1
|
Stassart RM, Gomez-Sanchez JA, Lloyd AC. Schwann Cells as Orchestrators of Nerve Repair: Implications for Tissue Regeneration and Pathologies. Cold Spring Harb Perspect Biol 2024; 16:a041363. [PMID: 38199866 PMCID: PMC11146315 DOI: 10.1101/cshperspect.a041363] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Peripheral nerves exist in a stable state in adulthood providing a rapid bidirectional signaling system to control tissue structure and function. However, following injury, peripheral nerves can regenerate much more effectively than those of the central nervous system (CNS). This multicellular process is coordinated by peripheral glia, in particular Schwann cells, which have multiple roles in stimulating and nurturing the regrowth of damaged axons back to their targets. Aside from the repair of damaged nerves themselves, nerve regenerative processes have been linked to the repair of other tissues and de novo innervation appears important in establishing an environment conducive for the development and spread of tumors. In contrast, defects in these processes are linked to neuropathies, aging, and pain. In this review, we focus on the role of peripheral glia, especially Schwann cells, in multiple aspects of nerve regeneration and discuss how these findings may be relevant for pathologies associated with these processes.
Collapse
Affiliation(s)
- Ruth M Stassart
- Paul-Flechsig-Institute of Neuropathology, University Clinic Leipzig, Leipzig 04103, Germany
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
- Instituto de Neurociencias CSIC-UMH, Sant Joan de Alicante 03550, Spain
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
2
|
Groh J, Abdelwahab T, Kattimani Y, Hörner M, Loserth S, Gudi V, Adalbert R, Imdahl F, Saliba AE, Coleman M, Stangel M, Simons M, Martini R. Microglia-mediated demyelination protects against CD8 + T cell-driven axon degeneration in mice carrying PLP defects. Nat Commun 2023; 14:6911. [PMID: 37903797 PMCID: PMC10616105 DOI: 10.1038/s41467-023-42570-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 10/16/2023] [Indexed: 11/01/2023] Open
Abstract
Axon degeneration and functional decline in myelin diseases are often attributed to loss of myelin but their relation is not fully understood. Perturbed myelinating glia can instigate chronic neuroinflammation and contribute to demyelination and axonal damage. Here we study mice with distinct defects in the proteolipid protein 1 gene that develop axonal damage which is driven by cytotoxic T cells targeting myelinating oligodendrocytes. We show that persistent ensheathment with perturbed myelin poses a risk for axon degeneration, neuron loss, and behavioral decline. We demonstrate that CD8+ T cell-driven axonal damage is less likely to progress towards degeneration when axons are efficiently demyelinated by activated microglia. Mechanistically, we show that cytotoxic T cell effector molecules induce cytoskeletal alterations within myelinating glia and aberrant actomyosin constriction of axons at paranodal domains. Our study identifies detrimental axon-glia-immune interactions which promote neurodegeneration and possible therapeutic targets for disorders associated with myelin defects and neuroinflammation.
Collapse
Affiliation(s)
- Janos Groh
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany.
