve as their structural backbones and highways for life-sustaining cargo transport and organelle dynamics. Understanding how these bundles are formed and then maintained will provide important explanations for axon biology and pathology. Currently, much is known about MTs and the proteins that bind and regulate them, but very little about how these factors functionally integrate to regulate axon biology. As an attempt to bridge between molecular mechanisms and their cellular relevance, we explain here the model of local axon homeostasis, based on our own experiments in Drosophila and published data primarily from vertebrates/mammals as well as C. elegans. The model proposes that (1) the physical forces imposed by motor protein-driven transport and dynamics in the confined axonal space, are a life-sustaining necessity, but pose a strong bias for MT bundles to become disorganised. (2) To counterbalance this risk, MT-binding and -regulating proteins of different classes work together to maintain and protect MT bundles as necessary transport highways. Loss of balance between these two fundamental processes can explain the development of axonopathies, in particular those linking to MT-regulating proteins, motors and transport defects. With this perspective in mind, we hope that more researchers incorporate MTs into their work, thus enhancing our chances of deciphering the complex regulatory networks that underpin axon biology and pathology.
Collapse
Affiliation(s)
- Ines Hahn
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - André Voelzmann
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Yu-Ting Liew
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Beatriz Costa-Gomes
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK
| | - Andreas Prokop
- Manchester Academic Health Science Centre, Faculty of Biology, Medicine and Health, The University of Manchester, School of Biology, Manchester, UK.
| |
Collapse
|
22
|
Fenn JD, Monsma PC, Brown A. Axonal neurofilaments exhibit frequent and complex folding behaviors. Cytoskeleton (Hoboken) 2019; 75:258-280. [PMID: 29683261 DOI: 10.1002/cm.21448] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/30/2018] [Accepted: 04/03/2018] [Indexed: 01/19/2023]
Abstract
Neurofilaments are flexible cytoskeletal polymers that are capable of folding and unfolding between their bouts of bidirectional movement along axons. Here we present a detailed characterization of this behavior in cultured neurons using kymograph analysis with approximately 30 ms temporal resolution. We analyzed 781 filaments ranging from 0.6-42 µm in length. We observed complex behaviors including pinch folds, hairpin folds, orientation changes (flips), and occasional severing and annealing events. On average, the filaments spent approximately 40% of their time in some sort of folded configuration. A small proportion of filaments (4%) moved while folded, but most (96%) moved in an outstretched configuration. Collectively, our observations suggest that motors may interact with neurofilaments at multiple points along their length, but preferentially at their ends. In addition, the prevalence of neurofilament folding and the tendency of neurofilaments to straighten out when they move, suggest that an important function of the movement of these polymers in axons may be to maintain them in an outstretched and longitudinally co-aligned configuration. Thus, neurofilament movement may function as much to organize these polymers as to move them, and this could explain why they spend so much time engaged in apparently unproductive bidirectional movement.
Collapse
Affiliation(s)
- J Daniel Fenn
- Department of Neuroscience, Ohio State University, Columbus, Ohio, 43210.,Medical Scientist Training Program, Ohio State University, Columbus, Ohio, 43210
| | - Paula C Monsma
- Department of Neuroscience, Ohio State University, Columbus, Ohio, 43210
| | - Anthony Brown
- Department of Neuroscience, Ohio State University, Columbus, Ohio, 43210
| |
Collapse
|
23
|
Rodrigues ACZ, Messi ML, Wang ZM, Abba MC, Pereyra A, Birbrair A, Zhang T, O’Meara M, Kwan P, Lopez EIS, Willis MS, Mintz A, Files DC, Furdui C, Oppenheim RW, Delbono O. The sympathetic nervous system regulates skeletal muscle motor innervation and acetylcholine receptor stability. Acta Physiol (Oxf) 2019; 225:e13195. [PMID: 30269419 PMCID: PMC7224611 DOI: 10.1111/apha.13195] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
AIM Symptoms of autonomic failure are frequently the presentation of advanced age and neurodegenerative diseases that impair adaptation to common physiologic stressors. The aim of this work was to examine the interaction between the sympathetic and motor nervous system, the involvement of the sympathetic nervous system (SNS) in neuromuscular junction (NMJ) presynaptic motor function, the stability of postsynaptic molecular organization, and the skeletal muscle composition and function. METHODS Since muscle weakness is a symptom of diseases characterized by autonomic dysfunction, we studied the impact of regional sympathetic ablation on muscle motor innervation by using transcriptome analysis, retrograde tracing of the sympathetic outflow to the skeletal muscle, confocal and electron microscopy, NMJ transmission by electrophysiological methods, protein analysis, and state of the art microsurgical techniques, in C57BL6, MuRF1KO and Thy-1 mice. RESULTS We found that the SNS regulates motor nerve synaptic vesicle release, skeletal muscle transcriptome, muscle force generated by motor nerve activity, axonal neurofilament phosphorylation, myelin thickness, and myofibre subtype composition and CSA. The SNS also modulates the levels of postsynaptic membrane acetylcholine receptor by regulating the Gαi2 -Hdac4-Myogenin-MuRF1pathway, which is prevented by the overexpression of the guanine nucleotide-binding protein Gαi2 (Q205L), a constitutively active mutant G protein subunit. CONCLUSION The SNS regulates NMJ transmission, maintains optimal Gαi2 expression, and prevents any increase in Hdac4, myogenin, MuRF1, and miR-206. SNS ablation leads to upregulation of MuRF1, muscle atrophy, and downregulation of postsynaptic AChR. Our findings are relevant to clinical conditions characterized by progressive decline of sympathetic innervation, such as neurodegenerative diseases and aging.
