1
|
Zou Y, Zhang X, Chen XY, Ma XF, Feng XY, Sun Y, Ma T, Ma QH, Zhao XD, Xu DE. Contactin -Associated protein1 Regulates Autophagy by Modulating the PI3K/AKT/mTOR Signaling Pathway and ATG4B Levels in Vitro and in Vivo. Mol Neurobiol 2024:10.1007/s12035-024-04425-9. [PMID: 39164481 DOI: 10.1007/s12035-024-04425-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/06/2024] [Indexed: 08/22/2024]
Abstract
Contactin-associated protein1 (Caspr1) plays an important role in the formation and stability of myelinated axons. In Caspr1 mutant mice, autophagy-related structures accumulate in neurons, causing axonal degeneration; however, the mechanism by which Caspr1 regulates autophagy remains unknown. To illustrate the mechanism of Caspr1 in autophagy process, we demonstrated that Caspr1 knockout in primary neurons from mice along with human cell lines, HEK-293 and HeLa, induced autophagy by downregulating the PI3K/AKT/mTOR signaling pathway to promote the conversion of microtubule-associated protein light chain 3 I (LC3-I) to LC3-II. In contrast, Caspr1 overexpression in cells contributed to the upregulation of this signaling pathway. We also demonstrated that Caspr1 knockout led to increased LC3-I protein expression in mice. In addition, Caspr1 could inhibit the expression of autophagy-related 4B cysteine peptidase (ATG4B) protein by directly binding to ATG4B in overexpressed Caspr1 cells. Intriguingly, we found an accumulation of ATG4B in the Golgi apparatuses of cells overexpressing Caspr1; therefore, we speculate that Caspr1 may restrict ATG4 secretion from the Golgi apparatus to the cytoplasm. Collectively, our results indicate that Caspr1 may regulate autophagy by modulating the PI3K/AKT/mTOR signaling pathway and the levels of ATG4 protein, both in vitro and in vivo. Thus, Caspr1 can be a potential therapeutic target in axonal damage and demyelinating diseases.
Collapse
Affiliation(s)
- Yan Zou
- Department of Neurosurgery, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xiao Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xin-Yi Chen
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Xiao-Fang Ma
- Hong Shan Hospital, Wuxi, 214000, Jiangsu, China
| | - Xiao-Yan Feng
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Yang Sun
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Tao Ma
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215004, Jiangsu, China
| | - Xu-Dong Zhao
- Department of Neurosurgery, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China.
- Wuxi Neurosurgical Institute, Wuxi, 214122, Jiangsu, China.
| | - De-En Xu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214002, Jiangsu, China.
- Department of Neurology, Jiangnan University Medical Center, the Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China.
| |
Collapse
|
2
|
Ding X, Wu Y, Rodriguez V, Ricco E, Okoh JT, Liu Y, Kraushaar DC, Rasband MN. Age-dependent regulation of axoglial interactions and behavior by oligodendrocyte AnkyrinG. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587609. [PMID: 38617359 PMCID: PMC11014615 DOI: 10.1101/2024.04.01.587609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The bipolar disorder (BD) risk gene ANK3 encodes the scaffolding protein AnkyrinG (AnkG). In neurons, AnkG regulates polarity and ion channel clustering at axon initial segments and nodes of Ranvier. Disruption of neuronal AnkG causes BD-like phenotypes in mice. During development, AnkG is also expressed at comparable levels in oligodendrocytes and facilitates the efficient assembly of paranodal junctions. However, the physiological roles of glial AnkG in the mature nervous system, and its contributions to BD-like phenotypes, remain unexplored. Here, we generated oligodendroglia-specific AnkG conditional knockout mice and observed the destabilization of axoglial interactions in aged but not young adult mice. In addition, these mice exhibited profound histological, electrophysiological, and behavioral pathophysiologies. Unbiased translatomic profiling revealed potential compensatory machineries. These results highlight the critical functions of glial AnkG in maintaining proper axoglial interactions throughout aging and suggests a previously unrecognized contribution of oligodendroglial AnkG to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Victoria Rodriguez
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Emily Ricco
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - James T. Okoh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yanhong Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Daniel C. Kraushaar