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
| | - Tassnim Abdelwahab
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Yogita Kattimani
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Michaela Hörner
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
- Department of Neurology, Section of Neurodegeneration, University Hospital Heidelberg, Heidelberg, Germany
| | - Silke Loserth
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Viktoria Gudi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Robert Adalbert
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary
- Institute of Health Sciences Education, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Fabian Imdahl
- Helmholtz Institute for RNA-based Infection Research, Helmholtz-Center for Infection Research, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research, Helmholtz-Center for Infection Research, Würzburg, Germany
- Institute of Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Michael Coleman
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Martin Stangel
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Translational Medicine, Novartis Institute of Biomedical Research, Basel, Switzerland
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster of Systems Neurology, Munich, Germany
| | - Rudolf Martini
- Department of Neurology, Section of Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
3
|
Mohamed WMY, Kwakowsky A. Editorial: Neuroinflammation and neurodegeneration from bench to bedside. Front Neurol 2023; 14:1296760. [PMID: 37869152 PMCID: PMC10588473 DOI: 10.3389/fneur.2023.1296760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Affiliation(s)
- Wael M. Y. Mohamed
- Department of Basic Medical Science, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Galway, Ireland
| |
Collapse
|
4
|
Zou Y, Wu S, Wen F, Ge Y, Luo S. PGC-1α Inhibits Schwann Cell Dedifferentiation and Delays Peripheral Nerve Degeneration by Targeting PON1. Cell Mol Neurobiol 2023; 43:3767-3781. [PMID: 37526811 DOI: 10.1007/s10571-023-01395-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/23/2023] [Indexed: 08/02/2023]
Abstract
PPARγ coactivator-1 alpha (PGC-1α) is an essential transcription factor co-activator that regulates gene transcription and neural regeneration. Schwann cells, which are unique glial cells in peripheral nerves that dedifferentiate after peripheral nerve injury (PNI) and are released from degenerative nerves. Wallerian degeneration is a series of stereotypical events that occurs in response to nerve fibers after PNI. The role of PGC-1α in Schwann cell dedifferentiation and Wallerian degeneration is not yet clear. As Wallerian degeneration plays a crucial role in PNI, we conducted a study to determine whether PGC-1α has an effect on peripheral nerve degeneration after injury. We examined the expression of PGC-1α after sciatic nerve crush or transection using Western blotting and found that PGC-1α expression increased after PNI. Then we utilized ex vivo and in vitro models to investigate the effects of PGC-1α inhibition and activation on Schwann cell dedifferentiation and nerve degeneration. Our findings indicate that PGC-1α negatively regulates Schwann cell dedifferentiation and nerve degeneration. Through the use of RNA-seq, siRNA/plasmid transfection and reversal experiments, we identified that PGC-1α targets inhibit the expression of paraoxonase 1 (PON1) during Schwann cell dedifferentiation in degenerated nerves. In summary, PGC-1α plays a crucial role in preventing Schwann cell dedifferentiation and its activation can reduce peripheral nerve degeneration by targeting PON1. PGC-1α inhibits Schwann cell dedifferentiation and peripheral nerve degeneration. PGC-1α negatively regulates Schwann cell dedifferentiation and peripheral nerve degeneration after injury by targeting PON1.
Collapse
Affiliation(s)
- Ying Zou
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Biology, School of Medicine, Jinan University, No. 601, West Huangpu Avenue, Tianhe District, Guangzhou, China.
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, No. 601, West Huangpu Avenue, Tianhe District, Guangzhou, China.
| | - Shu Wu
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, No. 601, West Huangpu Avenue, Tianhe District, Guangzhou, China
| | - Fei Wen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, No. 601, West Huangpu Avenue, Tianhe District, Guangzhou, China
| | - Yuanlong Ge
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, No. 601, West Huangpu Avenue, Tianhe District, Guangzhou, China.
| | - Shengkang Luo
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Biology, School of Medicine, Jinan University, No. 601, West Huangpu Avenue, Tianhe District, Guangzhou, China.
| |
Collapse
|
5
|
Mutschler C, Fazal SV, Schumacher N, Loreto A, Coleman MP, Arthur-Farraj P. Schwann cells are axo-protective after injury irrespective of myelination status in mouse Schwann cell-neuron cocultures. J Cell Sci 2023; 136:jcs261557. [PMID: 37642648 PMCID: PMC10546878 DOI: 10.1242/jcs.261557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Myelinating Schwann cell (SC)-dorsal root ganglion (DRG) neuron cocultures are an important technique for understanding cell-cell signalling and interactions during peripheral nervous system (PNS) myelination, injury, and regeneration. Although methods using rat SCs and neurons or mouse DRG explants are commonplace, there are no established protocols for compartmentalised myelinating cocultures with dissociated mouse cells. There consequently is a need for a coculture protocol that allows separate genetic manipulation of mouse SCs or neurons, or use of cells from different transgenic animals to complement in vivo mouse experiments. However, inducing myelination of dissociated mouse SCs in culture is challenging. Here, we describe a new method to coculture dissociated mouse SCs and DRG neurons in microfluidic chambers and induce robust myelination. Cocultures can be axotomised to study injury and used for drug treatments, and cells can be lentivirally transduced for live imaging. We used this model to investigate axon degeneration after traumatic axotomy and find that SCs, irrespective of myelination status, are axo-protective. At later timepoints after injury, live imaging of cocultures shows that SCs break up, ingest and clear axonal debris.