Collapse
Affiliation(s)
- Anna C. Z. Rodrigues
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Maria L. Messi
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Zhong-Min Wang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Martin C. Abba
- Basic and Applied Immunological Research Center (CINIBA), School of Medicine, National University of La Plata, Buenos Aires, Argentina
| | - Andrea Pereyra
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Alexander Birbrair
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Tan Zhang
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Meaghan O’Meara
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ping Kwan
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Elsa I. S. Lopez
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Monte S. Willis
- Department of Pathology, McAllister Heart Institute, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - D. Clark Files
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine, Pulmonary, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Cristina Furdui
- Department of Internal Medicine, Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Ronald W. Oppenheim
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Osvaldo Delbono
- Department of Internal Medicine, Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Neuroscience Program, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
24
|
Wang S, Ji D, Yang Q, Li M, Ma Z, Zhang S, Li H. NEFLb impairs early nervous system development via regulation of neuron apoptosis in zebrafish. J Cell Physiol 2018; 234:11208-11218. [PMID: 30569449 DOI: 10.1002/jcp.27771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
Neurofilament light chain (NEFL), a subunit of neurofilament, has been shown to play an important role in pathogenic neurodegenerative disease and in radial axonal growth. However, information remains largely lacking regarding the function of NEFL in early development to date. In this study, we demonstrated the presence of two nefl genes, nefla and neflb, in zebrafish, generated by fish-specific third round genome duplication. These duplicated nefl genes were predominantly expressed in the nervous system with an overlapping and distinct expression pattern. Both gene knockdown and rescue experiments show that it was neflb rather than nefla that played an indispensable role in nervous system development. It was also found that neflb knockdown resulted in striking apoptosis of the neurons in the brain and spinal cord, leading to morphological defects such as brain structure disorder and trunk bending. Thus, we report a previously uncharacterized role of NEFL that NEFLb impairs the early development of zebrafish nervous system via regulation of the neuron apoptosis in the brain and spinal cord.
Collapse
Affiliation(s)
- Su Wang
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China.,Laboratory for Evolution & Development, Department of Marine Biology, Ocean University of China, Qingdao, China
| | - Dongrui Ji
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China.,Laboratory for Evolution & Development, Department of Marine Biology, Ocean University of China, Qingdao, China
| | - Qingyun Yang
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China.,Laboratory for Evolution & Development, Department of Marine Biology, Ocean University of China, Qingdao, China
| | - Mingyue Li
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China.,Laboratory for Evolution & Development, Department of Marine Biology, Ocean University of China, Qingdao, China
| | - Zengyu Ma
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China.,Laboratory for Evolution & Development, Department of Marine Biology, Ocean University of China, Qingdao, China
| | - Shicui Zhang
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China.,Laboratory for Evolution & Development, Department of Marine Biology, Ocean University of China, Qingdao, China
| | - Hongyan Li
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, China.,Laboratory for Evolution & Development, Department of Marine Biology, Ocean University of China, Qingdao, China
| |
Collapse
|
25
|
Costa AR, Pinto-Costa R, Sousa SC, Sousa MM. The Regulation of Axon Diameter: From Axonal Circumferential Contractility to Activity-Dependent Axon Swelling. Front Mol Neurosci 2018; 11:319. [PMID: 30233318 PMCID: PMC6131297 DOI: 10.3389/fnmol.2018.00319] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/17/2018] [Indexed: 01/08/2023] Open
Abstract
In the adult nervous system axon caliber varies widely amongst different tracts. When considering a given axon, its diameter can further fluctuate in space and time, according to processes including the distribution of organelles and activity-dependent mechanisms. In addition, evidence is emerging supporting that in axons circumferential tension/contractility is present. Axonal diameter is generically regarded as being regulated by neurofilaments. When neurofilaments are absent or low, microtubule-dependent mechanisms can also contribute to the regulation of axon caliber. Despite this knowledge, the fine-tune mechanisms controlling diameter and circumferential tension throughout the lifetime of an axon, remain largely elusive. Recent data supports the role of the actin-spectrin-based membrane periodic skeleton and of non-muscle myosin II in the control of axon diameter. However, the cytoskeletal arrangement that underlies circumferential axonal contraction and expansion is still to be discovered. Here, we discuss in a critical viewpoint the existing knowledge on the regulation of axon diameter, with a specific focus on the possible role played by the axonal actin cytoskeleton.
Collapse
Affiliation(s)
- Ana Rita Costa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Instituto de Inovação e Investigação em Saúde, University of Porto, Porto, Portugal
| | - Rita Pinto-Costa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Instituto de Inovação e Investigação em Saúde, University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Sara Castro Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Instituto de Inovação e Investigação em Saúde, University of Porto, Porto, Portugal
| | - Mónica Mendes Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Instituto de Inovação e Investigação em Saúde, University of Porto, Porto, Portugal
| |
Collapse
|
26
|
Gedrova S, Galik J, Marsala M, Zavodska M, Pavel J, Sulla I, Gajdos M, Lukac I, Kafka J, Ledecky V, Sulla I, Karasova M, Reichel P, Trbolova A, Capik I, Lukacova V, Bimbova K, Bacova M, Stropkovska A, Lukacova N. Neuroprotective effect of local hypothermia in a computer-controlled compression model in minipig: Correlation of tissue sparing along the rostro-caudal axis with neurological outcome. Exp Ther Med 2017; 15:254-270. [PMID: 29399061 PMCID: PMC5769223 DOI: 10.3892/etm.2017.5432] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/20/2017] [Indexed: 11/05/2022] Open
Abstract
This study investigated the neuroprotective efficacy of local hypothermia in a minipig model of spinal cord injury (SCI) induced by a computer-controlled impactor device. The tissue integrity observed at the injury epicenter, and up to 3 cm cranially and caudally from the lesion site correlated with motor function. A computer-controlled device produced contusion lesions at L3 level with two different degrees of tissue sparing, depending upon pre-set impact parameters (8N- and 15N-force impact). Hypothermia with cold (4°C) saline or Dulbecco's modified Eagle's medium (DMEM)/F12 culture medium was applied 30 min after SCI (for 5 h) via a perfusion chamber (flow 2 ml/min). After saline hypothermia, the 8N-SCI group achieved faster recovery of hind limb function and the ability to walk from one to three steps at nine weeks in comparison with non-treated animals. Such improvements were not observed in saline-treated animals subjected to more severe 15N-SCI or in the group treated with DMEM/F12 medium. It was demonstrated that the tissue preservation in the cranial and caudal segments immediately adjacent to the lesion, and neurofilament protection in the lateral columns may be essential for modulation of the key spinal microcircuits leading to a functional outcome. Tissue sparing observed only in the caudal sections, even though significant, was not sufficient for functional improvement in the 15N-SCI model.