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
3
|
Chang C, Sell LB, Shi Q, Bhat MA. Mouse models of human CNTNAP1-associated congenital hypomyelinating neuropathy and genetic restoration of murine neurological deficits. Cell Rep 2023; 42:113274. [PMID: 37862170 PMCID: PMC10873044 DOI: 10.1016/j.celrep.2023.113274] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/22/2023] Open
Abstract
The Contactin-associated protein 1 (Cntnap1) mouse mutants fail to establish proper axonal domains in myelinated axons. Human CNTNAP1 mutations are linked to hypomyelinating neuropathy-3, which causes severe neurological deficits. To understand the human neuropathology and to model human CNTNAP1C323R and CNTNAP1R764C mutations, we generated Cntnap1C324R and Cntnap1R765C mouse mutants, respectively. Both Cntnap1 mutants show weight loss, reduced nerve conduction, and progressive motor dysfunction. The paranodal ultrastructure shows everted myelin loops and the absence of axo-glial junctions. Biochemical analysis reveals that these Cntnap1 mutant proteins are nearly undetectable in the paranodes, have reduced surface expression and stability, and are retained in the neuronal soma. Postnatal transgenic expression of Cntnap1 in the mutant backgrounds rescues the phenotypes and restores the organization of axonal domains with improved motor function. This study uncovers the mechanistic impact of two human CNTNAP1 mutations in a mouse model and provides proof of concept for gene therapy for CNTNAP1 patients.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Lacey B Sell
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; IBMS Neuroscience Graduate Program, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Qian Shi
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; IBMS Neuroscience Graduate Program, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; IBMS Neuroscience Graduate Program, Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
4
|
Shackleford G, Marziali LN, Sasaki Y, Claessens A, Ferri C, Weinstock NI, Rossor AM, Silvestri NJ, Wilson ER, Hurley E, Kidd GJ, Manohar S, Ding D, Salvi RJ, Feltri ML, D’Antonio M, Wrabetz L. A new mouse model of Charcot-Marie-Tooth 2J neuropathy replicates human axonopathy and suggest alteration in axo-glia communication. PLoS Genet 2022; 18:e1010477. [PMID: 36350884 PMCID: PMC9707796 DOI: 10.1371/journal.pgen.1010477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/29/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022] Open
Abstract
Myelin is essential for rapid nerve impulse propagation and axon protection. Accordingly, defects in myelination or myelin maintenance lead to secondary axonal damage and subsequent degeneration. Studies utilizing genetic (CNPase-, MAG-, and PLP-null mice) and naturally occurring neuropathy models suggest that myelinating glia also support axons independently from myelin. Myelin protein zero (MPZ or P0), which is expressed only by Schwann cells, is critical for myelin formation and maintenance in the peripheral nervous system. Many mutations in MPZ are associated with demyelinating neuropathies (Charcot-Marie-Tooth disease type 1B [CMT1B]). Surprisingly, the substitution of threonine by methionine at position 124 of P0 (P0T124M) causes axonal neuropathy (CMT2J) with little to no myelin damage. This disease provides an excellent paradigm to understand how myelinating glia support axons independently from myelin. To study this, we generated targeted knock-in MpzT124M mutant mice, a genetically authentic model of T124M-CMT2J neuropathy. Similar to patients, these mice develop axonopathy between 2 and 12 months of age, characterized by impaired motor performance, normal nerve conduction velocities but reduced compound motor action potential amplitudes, and axonal damage with only minor compact myelin modifications. Mechanistically, we detected metabolic changes that could lead to axonal degeneration, and prominent alterations in non-compact myelin domains such as paranodes, Schmidt-Lanterman incisures, and gap junctions, implicated in Schwann cell-axon communication and axonal metabolic support. Finally, we document perturbed mitochondrial size and distribution along MpzT124M axons suggesting altered axonal transport. Our data suggest that Schwann cells in P0T124M mutant mice cannot provide axons with sufficient trophic support, leading to reduced ATP biosynthesis and axonopathy. In conclusion, the MpzT124M mouse model faithfully reproduces the human neuropathy and represents a unique tool for identifying the molecular basis for glial support of axons.