Collapse
Affiliation(s)
- Clara Mutschler
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Shaline V. Fazal
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Nathalie Schumacher
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, 4000 Liège, Belgium
| | - Andrea Loreto
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Michael P. Coleman
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| |
Collapse
|
6
|
Mohamed W, Kumar J, Alghamdi BS, Soliman AH, Toshihide Y. Neurodegeneration and inflammation crosstalk: Therapeutic targets and perspectives. IBRO Neurosci Rep 2023; 14:95-110. [PMID: 37388502 PMCID: PMC10300452 DOI: 10.1016/j.ibneur.2022.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/19/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Glia, which was formerly considered to exist just to connect neurons, now plays a key function in a wide range of physiological events, including formation of memory, learning, neuroplasticity, synaptic plasticity, energy consumption, and homeostasis of ions. Glial cells regulate the brain's immune responses and confers nutritional and structural aid to neurons, making them an important player in a broad range of neurological disorders. Alzheimer's, ALS, Parkinson's, frontotemporal dementia (FTD), and epilepsy are a few of the neurodegenerative diseases that have been linked to microglia and astroglia cells, in particular. Synapse growth is aided by glial cell activity, and this activity has an effect on neuronal signalling. Each glial malfunction in diverse neurodegenerative diseases is distinct, and we will discuss its significance in the progression of the illness, as well as its potential for future treatment.
Collapse
Affiliation(s)
- Wael Mohamed
- Department of Basic Medical Sciences, Kulliyyah of Medicine, International Islamic University Malaysia (IIUM), Kuantan, Malaysia
- Clinical Pharmacology Department, Menoufia Medical School, Menoufia University, Menoufia, Egypt
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, UKM Medical Centre (UKMMC), Kuala Lumpur, Malaysia
| | | | | | | |
Collapse
|
7
|
Li X, Zhang T, Li C, Xu W, Guan Y, Li X, Cheng H, Chen S, Yang B, Liu Y, Ren Z, Song X, Jia Z, Wang Y, Tang J. Electrical stimulation accelerates Wallerian degeneration and promotes nerve regeneration after sciatic nerve injury. Glia 2023; 71:758-774. [PMID: 36484493 DOI: 10.1002/glia.24309] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022]
Abstract
Following peripheral nerve injury (PNI), Wallerian degeneration (WD) in the distal stump can generate a microenvironment favorable for nerve regeneration. Brief low-frequency electrical stimulation (ES) is an effective treatment for PNI, but the mechanism underlying its effect on WD remains unclear. Therefore, we hypothesized that ES could enhance nerve regeneration by accelerating WD. To verify this hypothesis, we used a rat model of sciatic nerve transection and provided ES at the distal stump of the injured nerve. The injured nerve was then evaluated after 1, 4, 7, 14 and 21 days post injury (dpi). The results showed that ES significantly promoted the degeneration and clearance of axons and myelin, and the dedifferentiation of Schwann cells. It upregulated the expression of BDNF and NGF and increased the number of monocytes and macrophages. Through transcriptome sequencing, we systematically investigated the effect of ES on the molecular processes involved in WD at 4 dpi. Evaluation of nerves bridged using silicone tubing after transection showed that ES accelerated early axonal and vascular regeneration while delaying gastrocnemius atrophy. These results demonstrate that ES promotes nerve regeneration by accelerating WD and upregulating the expression of neurotrophic factors.