Collapse
Affiliation(s)
- Stefania Gedrova
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| | - Jan Galik
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| | - Martin Marsala
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic.,Neuroregeneration Laboratory, Department of Anesthesiology, University of California-San Diego, La Jolla, CA 92037, USA
| | - Monika Zavodska
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| | - Jaroslav Pavel
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| | - Igor Sulla
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic.,Hospital of Slovak Railways, 040 01 Kosice, Slovak Republic
| | - Miroslav Gajdos
- Department of Neurosurgery, Faculty of Medicine, University of Pavol Jozef Safarik, 040 01 Kosice, Slovak Republic
| | - Imrich Lukac
- Department of Neurosurgery, Faculty of Medicine, University of Pavol Jozef Safarik, 040 01 Kosice, Slovak Republic
| | - Jozef Kafka
- Department of Neurosurgery, Faculty of Medicine, University of Pavol Jozef Safarik, 040 01 Kosice, Slovak Republic
| | - Valent Ledecky
- University of Veterinary Medicine and Pharmacy, 041 81 Kosice, Slovak Republic
| | - Igor Sulla
- University of Veterinary Medicine and Pharmacy, 041 81 Kosice, Slovak Republic
| | - Martina Karasova
- University of Veterinary Medicine and Pharmacy, 041 81 Kosice, Slovak Republic
| | - Peter Reichel
- University of Veterinary Medicine and Pharmacy, 041 81 Kosice, Slovak Republic
| | - Alexandra Trbolova
- University of Veterinary Medicine and Pharmacy, 041 81 Kosice, Slovak Republic
| | - Igor Capik
- University of Veterinary Medicine and Pharmacy, 041 81 Kosice, Slovak Republic
| | - Viktoria Lukacova
- Faculty of Economics, Technical University of Kosice, 040 01 Kosice, Slovak Republic
| | - Katarina Bimbova
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| | - Maria Bacova
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| | - Andrea Stropkovska
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| | - Nadezda Lukacova
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Kosice, Slovak Republic
| |
Collapse
|
27
|
Rutherford NJ, Brooks M, Riffe CJ, Gorion KMM, Howard JK, Dhillon JKS, Giasson BI. Prion-like transmission of α-synuclein pathology in the context of an NFL null background. Neurosci Lett 2017; 661:114-120. [PMID: 28964772 DOI: 10.1016/j.neulet.2017.09.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/26/2017] [Indexed: 02/02/2023]
Abstract
Neurofilaments are a major component of the axonal cytoskeleton in neurons and have been implicated in a number of neurodegenerative diseases due to their presence within characteristic pathological inclusions. Their contributions to these diseases are not yet fully understood, but previous studies investigated the effects of ablating the obligate subunit of neurofilaments, low molecular mass neurofilament subunit (NFL), on disease phenotypes in transgenic mouse models of Alzheimer's disease and tauopathy. Here, we tested the effects of ablating NFL in α-synuclein M83 transgenic mice expressing the human pathogenic A53T mutation, by breeding them onto an NFL null background. The induction and spread of α-synuclein inclusion pathology was triggered by the injection of preformed α-synuclein fibrils into the gastrocnemius muscle or hippocampus in M83 versus M83/NFL null mice. We observed no difference in the post-injection time to motor-impairment and paralysis endpoint or amount and distribution of α-synuclein inclusion pathology in the muscle injected M83 and M83/NFL null mice. Hippocampal injected M83/NFL null mice displayed subtle region-specific differences in the amount of α-synuclein inclusions however, pathology was observed in the same regions as the M83 mice. Overall, we observed only minor differences in the induction and transmission of α-synuclein pathology in these induced models of synucleinopathy in the presence or absence of NFL. This suggests that NFL and neurofilaments do not play a major role in influencing the induction and transmission of α-synuclein aggregation.
Collapse
Affiliation(s)
- Nicola J Rutherford
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Mieu Brooks
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Cara J Riffe
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Kimberly-Marie M Gorion
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Jasie K Howard
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Jess-Karan S Dhillon
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Benoit I Giasson
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
28
|
Developmental neurotoxicity of the hippocampus following in utero exposure to methylmercury: impairment in cell signaling. Arch Toxicol 2017; 92:513-527. [PMID: 28821999 DOI: 10.1007/s00204-017-2042-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/10/2017] [Indexed: 01/01/2023]
Abstract
In this study, we assessed some hippocampal signaling cascades and behavioral impairments in 30-day-old rat pups prenatally exposed to methylmercury (MeHg). Pregnant rats were exposed to 1.0 or 2.0 mg/kg MeHg by gavage in alternated days from gestational day 5 until parturition. We found increased anxiety-like and decreased exploration behavior evaluated by open field test and deficit of both short- and long-term memories by novel object recognition task, respectively, in MeHg-treated pups. Downregulated PI3K/Akt/mTOR pathway and activated/hypophosphorylated (Ser9) GSK3β in MeHg-treated pups could be upstream of hyperphosphorylated Tau (Ser396) destabilizing microtubules and contributing to neural dysfunction in the hippocampus of these rats. Hyperphosphorylated/activated p38MAPK and downregulated phosphoErk1/2 support a role for mitogen-activated protein kinase (MAPK) cascade on MeHg neurotoxicity. Decreased receptor of advanced glycation end products (RAGE) immunocontent supports the assumption that downregulated RAGE/Erk1/2 pathway could be involved in hypophosphorylated lysine/serine/proline (KSP) repeats on neurofilament subunits and disturbed axonal transport. Downregulated myelin basic protein (MBP), the major myelin protein, is compatible with dysmyelination and neurofilament hypophosphorylation. Increased glial fibrillary acidic protein (GFAP) levels suggest reactive astrocytes, and active apoptotic pathways BAD/BCL-2, BAX/BCL-XL, and caspase 3 suggest cell death. Taken together, our findings get light on important signaling mechanisms that could underlie the behavioral deficits in 30-day-old pups prenatally exposed to MeHg.