Collapse
Affiliation(s)
- Ghjuvan’Ghjacumu Shackleford
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Leandro N. Marziali
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Yo Sasaki
- Needleman Center for Neurometabolism and Axonal Therapeutics and Department of Genetics, Washington University School of Medicine in Saint Louis, St. Louis, Missouri, United States of America
| | - Anke Claessens
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Ferri
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Nadav I. Weinstock
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Alexander M. Rossor
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Nicholas J. Silvestri
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Emma R. Wilson
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Edward Hurley
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Grahame J. Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Senthilvelan Manohar
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Dalian Ding
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Richard J. Salvi
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - M. Laura Feltri
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| | - Maurizio D’Antonio
- Biology of Myelin Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Lawrence Wrabetz
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Department Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, New York, United States of America
| |
Collapse
|
5
|
Ishibashi T, Baba H. Paranodal Axoglial Junctions, an Essential Component in Axonal Homeostasis. Front Cell Dev Biol 2022; 10:951809. [PMID: 35874818 PMCID: PMC9299063 DOI: 10.3389/fcell.2022.951809] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022] Open
Abstract
In vertebrates, a high density of voltage-gated Na+ channel at nodes of Ranvier and of voltage-gated K+ channel at juxtaparanodes is necessary for rapid propagation of action potential, that is, for saltatory conduction in myelinated axons. Myelin loops attach to the axonal membrane and form paranodal axoglial junctions (PNJs) at paranodes adjacent to nodes of Ranvier. There is growing evidence that the PNJs contribute to axonal homeostasis in addition to their roles as lateral fences that restrict the location of nodal axolemmal proteins for effective saltatory conduction. Perturbations of PNJs, as in specific PNJ protein knockouts as well as in myelin lipid deficient mice, result in internodal axonal alterations, even if their internodal myelin is preserved. Here we review studies showing that PNJs play crucial roles in the myelinated axonal homeostasis. The present evidence points to two functions in particular: 1) PNJs facilitate axonal transport of membranous organelles as well as cytoskeletal proteins; and 2) they regulate the axonal distribution of type 1 inositol 1,4,5-trisphosphate receptor (IP3R1) in cerebellar Purkinje axons. Myelinated axonal homeostasis depends among others on the state of PNJs, and consequently, a better understanding of this dependency may contribute to the clarification of CNS disease mechanisms and the development of novel therapies.
Collapse
Affiliation(s)
- Tomoko Ishibashi
- Department of Functional Neurobiology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Japan
| | - Hiroko Baba
- Department of Occupational Therapy, Faculty of Rehabilitation, Niigata University of Health and Welfare, Niigata, Japan
| |
Collapse
|
6
|
Correale J, Ysrraelit MC. Multiple Sclerosis and Aging: The Dynamics of Demyelination and Remyelination. ASN Neuro 2022; 14:17590914221118502. [PMID: 35938615 PMCID: PMC9364177 DOI: 10.1177/17590914221118502] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system
(CNS) leading to demyelination and neurodegeneration. Life expectancy and age of onset in
MS patients have been rising over the last decades, and previous studies have shown that
age affects disease progression. Therefore, age appears as one of the most important
factors in accumulating disability in MS patients. Indeed, the degeneration of
oligodendrocytes (OGDs) and OGD precursors (OPCs) increases with age, in association with
increased inflammatory activity of astrocytes and microglia. Similarly, age-related
neuronal changes such as mitochondrial alterations, an increase in oxidative stress, and
disrupted paranodal junctions can impact myelin integrity. Conversely, once myelination is
complete, the long-term integrity of axons depends on OGD supply of energy. These
alterations determine pathological myelin changes consisting of myelin outfolding,
splitting, and accumulation of multilamellar fragments. Overall, these data demonstrate
that old mature OGDs lose their ability to produce and maintain healthy myelin over time,
to induce de novo myelination, and to remodel pre-existing myelinated
axons that contribute to neural plasticity in the CNS. Furthermore, as observed in other
tissues, aging induces a general decline in regenerative processes and, not surprisingly,
progressively hinders remyelination in MS. In this context, this review will provide an
overview of the current knowledge of age-related changes occurring in cells of the
oligodendroglial lineage and how they impact myelin synthesis, axonal degeneration, and
remyelination efficiency.