Collapse
Affiliation(s)
- Xiangling Li
- The School of Medicine, Jinzhou Medical University, Jinzhou, China.,Department of Orthopedics, The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Tieyuan Zhang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Chaochao Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Wenjing Xu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Yanjun Guan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Xiaoya Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Haofeng Cheng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Shengfeng Chen
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Boyao Yang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Yuli Liu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Zhiqi Ren
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Xiangyu Song
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China.,School of Medicine, Hebei North University, Zhangjiakou, China
| | - Zhibo Jia
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China.,School of Medicine, Hebei North University, Zhangjiakou, China
| | - Yu Wang
- Department of Orthopedics, The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jinshu Tang
- Department of Orthopedics, The Fourth Medical Center of the General Hospital of People's Liberation Army, Beijing, China
| |
Collapse
|
8
|
Piñero G, Vence M, Aranda ML, Cercato MC, Soto PA, Usach V, Setton-Avruj PC. All the PNS is a Stage: Transplanted Bone Marrow Cells Play an Immunomodulatory Role in Peripheral Nerve Regeneration. ASN Neuro 2023; 15:17590914231167281. [PMID: 37654230 PMCID: PMC10475269 DOI: 10.1177/17590914231167281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 09/02/2023] Open
Abstract
SUMMARY STATEMENT Bone marrow cell transplant has proven to be an effective therapeutic approach to treat peripheral nervous system injuries as it not only promoted regeneration and remyelination of the injured nerve but also had a potent effect on neuropathic pain.
Collapse
Affiliation(s)
- Gonzalo Piñero
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
- Department of Pathology, Mount Sinai Hospital, New York, NY, USA
| | - Marianela Vence
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Marcos L. Aranda
- Universidad de Buenos Aires-CONICET, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Ciudad Autónoma de Buenos Aires, Argentina
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Magalí C. Cercato
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Paula A. Soto
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Vanina Usach
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia C. Setton-Avruj
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
9
|
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: 62] [Impact Index Per Article: 31.0] [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
|
10
|
Traumatic axonopathy in spinal tracts after impact acceleration head injury: Ultrastructural observations and evidence of SARM1-dependent axonal degeneration. Exp Neurol 2023; 359:114252. [PMID: 36244414 DOI: 10.1016/j.expneurol.2022.114252] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022]
Abstract
Traumatic axonal injury (TAI) and the associated axonopathy are common consequences of traumatic brain injury (TBI) and contribute to significant neurological morbidity. It has been previously suggested that TAI activates a highly conserved program of axonal self-destruction known as Wallerian degeneration (WD). In the present study, we utilize our well-established impact acceleration model of TBI (IA-TBI) to characterize the pathology of injured myelinated axons in the white matter tracks traversing the ventral, lateral, and dorsal spinal columns in the mouse and assess the effect of Sterile Alpha and TIR Motif Containing 1 (Sarm1) gene knockout on acute and subacute axonal degeneration and myelin pathology. In silver-stained preparations, we found that IA-TBI results in white matter pathology as well as terminal field degeneration across the rostrocaudal axis of the spinal cord. At the ultrastructural level, we found that traumatic axonopathy is associated with diverse types of axonal and myelin pathology, ranging from focal axoskeletal perturbations and focal disruption of the myelin sheath to axonal fragmentation. Several morphological features such as neurofilament compaction, accumulation of organelles and inclusions, axoskeletal flocculation, myelin degeneration and formation of ovoids are similar to profiles encountered in classical examples of WD. Other profiles such as excess myelin figures and inner tongue evaginations are more typical of chronic neuropathies. Stereological analysis of pathological axonal and myelin profiles in the ventral, lateral, and dorsal columns of the lower cervical cord (C6) segments from wild type and Sarm1 KO mice at 3 and 7 days post IA-TBI (n = 32) revealed an up to 90% reduction in the density of pathological profiles in Sarm1 KO mice after IA-TBI. Protection was evident across all white matter tracts assessed, but showed some variability. Finally, Sarm1 deletion ameliorated the activation of microglia associated with TAI. Our findings demonstrate the presence of severe traumatic axonopathy in multiple ascending and descending long tracts after IA-TBI with features consistent with some chronic axonopathies and models of WD and the across-tract protective effect of Sarm1 deletion.