Collapse
|
29
|
Jones MR, Villalón E, Northcutt AJ, Calcutt NA, Garcia ML. Differential effects of myostatin deficiency on motor and sensory axons. Muscle Nerve 2017; 56:E100-E107. [PMID: 28073155 DOI: 10.1002/mus.25570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/04/2017] [Accepted: 01/08/2017] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Deletion of myostatin in mice (MSTN-/- ) alters structural properties of peripheral axons. However, properties like axon diameter and myelin thickness were analyzed in mixed nerves, so it is unclear whether loss of myostatin affects motor, sensory, or both types of axons. METHODS Using the MSTN-/- mouse model, we analyzed the effects of increasing the number of muscle fibers on axon diameter, myelin thickness, and internode length in motor and sensory axons. RESULTS Axon diameter and myelin thickness were increased in motor axons of MSTN-/- mice without affecting internode length or axon number. The number of sensory axons was increased without affecting their structural properties. DISCUSSION These results suggest that motor and sensory axons establish structural properties by independent mechanisms. Moreover, in motor axons, instructive cues from the neuromuscular junction may play a role in co-regulating axon diameter and myelin thickness, whereas internode length is established independently. Muscle Nerve 56: E100-E107, 2017.
Collapse
Affiliation(s)
- Maria R Jones
- Department of Biological Sciences, University of Missouri, Columbia, Missouri, USA.,C.S. Bond Life Sciences Center, University of Missouri, 1201 East Rollins Road, Columbia, Missouri, 65211, USA
| | - Eric Villalón
- Department of Biological Sciences, University of Missouri, Columbia, Missouri, USA.,C.S. Bond Life Sciences Center, University of Missouri, 1201 East Rollins Road, Columbia, Missouri, 65211, USA
| | - Adam J Northcutt
- Department of Biological Sciences, University of Missouri, Columbia, Missouri, USA
| | - Nigel A Calcutt
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Michael L Garcia
- Department of Biological Sciences, University of Missouri, Columbia, Missouri, USA.,C.S. Bond Life Sciences Center, University of Missouri, 1201 East Rollins Road, Columbia, Missouri, 65211, USA
| |
Collapse
|
30
|
Yuan A, Rao MV, Veeranna, Nixon RA. Neurofilaments and Neurofilament Proteins in Health and Disease. Cold Spring Harb Perspect Biol 2017; 9:9/4/a018309. [PMID: 28373358 DOI: 10.1101/cshperspect.a018309] [Citation(s) in RCA: 491] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SUMMARYNeurofilaments (NFs) are unique among tissue-specific classes of intermediate filaments (IFs) in being heteropolymers composed of four subunits (NF-L [neurofilament light]; NF-M [neurofilament middle]; NF-H [neurofilament heavy]; and α-internexin or peripherin), each having different domain structures and functions. Here, we review how NFs provide structural support for the highly asymmetric geometries of neurons and, especially, for the marked radial expansion of myelinated axons crucial for effective nerve conduction velocity. NFs in axons extensively cross-bridge and interconnect with other non-IF components of the cytoskeleton, including microtubules, actin filaments, and other fibrous cytoskeletal elements, to establish a regionally specialized network that undergoes exceptionally slow local turnover and serves as a docking platform to organize other organelles and proteins. We also discuss how a small pool of oligomeric and short filamentous precursors in the slow phase of axonal transport maintains this network. A complex pattern of phosphorylation and dephosphorylation events on each subunit modulates filament assembly, turnover, and organization within the axonal cytoskeleton. Multiple factors, and especially turnover rate, determine the size of the network, which can vary substantially along the axon. NF gene mutations cause several neuroaxonal disorders characterized by disrupted subunit assembly and NF aggregation. Additional NF alterations are associated with varied neuropsychiatric disorders. New evidence that subunits of NFs exist within postsynaptic terminal boutons and influence neurotransmission suggests how NF proteins might contribute to normal synaptic function and neuropsychiatric disease states.
Collapse
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016
| | - Mala V Rao
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016
| | - Veeranna
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016
| | - Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York 10962.,Department of Psychiatry, New York University School of Medicine, New York, New York 10016.,Cell Biology, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
31
|
Hoffman A, Taleski G, Sontag E. The protein serine/threonine phosphatases PP2A, PP1 and calcineurin: A triple threat in the regulation of the neuronal cytoskeleton. Mol Cell Neurosci 2017; 84:119-131. [PMID: 28126489 DOI: 10.1016/j.mcn.2017.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/16/2017] [Accepted: 01/21/2017] [Indexed: 01/08/2023] Open
Abstract
The microtubule, F-actin and neurofilament networks play a critical role in neuronal cell morphogenesis, polarity and synaptic plasticity. Significantly, the assembly/disassembly and stability of these cytoskeletal networks is crucially modulated by protein phosphorylation and dephosphorylation events. Herein, we aim to more closely examine the role played by three major neuronal Ser/Thr protein phosphatases, PP2A, PP1 and calcineurin, in the homeostasis of the neuronal cytoskeleton. There is strong evidence that these enzymes interact with and dephosphorylate a variety of cytoskeletal proteins, resulting in major regulation of neuronal cytoskeletal dynamics. Conversely, we also discuss how multi-protein cytoskeletal scaffolds can also influence the regulation of these phosphatases, with important implications for neuronal signalling and homeostasis. Not surprisingly, deregulation of these cytoskeletal scaffolds and phosphatase dysfunction are associated with many neurological diseases.
Collapse
Affiliation(s)
- Alexander Hoffman
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Goce Taleski
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, and Hunter Medical Research Institute, University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
32
|
|
33
|
Targeting caspase-6 and caspase-8 to promote neuronal survival following ischemic stroke. Cell Death Dis 2015; 6:e1967. [PMID: 26539914 PMCID: PMC4670918 DOI: 10.1038/cddis.2015.272] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/18/2015] [Accepted: 08/24/2015] [Indexed: 01/06/2023]
Abstract
Previous studies show that caspase-6 and caspase-8 are involved in neuronal apoptosis and regenerative failure after trauma of the adult central nervous system (CNS). In this study, we evaluated whether caspase-6 or -8 inhibitors can reduce cerebral or retinal injury after ischemia. Cerebral infarct volume, relative to appropriate controls, was significantly reduced in groups treated with caspase-6 or -8 inhibitors. Concomitantly, these treatments also reduced neurological deficits, reduced edema, increased cell proliferation, and increased neurofilament levels in the injured cerebrum. Caspase-6 and -8 inhibitors, or siRNAs, also increased retinal ganglion cell survival at 14 days after ischemic injury. Caspase-6 or -8 inhibition also decreased caspase-3, -6, and caspase-8 cleavage when assayed by western blot and reduced caspase-3 and -6 activities in colorimetric assays. We have shown that caspase-6 or caspase-8 inhibition decreases the neuropathological consequences of cerebral or retinal infarction, thereby emphasizing their importance in ischemic neuronal degeneration. As such, caspase-6 and -8 are potential targets for future therapies aimed at attenuating the devastating functional losses that result from retinal or cerebral stroke.