Collapse
Affiliation(s)
- Jorge Correale
- Departamento de Neurología, 58782Fleni, Buenos Aires, Argentina
| | | |
Collapse
|
7
|
Tang M, Liu T, Jiang P, Dang R. The interaction between autophagy and neuroinflammation in major depressive disorder: From pathophysiology to therapeutic implications. Pharmacol Res 2021; 168:105586. [PMID: 33812005 DOI: 10.1016/j.phrs.2021.105586] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022]
Abstract
The past decade has revealed neuroinflammation as an important mechanism of major depressive disorder (MDD). Nod-like receptors family pyrin domain containing 3 (NLRP3) inflammasome is the key regulator interleukin-1β (IL-1β) maturation, whose activation has been reported in MDD patients and various animal models. Function as a dominant driver of neuroinflammation, NLRP3 bridges the gap between immune activation with stress exposure, and further leads to subsequent occurrence of neuropsychiatric disorders such as MDD. Of note, autophagy is a tightly regulated cellular degradation pathway that removes damaged organelles and intracellular pathogens, and maintains cellular homeostasis from varying insults. Serving as a critical cellular monitoring system, normal functioned autophagy signaling prevents excessive NLRP3 inflammasome activation and subsequent release of IL-1 family cytokines. This review will describe the current understanding of how autophagy regulates NLRP3 inflammasome activity and discuss the implications of this regulation on the pathogenesis of MDD. The extensive crosstalk between autophagy pathway and NLRP3 inflammasome is further discussed, as it is critical for developing new therapeutic strategies for MDD aimed at modulating the neuroinflammatory responses.
Collapse
Affiliation(s)
- Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ting Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pei Jiang
- Institute of Clinical Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272000, China.
| | - Ruili Dang
- Institute of Clinical Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272000, China.
| |
Collapse
|
8
|
Sakita M, Murakami S, Nonaka K, Sakamoto R, Saito T, Isobe W, Kumagai S. Different patterns in age-related morphometric alteration of myelinated fibers and capillaries of the tibial nerve: a longitudinal study in normal rats. J Anat 2020; 236:1101-1111. [PMID: 32052433 DOI: 10.1111/joa.13168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
Age-related regression of myelinated fibers in peripheral nerves of the lower limbs is strongly influenced by capillaries and results in balance dysfunction and falls. However, the temporal relationships between alteration patterns of myelinated fibers and capillaries have not yet been clarified. This study aimed to investigate age-related morphological and histological changes of both myelinated fibers and capillaries in peripheral nerves to clarify whether myelinated fibers or capillaries change earlier. Seven male Wistar rats each were randomly selected at 20 weeks (young group), 70 weeks (middle group), and 97 weeks (old group) for histological evaluations. The left and right tibial nerves were removed morphologically and histologically to examine myelinated fibers and capillaries. Axon diameter and myelin thickness were almost unaltered in the middle group compared with the young group but were significantly reduced in the old group when compared with the other two groups. However, the capillary diameter and number of microvascular branch points were substantially reduced in the middle group. The current study demonstrates that myelinated fibers of peripheral nerves show signs of regression in elderly rats, whereas capillaries start to reduce in middle-aged animals. In normal aging of the tibial nerve, capillaries may regress before myelinated fibers.