Collapse
|
11
|
Wakhloo D, Oberhauser J, Madira A, Mahajani S. From cradle to grave: neurogenesis, neuroregeneration and neurodegeneration in Alzheimer's and Parkinson's diseases. Neural Regen Res 2022; 17:2606-2614. [PMID: 35662189 PMCID: PMC9165389 DOI: 10.4103/1673-5374.336138] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/16/2021] [Accepted: 12/02/2021] [Indexed: 11/29/2022] Open
Abstract
Two of the most common neurodegenerative disorders - Alzheimer's and Parkinson's diseases - are characterized by synaptic dysfunction and degeneration that culminate in neuronal loss due to abnormal protein accumulation. The intracellular aggregation of hyper-phosphorylated tau and the extracellular aggregation of amyloid beta plaques form the basis of Alzheimer's disease pathology. The major hallmark of Parkinson's disease is the loss of dopaminergic neurons in the substantia nigra pars compacta, following the formation of Lewy bodies, which consists primarily of alpha-synuclein aggregates. However, the discrete mechanisms that contribute to neurodegeneration in these disorders are still poorly understood. Both neuronal loss and impaired adult neurogenesis have been reported in animal models of these disorders. Yet these findings remain subject to frequent debate due to a lack of conclusive evidence in post mortem brain tissue from human patients. While some publications provide significant findings related to axonal regeneration in Alzheimer's and Parkinson's diseases, they also highlight the limitations and obstacles to the development of neuroregenerative therapies. In this review, we summarize in vitro and in vivo findings related to neurogenesis, neuroregeneration and neurodegeneration in the context of Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Debia Wakhloo
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jane Oberhauser
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Angela Madira
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Sameehan Mahajani
- Deparment of Neuropathology, Stanford University, School of Medicine, Stanford, CA, USA
| |
Collapse
|
12
|
An Optimized Comparative Proteomic Approach as a Tool in Neurodegenerative Disease Research. Cells 2022; 11:cells11172653. [PMID: 36078061 PMCID: PMC9454658 DOI: 10.3390/cells11172653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Recent advances in proteomic technologies now allow unparalleled assessment of the molecular composition of a wide range of sample types. However, the application of such technologies and techniques should not be undertaken lightly. Here, we describe why the design of a proteomics experiment itself is only the first step in yielding high-quality, translatable results. Indeed, the effectiveness and/or impact of the majority of contemporary proteomics screens are hindered not by commonly considered technical limitations such as low proteome coverage but rather by insufficient analyses. Proteomic experimentation requires a careful methodological selection to account for variables from sample collection, through to database searches for peptide identification to standardised post-mass spectrometry options directed analysis workflow, which should be adjusted for each study, from determining when and how to filter proteomic data to choosing holistic versus trend-wise analyses for biologically relevant patterns. Finally, we highlight and discuss the difficulties inherent in the modelling and study of the majority of progressive neurodegenerative conditions. We provide evidence (in the context of neurodegenerative research) for the benefit of undertaking a comparative approach through the application of the above considerations in the alignment of publicly available pre-existing data sets to identify potential novel regulators of neuronal stability.
Collapse
|
13
|
Elbaz B, Yang L, Vardy M, Isaac S, Rader BL, Kawaguchi R, Traka M, Woolf CJ, Renthal W, Popko B. Sensory neurons display cell-type-specific vulnerability to loss of neuron-glia interactions. Cell Rep 2022; 40:111130. [PMID: 35858549 PMCID: PMC9354470 DOI: 10.1016/j.celrep.2022.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/22/2021] [Accepted: 07/01/2022] [Indexed: 11/11/2022] Open
Abstract
Peripheral nervous system (PNS) injuries initiate transcriptional changes in glial cells and sensory neurons that promote axonal regeneration. While the factors that initiate the transcriptional changes in glial cells are well characterized, the full range of stimuli that initiate the response of sensory neurons remain elusive. Here, using a genetic model of glial cell ablation, we find that glial cell loss results in transient PNS demyelination without overt axonal loss. By profiling sensory ganglia at single-cell resolution, we show that glial cell loss induces a transcriptional injury response preferentially in proprioceptive and Aβ RA-LTMR neurons. The transcriptional response of sensory neurons to mechanical injury has been assumed to be a cell-autonomous response. By identifying a similar response in non-injured, demyelinated neurons, our study suggests that this represents a non-cell-autonomous transcriptional response of sensory neurons to glial cell loss and demyelination.