Collapse
|
34
|
Jones MR, Villalón E, Garcia ML. Genetic Manipulation of Neurofilament Protein Phosphorylation. Methods Enzymol 2015; 568:461-76. [PMID: 26795480 DOI: 10.1016/bs.mie.2015.07.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neurofilament biology is important to understanding structural properties of axons, such as establishment of axonal diameter by radial growth. In order to study the function of neurofilaments, a series of genetically modified mice have been generated. Here, we describe a brief history of genetic modifications used to study neurofilaments, as well as an overview of the steps required to generate a gene-targeted mouse. In addition, we describe steps utilized to analyze neurofilament phosphorylation status using immunoblotting. Taken together, these provide comprehensive analysis of neurofilament function in vivo, which can be applied to many systems.
Collapse
Affiliation(s)
- Maria R Jones
- Department of Biological Sciences, University of Missouri, Columbia, Missouri, USA; C.S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Eric Villalón
- Department of Biological Sciences, University of Missouri, Columbia, Missouri, USA; C.S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Michael L Garcia
- Department of Biological Sciences, University of Missouri, Columbia, Missouri, USA; C.S. Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA.
| |
Collapse
|
35
|
A Stochastic Multiscale Model That Explains the Segregation of Axonal Microtubules and Neurofilaments in Neurological Diseases. PLoS Comput Biol 2015; 11:e1004406. [PMID: 26285012 PMCID: PMC4540448 DOI: 10.1371/journal.pcbi.1004406] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/17/2015] [Indexed: 11/19/2022] Open
Abstract
The organization of the axonal cytoskeleton is a key determinant of the normal function of an axon, which is a long thin projection of a neuron. Under normal conditions two axonal cytoskeletal polymers, microtubules and neurofilaments, align longitudinally in axons and are interspersed in axonal cross-sections. However, in many neurotoxic and neurodegenerative disorders, microtubules and neurofilaments segregate apart from each other, with microtubules and membranous organelles clustered centrally and neurofilaments displaced to the periphery. This striking segregation precedes the abnormal and excessive neurofilament accumulation in these diseases, which in turn leads to focal axonal swellings. While neurofilament accumulation suggests an impairment of neurofilament transport along axons, the underlying mechanism of their segregation from microtubules remains poorly understood for over 30 years. To address this question, we developed a stochastic multiscale model for the cross-sectional distribution of microtubules and neurofilaments in axons. The model describes microtubules, neurofilaments and organelles as interacting particles in a 2D cross-section, and is built upon molecular processes that occur on a time scale of seconds or shorter. It incorporates the longitudinal transport of neurofilaments and organelles through this domain by allowing stochastic arrival and departure of these cargoes, and integrates the dynamic interactions of these cargoes with microtubules mediated by molecular motors. Simulations of the model demonstrate that organelles can pull nearby microtubules together, and in the absence of neurofilament transport, this mechanism gradually segregates microtubules from neurofilaments on a time scale of hours, similar to that observed in toxic neuropathies. This suggests that the microtubule-neurofilament segregation can be a consequence of the selective impairment of neurofilament transport. The model generates the experimentally testable prediction that the rate and extent of segregation will be dependent on the sizes of the moving organelles as well as the density of their traffic. The shape and function of axons is dependent on a dynamic system of microscopic intracellular protein polymers (microtubules, neurofilaments and microfilaments) that comprise the axonal cytoskeleton. Neurofilaments are cargoes of intracellular transport that move along microtubule tracks, and they accumulate abnormally in axons in many neurotoxic and neurodegenerative disorders. Intriguingly, it has been reported that neurofilaments and microtubules, which are normally interspersed in axonal cross-sections, often segregate apart from each other in these disorders, which is something that is never observed in healthy axons. Here we describe a stochastic multiscale computational model that explains the mechanism of this striking segregation and offers insights into the mechanism of neurofilament accumulation in disease.
Collapse
|
36
|
Pisciotta C, Bai Y, Brennan KM, Wu X, Grider T, Feely S, Wang S, Moore S, Siskind C, Gonzalez M, Zuchner S, Shy ME. Reduced neurofilament expression in cutaneous nerve fibers of patients with CMT2E. Neurology 2015; 85:228-34. [PMID: 26109717 DOI: 10.1212/wnl.0000000000001773] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/18/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the effects of NEFL Glu396Lys mutation on the expression and assembly of neurofilaments (NFs) in cutaneous nerve fibers of patients with Charcot-Marie-Tooth disease type 2E (CMT2E). METHODS A large family with CMT2E underwent clinical, electrophysiologic, and skin biopsy studies. Biopsies were processed by indirect immunofluorescence (IF), electron microscopy (EM), and Western blot analysis. RESULTS The clinical features demonstrated intrafamilial phenotypic variability, and the electrophysiologic findings revealed nerve conductions that were either slow or in the intermediate range. All patients had reduced or absent compound muscular action potential amplitudes. Skin biopsies showed axons labeled with the axonal markers protein gene product 9.5 and α-tubulin, but not with NFs. The results of Western blot analysis were consistent with those of IF, showing reduced or absent NFs and normal expression of α-tubulin. EM revealed clusters of regenerated fibers, in absence of myelin sheath abnormalities. Both IF and EM failed to show NF aggregates in dermal axons. The morphometric analysis showed a smaller axonal caliber in patients than in controls. The study of the nodal/paranodal architecture demonstrated that sodium channels and Caspr were correctly localized in patients with CMT2E. CONCLUSIONS Decrease in NF abundance may be a pathologic marker of CMT2E. The lack of NF aggregates, consistent with prior studies, suggests that they occur proximally leading to subsequent alterations in the axonal cytoskeleton. The small axonal caliber, along with the normal molecular architecture of nodes and paranodes, explain the reduced velocities detected in patients with CMT2E. Our results also demonstrate that skin biopsy can provide evidence of pathologic and pathogenic abnormalities in patients with CMT2E.