Collapse
Affiliation(s)
- Masahiro Sakita
- Graduate School of Health Sciences, Kyoto Tachibana University, Kyoto, Japan
| | - Shinichiro Murakami
- Department of Physical Therapy, Faculty of Health Care Sciences, Himeji-Dokkyo University, Hyogo, Japan
| | - Koji Nonaka
- Department of Rehabilitation, Faculty of Health Sciences, Naragakuen University, Nara, Japan
| | - Ryuji Sakamoto
- Department of Physical Therapy, Takarazuka University of Medical and Health Care, Hyogo, Japan
| | - Takafumi Saito
- Department of Physical Therapy, Aso Rehabilitation College, Fukuoka, Japan
| | - Wataru Isobe
- Department of Rehabilitation, Mitsubishi Kyoto Hospital, Kyoto, Japan
| | - Shuzo Kumagai
- Laboratory of Health and Exercise Epidemiology, Center for Health Science and Counseling, Kyushu University, Fukuoka, Japan
| |
Collapse
|
9
|
Treatment resistance in major depression is correlated with increased plasma levels of neurofilament light protein reflecting axonal damage. Med Hypotheses 2019; 127:159-161. [DOI: 10.1016/j.mehy.2019.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 02/19/2019] [Accepted: 03/21/2019] [Indexed: 12/28/2022]
|
10
|
Yermakov LM, Hong LA, Drouet DE, Griggs RB, Susuki K. Functional Domains in Myelinated Axons. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:65-83. [PMID: 31760639 DOI: 10.1007/978-981-32-9636-7_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Propagation of action potentials along axons is optimized through interactions between neurons and myelinating glial cells. Myelination drives division of the axons into distinct molecular domains including nodes of Ranvier. The high density of voltage-gated sodium channels at nodes generates action potentials allowing for rapid and efficient saltatory nerve conduction. At paranodes flanking both sides of the nodes, myelinating glial cells interact with axons, forming junctions that are essential for node formation and maintenance. Recent studies indicate that the disruption of these specialized axonal domains is involved in the pathophysiology of various neurological diseases. Loss of paranodal axoglial junctions due to genetic mutations or autoimmune attack against the paranodal proteins leads to nerve conduction failure and neurological symptoms. Breakdown of nodal and paranodal proteins by calpains, the calcium-dependent cysteine proteases, may be a common mechanism involved in various nervous system diseases and injuries. This chapter reviews recent progress in neurobiology and pathophysiology of specialized axonal domains along myelinated nerve fibers.
Collapse
Affiliation(s)
- Leonid M Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Lulu A Hong
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Domenica E Drouet
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Ryan B Griggs
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA.
| |
Collapse
|
11
|
Baba H, Ishibashi T. The Role of Sulfatides in Axon–Glia Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:165-179. [DOI: 10.1007/978-981-32-9636-7_11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
12
|
Mariano V, Domínguez-Iturza N, Neukomm LJ, Bagni C. Maintenance mechanisms of circuit-integrated axons. Curr Opin Neurobiol 2018; 53:162-173. [PMID: 30241058 DOI: 10.1016/j.conb.2018.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022]
Abstract
Adult, circuit-integrated neurons must be maintained and supported for the life span of their host. The attenuation of either maintenance or plasticity leads to impaired circuit function and ultimately to neurodegenerative disorders. Over the last few years, significant discoveries of molecular mechanisms were made that mediate the formation and maintenance of axons. Here, we highlight intrinsic and extrinsic mechanisms that ensure the health and survival of axons. We also briefly discuss examples of mutations associated with impaired axonal maintenance identified in specific neurological conditions. A better understanding of these mechanisms will therefore help to define targets for therapeutic interventions.
Collapse
Affiliation(s)
- Vittoria Mariano
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland; Department of Neurosciences KU Leuven, VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Nuria Domínguez-Iturza
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland; Department of Neurosciences KU Leuven, VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Lukas J Neukomm
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland.