Collapse
Affiliation(s)
- Benayahu Elbaz
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA.
| | - Lite Yang
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Maia Vardy
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Sara Isaac
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Braesen L Rader
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Maria Traka
- Department of Anatomy, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; F.M. Kirby Neurobiology Center, Boston Children's Hospital, 3 Blackfan Circle, Boston, MA 02115, USA
| | - William Renthal
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Brian Popko
- Department of Neurology, Northwestern Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
14
|
Contreras E, Bolívar S, Navarro X, Udina E. New insights into peripheral nerve regeneration: The role of secretomes. Exp Neurol 2022; 354:114069. [DOI: 10.1016/j.expneurol.2022.114069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/05/2022] [Accepted: 04/03/2022] [Indexed: 11/04/2022]
|
15
|
Quantification of Neurite Degeneration with Enhanced Accuracy and Efficiency in an In Vitro Model of Parkinson's Disease. eNeuro 2022; 9:ENEURO.0327-21.2022. [PMID: 35210286 PMCID: PMC8938979 DOI: 10.1523/eneuro.0327-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 01/21/2022] [Accepted: 01/30/2022] [Indexed: 11/21/2022] Open
Abstract
Neurite degeneration is associated with early stages of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease (PD), and amyotrophic lateral sclerosis. One method that is commonly used to analyze neurite degeneration involves calculation of a Degeneration Index (DI) following utilization of the Analyze Particles tool of ImageJ to detect neurite fragments in micrographs of cultured cells. However, DI analyses are prone to several types of measurement error, can be time consuming to perform, and are limited in application. Here, we describe an improved method for performing DI analyses. Accuracy of measurements was enhanced through modification of selection criteria for detecting neurite fragments, removal of image artifacts and non-neurite materials from images, and optimization of image contrast. Such enhancements were implemented into an ImageJ macro that enables rapid and fully automated DI analysis of multiple images. The macro features operations for automated removal of cell bodies from micrographs, thus expanding the application of DI analyses to use in experiments involving dissociated cultures. We present experimental findings supporting that, compared with the conventional method, the enhanced analysis method yields measurements with increased accuracy and requires significantly less time to perform. Furthermore, we demonstrate the utility of the method to investigate neurite degeneration in a cell culture model of PD by conducting an experiment revealing the effects of c-Jun N-terminal kinase (JNK) on neurite degeneration induced by oxidative stress in human mesencephalic cells. This improved analysis method may be used to gain novel insight into factors underlying neurite degeneration and the progression of neurodegenerative disorders.
Collapse
|
16
|
Babetto E, Beirowski B. Stressed axons craving for glial sugar: links to regeneration? Neural Regen Res 2022; 17:304-306. [PMID: 34269193 PMCID: PMC8463966 DOI: 10.4103/1673-5374.317965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Elisabetta Babetto
- Hunter James Kelly Research Institute; Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Bogdan Beirowski
- Hunter James Kelly Research Institute; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
17
|
Yong Y, Hunter-Chang S, Stepanova E, Deppmann C. Axonal spheroids in neurodegeneration. Mol Cell Neurosci 2021; 117:103679. [PMID: 34678457 PMCID: PMC8742877 DOI: 10.1016/j.mcn.2021.103679] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 10/20/2022] Open
Abstract
Axonal spheroids are bubble-like biological features that form on most degenerating axons, yet little is known about their influence on degenerative processes. Their formation and growth has been observed in response to various degenerative triggers such as injury, oxidative stress, inflammatory factors, and neurotoxic molecules. They often contain cytoskeletal elements and organelles, and, depending on the pathological insult, can colocalize with disease-related proteins such as amyloid precursor protein (APP), ubiquitin, and motor proteins. Initial formation of axonal spheroids depends on the disruption of axonal and membrane tension governed by cytoskeleton structure and calcium levels. Shortly after spheroid formation, the engulfment signal phosphatidylserine (PS) is exposed on the outer leaflet of spheroid plasma membrane, suggesting an important role for axonal spheroids in phagocytosis and debris clearance during degeneration. Spheroids can grow until they rupture, allowing pro-degenerative factors to exit the axon into extracellular space and accelerating neurodegeneration. Though much remains to be discovered in this area, axonal spheroid research promises to lend insight into the etiologies of neurodegenerative disease, and may be an important target for therapeutic intervention. This review summarizes over 100 years of work, describing what is known about axonal spheroid structure, regulation and function.