Collapse
Affiliation(s)
- Chiara Pisciotta
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL.
| | - Yunhong Bai
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Kathryn M Brennan
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Xingyao Wu
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Tiffany Grider
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Shawna Feely
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Suola Wang
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Steven Moore
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Carly Siskind
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Michael Gonzalez
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Stephan Zuchner
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| | - Michael E Shy
- From the Departments of Neurology (C.P., Y.B., K.M.B., X.W., T.G., S.F., S.W., M.E.S.) and Pathology (S.M.), University of Iowa Hospitals and Clinics, Iowa City; Departments of Neurology (C.S.), Stanford University, CA; Dr. John T. Macdonald Foundation Department of Human Genetics (M.G., S.Z.), University of Miami Miller School of Medicine, FL
| |
Collapse
|
37
|
Neuroproteomics in the auditory brainstem: Candidate proteins for ultrafast and precise information processing. Mol Cell Neurosci 2015; 64:9-23. [DOI: 10.1016/j.mcn.2014.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 07/25/2014] [Accepted: 08/12/2014] [Indexed: 12/18/2022] Open
|
38
|
McLean NA, Popescu BF, Gordon T, Zochodne DW, Verge VMK. Delayed nerve stimulation promotes axon-protective neurofilament phosphorylation, accelerates immune cell clearance and enhances remyelination in vivo in focally demyelinated nerves. PLoS One 2014; 9:e110174. [PMID: 25310564 PMCID: PMC4195712 DOI: 10.1371/journal.pone.0110174] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/17/2014] [Indexed: 01/19/2023] Open
Abstract
Rapid and efficient axon remyelination aids in restoring strong electrochemical communication with end organs and in preventing axonal degeneration often observed in demyelinating neuropathies. The signals from axons that can trigger more effective remyelination in vivo are still being elucidated. Here we report the remarkable effect of delayed brief electrical nerve stimulation (ES; 1 hour @ 20 Hz 5 days post-demyelination) on ensuing reparative events in a focally demyelinated adult rat peripheral nerve. ES impacted many parameters underlying successful remyelination. It effected increased neurofilament expression and phosphorylation, both implicated in axon protection. ES increased expression of myelin basic protein (MBP) and promoted node of Ranvier re-organization, both of which coincided with the early reappearance of remyelinated axons, effects not observed at the same time points in non-stimulated demyelinated nerves. The improved ES-associated remyelination was accompanied by enhanced clearance of ED-1 positive macrophages and attenuation of glial fibrillary acidic protein expression in accompanying Schwann cells, suggesting a more rapid clearance of myelin debris and return of Schwann cells to a nonreactive myelinating state. These benefits of ES correlated with increased levels of brain derived neurotrophic factor (BDNF) in the acute demyelination zone, a key molecule in the initiation of the myelination program. In conclusion, the tremendous impact of delayed brief nerve stimulation on enhancement of the innate capacity of a focally demyelinated nerve to successfully remyelinate identifies manipulation of this axis as a novel therapeutic target for demyelinating pathologies.
Collapse
Affiliation(s)
- Nikki A. McLean
- CMSNRC (Cameco MS Neuroscience Research Center) and Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Bogdan F. Popescu
- CMSNRC (Cameco MS Neuroscience Research Center) and Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Tessa Gordon
- Department of Surgery, Division of Plastic Reconstructive Surgery, University of Toronto, Toronto, ON, Canada
| | - Douglas W. Zochodne
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Valerie M. K. Verge
- CMSNRC (Cameco MS Neuroscience Research Center) and Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, SK, Canada
- * E-mail:
| |
Collapse
|
39
|
Abstract
Axons in the vertebrate nervous system only expand beyond ∼ 1 μm in diameter if they become myelinated. This expansion is due in large part to the accumulation of space-filling cytoskeletal polymers called neurofilaments, which are cargoes of axonal transport. One possible mechanism for this accumulation is a decrease in the rate of neurofilament transport. To test this hypothesis, we used a fluorescence photoactivation pulse-escape technique to compare the kinetics of neurofilament transport in contiguous myelinated and unmyelinated segments of axons in long-term myelinating cocultures established from the dorsal root ganglia of embryonic rats. The myelinated segments contained more neurofilaments and had a larger cross-sectional area than the contiguous unmyelinated segments, and this correlated with a local slowing of neurofilament transport. By computational modeling of the pulse-escape kinetics, we found that this slowing of neurofilament transport could be explained by an increase in the proportion of the time that the neurofilaments spent pausing and that this increase in pausing was sufficient to explain the observed neurofilament accumulation. Thus we propose that myelinating cells can regulate the neurofilament content and morphology of axons locally by modulating the kinetics of neurofilament transport.
Collapse
|
40
|
Loss of prion protein leads to age-dependent behavioral abnormalities and changes in cytoskeletal protein expression. Mol Neurobiol 2014; 50:923-36. [PMID: 24604355 DOI: 10.1007/s12035-014-8655-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/29/2014] [Indexed: 12/13/2022]
Abstract
The cellular prion protein (PrPC) is a highly conserved protein whose exact physiological role remains elusive. In the present study, we investigated age-dependent behavioral abnormalities in PrPC-knockout (Prnp0/0) mice and wild-type (WT) controls. Prnp0/0 mice showed age-dependent behavioral deficits in memory performance, associative learning, basal anxiety, and nest building behavior. Using a hypothesis-free quantitative proteomic investigation, we found that loss of PrPC affected the levels of neurofilament proteins in an age-dependent manner. In order to understand the biochemical basis of these observations, we analyzed the phosphorylation status of neurofilament heavy chain (NF-H). We found a reduction in NF-H phosphorylation in both Prnp0/0 mice and in PrPC-deficient cells. The expression of Fyn and phospho-Fyn, a potential regulator for NF phosphorylation, was associated with PrPC ablation. The number of β-tubulin III-positive neurons in the hippocampus was diminished in Prnp0/0 mice relative to WT mice. These data indicate that PrPC plays an important role in cytoskeletal organization, brain function, and age-related neuroprotection. Our work represents the first direct biochemical link between these proteins and the observed behavioral phenotypes.