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Switzerland; Department of Biomedicine and Prevention, University of Rome Tor Vergata, Italy.
| |
Collapse
|
13
|
Conant A, Curiel J, Pizzino A, Sabetrasekh P, Murphy J, Bloom M, Evans SH, Helman G, Taft RJ, Simons C, Whitehead MT, Moore SA, Vanderver A. Absence of Axoglial Paranodal Junctions in a Child With CNTNAP1 Mutations, Hypomyelination, and Arthrogryposis. J Child Neurol 2018; 33:642-650. [PMID: 29882456 PMCID: PMC6800098 DOI: 10.1177/0883073818776157] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Leukodystrophies and genetic leukoencephalopathies are a heterogeneous group of heritable disorders that affect the glial-axonal unit. As more patients with unsolved leukodystrophies and genetic leukoencephalopathies undergo next generation sequencing, causative mutations in genes leading to central hypomyelination are being identified. Two such individuals presented with arthrogryposis multiplex congenita, congenital hypomyelinating neuropathy, and central hypomyelination with early respiratory failure. Whole exome sequencing identified biallelic mutations in the CNTNAP1 gene: homozygous c.1163G>C (p.Arg388Pro) and compound heterozygous c.967T>C (p.Cys323Arg) and c.319C>T (p.Arg107*). Sural nerve and quadriceps muscle biopsies demonstrated progressive, severe onion bulb and axonal pathology. By ultrastructural evaluation, septate axoglial paranodal junctions were absent from nodes of Ranvier. Serial brain magnetic resonance images revealed hypomyelination, progressive atrophy, and reduced diffusion in the globus pallidus in both patients. These 2 families illustrate severe progressive peripheral demyelinating neuropathy due to the absence of septate paranodal junctions and central hypomyelination with neurodegeneration in CNTNAP1-associated arthrogryposis multiplex congenita.
Collapse
Affiliation(s)
- Alexander Conant
- 1 Department of Neurology, Children's National Health System, Washington, DC, USA
| | - Julian Curiel
- 2 Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amy Pizzino
- 1 Department of Neurology, Children's National Health System, Washington, DC, USA
| | - Parisa Sabetrasekh
- 1 Department of Neurology, Children's National Health System, Washington, DC, USA
| | - Jennifer Murphy
- 3 National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Miriam Bloom
- 4 Department of Pediatric Hospitalist Medicine, Children's National Health System, Washington, DC, USA
| | - Sarah H Evans
- 5 Department of Physical Medicine and Rehabilitation, Children's National Health System, Washington, DC, USA
| | - Guy Helman
- 1 Department of Neurology, Children's National Health System, Washington, DC, USA.,6 Center for Genetic Medicine, Children's National Health System, Washington DC, USA.,7 Murdoch Children's Research Institute, Parkville, Melbourne, Australia
| | - Ryan J Taft
- 8 Illumina, San Diego, CA, USA.,9 Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia
| | - Cas Simons
- 7 Murdoch Children's Research Institute, Parkville, Melbourne, Australia.,9 Institute for Molecular Bioscience, University of Queensland, St. Lucia, Queensland, Australia
| | - Matthew T Whitehead
- 10 Neuroradiology Department, Children's National Health System, Washington, DC, USA.,11 George Washington University School of Medicine, Washington, DC, USA
| | - Steven A Moore
- 12 Department of Pathology, University of Iowa Carver College of Medicine and Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Iowa City, IA, USA
| | - Adeline Vanderver
- 1 Department of Neurology, Children's National Health System, Washington, DC, USA.,2 Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.,3 National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.,11 George Washington University School of Medicine, Washington, DC, USA
| |
Collapse
|
14
|
Experimental Traumatic Brain Injury Identifies Distinct Early and Late Phase Axonal Conduction Deficits of White Matter Pathophysiology, and Reveals Intervening Recovery. J Neurosci 2018; 38:8723-8736. [PMID: 30143572 PMCID: PMC6181309 DOI: 10.1523/jneurosci.0819-18.2018] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/15/2018] [Accepted: 07/10/2018] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injury (TBI) patients often exhibit slowed information processing speed that can underlie diverse symptoms. Processing speed depends on neural circuit function at synapses, in the soma, and along axons. Long axons in white matter (WM) tracts are particularly vulnerable to TBI. We hypothesized that disrupted axon–myelin interactions that slow or block action potential conduction in WM tracts may contribute to slowed processing speed after TBI. Concussive TBI in male/female mice was used to produce traumatic axonal injury in the corpus callosum (CC), similar to WM pathology in human TBI cases. Compound action potential velocity was slowed along myelinated axons at 3 d after TBI with partial recovery by 2 weeks, suggesting early demyelination followed by remyelination. Ultrastructurally, dispersed demyelinated axons and disorganized myelin attachment to axons at paranodes were apparent within CC regions exhibiting traumatic axonal injury. Action potential conduction is exquisitely sensitive