Collapse
Affiliation(s)
- Yu Yong
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Sarah Hunter-Chang
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Ekaterina Stepanova
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Christopher Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
18
|
Xu J, Wen J, Fu L, Liao L, Zou Y, Zhang J, Deng J, Zhang H, Liu J, Wang X, Zuo D, Guo J. Macrophage-specific RhoA knockout delays Wallerian degeneration after peripheral nerve injury in mice. J Neuroinflammation 2021; 18:234. [PMID: 34654444 PMCID: PMC8520251 DOI: 10.1186/s12974-021-02292-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022] Open
Abstract
Background Plenty of macrophages are recruited to the injured nerve to play key roles in the immunoreaction and engulf the debris of degenerated axons and myelin during Wallerian degeneration, thus creating a conducive microenvironment for nerve regeneration. Recently, drugs targeting the RhoA pathway have been widely used to promote peripheral axonal regeneration. However, the role of RhoA in macrophage during Wallerian degeneration and nerve regeneration after peripheral nerve injury is still unknown. Herein, we come up with the hypothesis that RhoA might influence Wallerian degeneration and nerve regeneration by affecting the migration and phagocytosis of macrophages after peripheral nerve injury. Methods Immunohistochemistry, Western blotting, H&E staining, and electrophysiology were performed to access the Wallerian degeneration and axonal regeneration after sciatic nerve transection and crush injury in the LyzCre+/−; RhoAflox/flox (cKO) mice or Lyz2Cre+/− (Cre) mice, regardless of sex. Macrophages’ migration and phagocytosis were detected in the injured nerves and the cultured macrophages. Moreover, the expression and potential roles of ROCK and MLCK were also evaluated in the cultured macrophages. Results 1. RhoA was specifically knocked out in macrophages of the cKO mice; 2. The segmentation of axons and myelin, the axonal regeneration, and nerve conduction in the injured nerve were significantly impeded while the myoatrophy was more severe in the cKO mice compared with those in Cre mice; 3. RhoA knockout attenuated the migration and phagocytosis of macrophages in vivo and in vitro; 4. ROCK and MLCK were downregulated in the cKO macrophages while inhibition of ROCK and MLCK could weaken the migration and phagocytosis of macrophages. Conclusions Our findings suggest that RhoA depletion in macrophages exerts a detrimental effect on Wallerian degeneration and nerve regeneration, which is most likely due to the impaired migration and phagocytosis of macrophages resulted from disrupted RhoA/ROCK/MLCK pathway. Since previous research has proved RhoA inhibition in neurons was favoring for axonal regeneration, the present study reminds us of that the cellular specificity of RhoA-targeted drugs is needed to be considered in the future application for treating peripheral nerve injury.
Collapse
Affiliation(s)
- Jiawei Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jinkun Wen
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Department of Neurology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China
| | - Lanya Fu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Liqiang Liao
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China
| | - Ying Zou
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jiaqi Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Junyao Deng
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Haowen Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Jingmin Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Xianghai Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China.,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China
| | - Daming Zuo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jiasong Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou Ave North 1838, Guangzhou, 510515, China. .,Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, China. .,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510700, China. .,Department of Spine Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China. .,Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou, 510515, China.