Collapse
|
41
|
Bohórquez DV, Samsa LA, Roholt A, Medicetty S, Chandra R, Liddle RA. An enteroendocrine cell-enteric glia connection revealed by 3D electron microscopy. PLoS One 2014; 9:e89881. [PMID: 24587096 PMCID: PMC3935946 DOI: 10.1371/journal.pone.0089881] [Citation(s) in RCA: 181] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/28/2014] [Indexed: 11/25/2022] Open
Abstract
The enteroendocrine cell is the cornerstone of gastrointestinal chemosensation. In the intestine and colon, this cell is stimulated by nutrients, tastants that elicit the perception of flavor, and bacterial by-products; and in response, the cell secretes hormones like cholecystokinin and peptide YY – both potent regulators of appetite. The development of transgenic mice with enteroendocrine cells expressing green fluorescent protein has allowed for the elucidation of the apical nutrient sensing mechanisms of the cell. However, the basal secretory aspects of the enteroendocrine cell remain largely unexplored, particularly because a complete account of the enteroendocrine cell ultrastructure does not exist. Today, the fine ultrastructure of a specific cell can be revealed in the third dimension thanks to the invention of serial block face scanning electron microscopy (SBEM). Here, we bridged confocal microscopy with SBEM to identify the enteroendocrine cell of the mouse and study its ultrastructure in the third dimension. The results demonstrated that 73.5% of the peptide-secreting vesicles in the enteroendocrine cell are contained within an axon-like basal process. We called this process a neuropod. This neuropod contains neurofilaments, which are typical structural proteins of axons. Surprisingly, the SBEM data also demonstrated that the enteroendocrine cell neuropod is escorted by enteric glia – the cells that nurture enteric neurons. We extended these structural findings into an in vitro intestinal organoid system, in which the addition of glial derived neurotrophic factors enhanced the development of neuropods in enteroendocrine cells. These findings open a new avenue of exploration in gastrointestinal chemosensation by unveiling an unforeseen physical relationship between enteric glia and enteroendocrine cells.
Collapse
Affiliation(s)
- Diego V. Bohórquez
- Departments of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| | - Leigh A. Samsa
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Andrew Roholt
- Renovo Neural Incorporation, Cleveland, Ohio, United States of America
| | - Satish Medicetty
- Renovo Neural Incorporation, Cleveland, Ohio, United States of America
| | - Rashmi Chandra
- Departments of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Rodger A. Liddle
- Departments of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Veterans Affairs Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
42
|
Ward RE, Huang W, Kostusiak M, Pallier PN, Michael-Titus AT, Priestley JV. A characterization of white matter pathology following spinal cord compression injury in the rat. Neuroscience 2013; 260:227-39. [PMID: 24361176 DOI: 10.1016/j.neuroscience.2013.12.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 12/09/2013] [Accepted: 12/09/2013] [Indexed: 12/25/2022]
Abstract
Our laboratory has previously described the characteristics of neuronal injury in a rat compression model of spinal cord injury (SCI), focussing on the impact of this injury on the gray matter. However, white matter damage is known to play a critical role in functional outcome following injury. Therefore, in the present study, we used immunohistochemistry and electron microscopy to examine the alterations to the white matter that are initiated by compression SCI applied at T12 vertebral level. A significant loss of axonal and dendritic cytoskeletal proteins was observed at the injury epicenter within 1day of injury. This was accompanied by axonal dysfunction, as demonstrated by the accumulation of β-amyloid precursor protein (β-APP), with a peak at 3days post-SCI. A similar, acute loss of cytoskeletal proteins was observed up to 5mm away from the injury epicenter and was particularly evident rostral to the lesion site, whereas β-APP accumulation was prominent in tracts proximal to the injury. Early myelin loss was confirmed by myelin basic protein (MBP) immunostaining and by electron microscopy, which also highlighted the infiltration of inflammatory and red blood cells. However, 6weeks after injury, areas of new Schwann cell and oligodendrocyte myelination were observed. This study demonstrates that substantial white matter damage occurs following compression SCI in the rat. Moreover, the loss of cytoskeletal proteins and accumulation of β-APP up to 5mm away from the lesion site within 1day of injury indicates the rapid manner in which the axonal damage extends in the rostro-caudal axis. This is likely due to both Wallerian degeneration and spread of secondary cell death, with the latter affecting axons both proximal and distal to the injury.
Collapse
Affiliation(s)
- R E Ward
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| | - W Huang
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; The Institute of Medical Sciences, School of Medical Science, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - M Kostusiak
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - P N Pallier
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - A T Michael-Titus
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - J V Priestley
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
43
|
Huang Z, Zhuo Y, Shen Z, Wang Y, Wang L, Li H, Chen J, Chen W. The role of NEFL in cell growth and invasion in head and neck squamous cell carcinoma cell lines. J Oral Pathol Med 2013; 43:191-8. [PMID: 23992471 DOI: 10.1111/jop.12109] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2013] [Indexed: 02/01/2023]
Abstract
The neurofilament light polypeptide (NEFL) gene located on chromosome 8q21 is associated with the cancer of several organs and is regarded as a potential tumor suppressor gene. However, the role of the NEFL protein has not yet been studied in cancer cells. Although evidence suggests that there is a correlation between NEFL expression and cancer, studies regarding the role of the NEFL protein have been mostly limited to neurological diseases, such as Charcot-Marie-Tooth's disease (CMT). Most of these studies have not explored the role of NEFL in cancer cell apoptosis and/or invasion. In this study, NEFL expression was manipulated, and apoptosis and invasion were compared in head and neck squamous cell carcinoma cell lines. The results show that the expression of NEFL induces cancer cell apoptosis and inhibits invasion in these cell lines, suggesting that NEFL may play a role in cancer cell apoptosis and invasion.