to paranode abnormalities. Molecular identification of paranodes and nodes of Ranvier detected asymmetrical paranode pairs and abnormal heminodes after TBI. Fluorescent labeling of oligodendrocyte progenitors in NG2CreER;mTmG mice showed increased synthesis of new membranes extended along axons to paranodes, indicating remyelination after TBI. At later times after TBI, an overall loss of conducting axons was observed at 6 weeks followed by CC atrophy at 8 weeks. These studies identify a progression of both myelinated axon conduction deficits and axon–myelin pathology in the CC, implicating WM injury in impaired information processing at early and late phases after TBI. Furthermore, the intervening recovery reveals a potential therapeutic window. SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) is a major global health concern. Across the spectrum of TBI severities, impaired information processing can contribute to diverse functional deficits that underlie persistent symptoms. We used experimental TBI to exploit technical advantages in mice while modeling traumatic axonal injury in white matter tracts, which is a key pathological feature of human TBI. A combination of approaches revealed slowed and failed signal conduction along with damage to the structure and molecular composition of myelinated axons in the white matter after TBI. An early regenerative response was not sustained yet reveals a potential time window for intervention. These insights into white matter abnormalities underlying axon conduction deficits can inform strategies to improve treatment options for TBI patients.
Collapse
|
15
|
Wang Y, Song M, Song F. Neuronal autophagy and axon degeneration. Cell Mol Life Sci 2018; 75:2389-2406. [PMID: 29675785 PMCID: PMC11105516 DOI: 10.1007/s00018-018-2812-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/13/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023]
Abstract
Axon degeneration is a pathophysiological process of axonal dying and breakdown, which is characterized by several morphological features including the accumulation of axoplasmic organelles, disassembly of microtubules, and fragmentation of the axonal cytoskeleton. Autophagy, a highly conserved lysosomal-degradation machinery responsible for the control of cellular protein quality, is widely believed to be essential for the maintenance of axonal homeostasis in neurons. In recent years, more and more evidence suggests that dysfunctional autophagy is associated with axonal degeneration in many neurodegenerative diseases. Here, we review the core machinery of autophagy in neuronal cells, and provide several major steps that interfere with autophagy flux in neurodegenerative conditions. Furthermore, this review highlights the potential role of neuronal autophagy in axon degeneration, and presents some possible molecular mechanisms by which dysfunctional autophagy leads to axon degeneration in pathological conditions.
Collapse
Affiliation(s)
- Yu Wang
- Department of Toxicology, School of Public Health, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Mingxue Song
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fuzhou, 350108, Fujian, People's Republic of China
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
16
|
Krause Neto W, Silva WDA, Ciena AP, de Souza RR, Anaruma CA, Gama EF. Aging Induces Changes in the Somatic Nerve and Postsynaptic Component without Any Alterations in Skeletal Muscles Morphology and Capacity to Carry Load of Wistar Rats. Front Neurosci 2017; 11:688. [PMID: 29326543 PMCID: PMC5741656 DOI: 10.3389/fnins.2017.00688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/22/2017] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to analyze the morphology of the peripheral nerve, postsynaptic compartment, skeletal muscles and weight-bearing capacity of Wistar rats at specific ages. Twenty rats were divided into groups: 10 months-old (ADULT) and 24 months-old (OLD). After euthanasia, we prepared and analyzed the tibial nerve using transmission electron microscopy and the soleus and plantaris muscles for cytofluorescence and histochemistry. For the comparison of the results between groups we used dependent and independent Student's t-test with level of significance set at p ≤ 0.05. For the tibial nerve, the OLD group presented the following alterations compared to the ADULT group: larger area and diameter of both myelinated fibers and axons, smaller area occupied by myelinated and unmyelinated axons, lower numerical density of myelinated fibers, and fewer myelinated fibers with normal morphology. Both aged soleus and plantaris end-plate showed greater total perimeter, stained perimeter, total area and stained area compared to ADULT group (p < 0.05). Yet, aged soleus end-plate presented greater dispersion than ADULT samples (p < 0.05). For the morphology of soleus and plantaris muscles, density of the interstitial volume was greater in the OLD group (p < 0.05). No statistical difference was found between groups in the weight-bearing tests. The results of the present study demonstrated that the aging process induces changes in the peripheral nerve and postsynaptic compartment without any change in skeletal muscles and ability to carry load in Wistar rats.