Collapse
Affiliation(s)
- Zhiquan Huang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Key Laboratory of malignant tumor gene regulation and target therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, Guangdong, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Aidong Yuan
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, NY 10962, USA.
| | | | | | | |
Collapse
|
45
|
|
46
|
Rao MV, Yuan A, Campbell J, Kumar A, Nixon RA. The C-terminal domains of NF-H and NF-M subunits maintain axonal neurofilament content by blocking turnover of the stationary neurofilament network. PLoS One 2012; 7:e44320. [PMID: 23028520 PMCID: PMC3448626 DOI: 10.1371/journal.pone.0044320] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/01/2012] [Indexed: 02/03/2023] Open
Abstract
Newly synthesized neurofilaments or protofilaments are incorporated into a highly stable stationary cytoskeleton network as they are transported along axons. Although the heavily phosphorylated carboxyl-terminal tail domains of the heavy and medium neurofilament (NF) subunits have been proposed to contribute to this process and particularly to stability of this structure, their function is still obscure. Here we show in NF-H/M tail deletion [NF-(H/M)(tailΔ)] mice that the deletion of both of these domains selectively lowers NF levels 3-6 fold along optic axons without altering either rates of subunit synthesis or the rate of slow axonal transport of NF. Pulse labeling studies carried out over 90 days revealed a significantly faster rate of disappearance of NF from the stationary NF network of optic axons in NF-(H/M)(tailΔ) mice. Faster NF disappearance was accompanied by elevated levels of NF-L proteolytic fragments in NF-(H/M)(tailΔ) axons. We conclude that NF-H and NF-M C-terminal domains do not normally regulate NF transport rates as previously proposed, but instead increase the proteolytic resistance of NF, thereby stabilizing the stationary neurofilament cytoskeleton along axons.
Collapse
Affiliation(s)
- Mala V Rao
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York, United States of America.
| | | | | | | | | |
Collapse
|
47
|
Song F, Zhang Q, Kou R, Zou C, Gao Y, Xie K. 2,5-hexanedione altered the degradation of low-molecular-weight neurofilament in rat nerve tissues. Food Chem Toxicol 2012; 50:4277-84. [PMID: 22967723 DOI: 10.1016/j.fct.2012.08.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 08/07/2012] [Accepted: 08/23/2012] [Indexed: 02/03/2023]
Abstract
Occupational exposure to n-hexane produces a central-peripheral distal axonopathy, which is characterized by giant axonal swellings filled with neurofilaments (NFs). To investigate the change of NFs degradation and their possible role in n-hexane neuropathy, adult male Wistar rats were administered intraperitoneally at a dosage of 400 mg/kg/day 2,5-hexanedione (2,5-HD) for 4 weeks. The time course of low-molecular-weight neurofilament (NF-L) degradation and autophagy-related protein in rat sciatic nerves and spinal cords was determined by Western blotting. The results demonstrated that the administration of 2,5-HD inhibited NF-L degradation to an undetectable level in sciatic nerves. Furthermore, a significant reduction of NF-L degradation in spinal cords was observed in the early stage of 2,5-HD exposure. In the meantime, 2,5-HD significantly decreased the level of Beclin-1, a key autophagy-regulated protein in sciatic nerves of rats while increased the level of P62, a selective substrate of autophagy degrading pathway, which indicated a dysfunctional autophagy in rat nerve tissues. Collectively, our findings suggested that the inhibition of autophagy by 2,5-HD might be responsible for the reduction of NF-L degradation in rat sciatic nerves, and involved in the pathogenesis of 2,5-HD-induced axonopathy.
Collapse
Affiliation(s)
- Fuyong Song
- Institute of Toxicology, School of Public Health, Shandong University, Jinan, Shandong, 250012, PR China
| | | | | | | | | | | |
Collapse
|
48
|
Wakeling EN, Joussemet B, Costiou P, Fanuel D, Moullier P, Barkats M, Fyfe JC. Failure of lower motor neuron radial outgrowth precedes retrograde degeneration in a feline model of spinal muscular atrophy. J Comp Neurol 2012; 520:1737-50. [PMID: 22120001 DOI: 10.1002/cne.23010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Feline spinal muscular atrophy (SMA) is a fully penetrant, autosomal recessive lower motor neuron disease in domestic cats that clinically resembles human SMA Type III. A whole genome linkage scan identified a ∼140-kb deletion that abrogates expression of LIX1, a novel SMA candidate gene of unknown function. To characterize the progression of feline SMA, we assessed pathological changes in muscle and spinal cord from 3 days of age to beyond onset of clinical signs. Electromyographic (EMG) analysis indicating denervation occurred between 10 and 12 weeks, with the first neurological signs occurring at the same time. Compound motor action potential (CMAP) amplitudes were significantly reduced in the soleus and extensor carpi radialis muscles at 8-11 weeks. Quadriceps femoris muscle fibers from affected cats appeared smaller at 10 weeks; by 12 weeks atrophic fibers were more prevalent than in age-matched controls. In affected cats, significant loss of L5 ventral root axons was observed at 12 weeks. By 21 weeks of age, affected cats had 40% fewer L5 motor axons than normal. There was no significant difference in total L5 soma number, even at 21 weeks; thus degeneration begins distal to the cell body and proceeds retrogradely. Morphometric analysis of L5 ventral roots and horns revealed that 4 weeks prior to axon loss, motor axons in affected cats failed to undergo radial enlargement, suggesting a role for the putative disease gene LIX1 in radial growth of axons.
Collapse
Affiliation(s)
- Erin N Wakeling
- Genetics Program, Michigan State University East Lansing, Michigan 48824, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Serum biomarkers of neurologic injury in cardiac operations. Ann Thorac Surg 2012; 94:1026-33. [PMID: 22857981 DOI: 10.1016/j.athoracsur.2012.04.142] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 04/19/2012] [Accepted: 04/23/2012] [Indexed: 01/03/2023]
Abstract
Assessment of subtle neurocognitive decline after surgical procedures has been hampered by heterogeneous testing techniques and a lack of reproducibility. This review summarizes the sensitivity and specificity of biomarkers of neurologic injury to determine whether they can be applied in the postoperative period to accurately predict neurocognitive decline. Creatine kinase-brain type, neuron-specific enolase, and S100B can be released into serum during operations by extracranial sources. Glial fibrillary acidic protein is a sensitive marker, and there are extracranial sources that are antigenically different from the brain-derived form. Serum levels of tau protein after acute neurologic injury do not reliability correlate with incidence.
Collapse
|
50
|
Duffy KR, Crowder NA, LeDue EE. Investigation of cytoskeleton proteins in neurons of the cat lateral geniculate nucleus. J Comp Neurol 2011; 520:186-99. [DOI: 10.1002/cne.22727] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|