Collapse
Affiliation(s)
- Walter Krause Neto
- Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, Brazil
| | - Wellington de Assis Silva
- Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, Brazil
| | - Adriano P Ciena
- Laboratory of Morphology and Physical Activity, Department of Physical Education, São Paulo State University, Rio Claro, Brazil
| | - Romeu R de Souza
- Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, Brazil
| | - Carlos A Anaruma
- Laboratory of Morphology and Physical Activity, Department of Physical Education, São Paulo State University, Rio Claro, Brazil
| | - Eliane F Gama
- Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, Brazil
| |
Collapse
|
17
|
Pan S, Chan JR. Regulation and dysregulation of axon infrastructure by myelinating glia. J Cell Biol 2017; 216:3903-3916. [PMID: 29114067 PMCID: PMC5716274 DOI: 10.1083/jcb.201702150] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/06/2017] [Accepted: 10/18/2017] [Indexed: 12/21/2022] Open
Abstract
Pan and Chan discuss the role of myelinating glia in axonal development and the impact of demyelination on axon degeneration. Axon loss and neurodegeneration constitute clinically debilitating sequelae in demyelinating diseases such as multiple sclerosis, but the underlying mechanisms of secondary degeneration are not well understood. Myelinating glia play a fundamental role in promoting the maturation of the axon cytoskeleton, regulating axon trafficking parameters, and imposing architectural rearrangements such as the nodes of Ranvier and their associated molecular domains. In the setting of demyelination, these changes may be reversed or persist as maladaptive features, leading to axon degeneration. In this review, we consider recent insights into axon–glial interactions during development and disease to propose that disruption of the cytoskeleton, nodal architecture, and other components of axon infrastructure is a potential mediator of pathophysiological damage after demyelination.
Collapse
Affiliation(s)
- Simon Pan
- Department of Neurology, University of California, San Francisco, San Francisco, CA .,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA
| | - Jonah R Chan
- Department of Neurology, University of California, San Francisco, San Francisco, CA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
18
|
Griggs RB, Yermakov LM, Susuki K. Formation and disruption of functional domains in myelinated CNS axons. Neurosci Res 2016; 116:77-87. [PMID: 27717670 DOI: 10.1016/j.neures.2016.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/19/2016] [Accepted: 09/23/2016] [Indexed: 12/15/2022]
Abstract
Communication in the central nervous system (CNS) occurs through initiation and propagation of action potentials at excitable domains along axons. Action potentials generated at the axon initial segment (AIS) are regenerated at nodes of Ranvier through the process of saltatory conduction. Proper formation and maintenance of the molecular structure at the AIS and nodes are required for sustaining conduction fidelity. In myelinated CNS axons, paranodal junctions between the axolemma and myelinating oligodendrocytes delineate nodes of Ranvier and regulate the distribution and localization of specialized functional elements, such as voltage-gated sodium channels and mitochondria. Disruption of excitable domains and altered distribution of functional elements in CNS axons is associated with demyelinating diseases such as multiple sclerosis, and is likely a mechanism common to other neurological disorders. This review will provide a brief overview of the molecular structure of the AIS and nodes of Ranvier, as well as the distribution of mitochondria in myelinated axons. In addition, this review highlights important structural and functional changes within myelinated CNS axons that are associated with neurological dysfunction.
Collapse
Affiliation(s)
- Ryan B Griggs
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Leonid M Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States.
| |
Collapse
|