| |
Collapse
|
19
|
Arthur-Farraj P, Coleman MP. Lessons from Injury: How Nerve Injury Studies Reveal Basic Biological Mechanisms and Therapeutic Opportunities for Peripheral Nerve Diseases. Neurotherapeutics 2021; 18:2200-2221. [PMID: 34595734 PMCID: PMC8804151 DOI: 10.1007/s13311-021-01125-3] [Citation(s) in RCA: 23] [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] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Since Waller and Cajal in the nineteenth and early twentieth centuries, laboratory traumatic peripheral nerve injury studies have provided great insight into cellular and molecular mechanisms governing axon degeneration and the responses of Schwann cells, the major glial cell type of peripheral nerves. It is now evident that pathways underlying injury-induced axon degeneration and the Schwann cell injury-specific state, the repair Schwann cell, are relevant to many inherited and acquired disorders of peripheral nerves. This review provides a timely update on the molecular understanding of axon degeneration and formation of the repair Schwann cell. We discuss how nicotinamide mononucleotide adenylyltransferase 2 (NMNAT2) and sterile alpha TIR motif containing protein 1 (SARM1) are required for axon survival and degeneration, respectively, how transcription factor c-JUN is essential for the Schwann cell response to nerve injury and what each tells us about disease mechanisms and potential therapies. Human genetic association with NMNAT2 and SARM1 strongly suggests aberrant activation of programmed axon death in polyneuropathies and motor neuron disorders, respectively, and animal studies suggest wider involvement including in chemotherapy-induced and diabetic neuropathies. In repair Schwann cells, cJUN is aberrantly expressed in a wide variety of human acquired and inherited neuropathies. Animal models suggest it limits axon loss in both genetic and traumatic neuropathies, whereas in contrast, Schwann cell secreted Neuregulin-1 type 1 drives onion bulb pathology in CMT1A. Finally, we discuss opportunities for drug-based and gene therapies to prevent axon loss or manipulate the repair Schwann cell state to treat acquired and inherited neuropathies and neuronopathies.
Collapse
Affiliation(s)
- Peter Arthur-Farraj
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| | - Michael P Coleman
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge, CB2 0PY, UK.
| |
Collapse
|
20
|
Abstract
Significant advances have been made in recent years in identifying the genetic components of Wallerian degeneration, the process that brings the progressive destruction and removal of injured axons. It has now been accepted that Wallerian degeneration is an active and dynamic cellular process that is well regulated at molecular and cellular levels. In this review, we describe our current understanding of Wallerian degeneration, focusing on the molecular players and mechanisms that mediate the injury response, activate the degenerative program, transduce the death signal, execute the destruction order, and finally, clear away the debris. By highlighting the starring roles and sketching out the molecular script of Wallerian degeneration, we hope to provide a useful framework to understand Wallerian and Wallerian-like degeneration and to lay a foundation for developing new therapeutic strategies to treat axon degeneration in neural injury as well as in neurodegenerative disease. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kai Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201203, China; , , .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingsheng Jiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201203, China; , , .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanshan Fang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201203, China; , , .,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
21
|
Dubový P, Hradilová-Svíženská I, Brázda V, Joukal M. Toll-Like Receptor 9-Mediated Neuronal Innate Immune Reaction Is Associated with Initiating a Pro-Regenerative State in Neurons of the Dorsal Root Ganglia Non-Associated with Sciatic Nerve Lesion. Int J Mol Sci 2021; 22:ijms22147446. [PMID: 34299065 PMCID: PMC8304752 DOI: 10.3390/ijms22147446] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/27/2022] Open
Abstract
One of the changes brought about by Wallerian degeneration distal to nerve injury is disintegration of axonal mitochondria and consequent leakage of mitochondrial DNA (mtDNA)—the natural ligand for the toll-like receptor 9 (TLR9). RT-PCR and immunohistochemical or Western blot analyses were used to detect TLR9 mRNA and protein respectively in the lumbar (L4-L5) and cervical (C7-C8) dorsal root ganglia (DRG) ipsilateral and contralateral to a sterile unilateral sciatic nerve compression or transection. The unilateral sciatic nerve lesions led to bilateral increases in levels of both TLR9 mRNA and protein not only in the lumbar but also in the remote cervical DRG compared with naive or sham-operated controls. This upregulation of TLR9 was linked to activation of the Nuclear Factor kappa B (NFκB) and nuclear translocation of the Signal Transducer and Activator of Transcription 3 (STAT3), implying innate neuronal immune reaction and a pro-regenerative state in uninjured primary sensory neurons of the cervical DRG. The relationship of TLR9 to the induction of a pro-regenerative state in the cervical DRG neurons was confirmed by the shorter lengths of regenerated axons distal to ulnar nerve crush following a previous sciatic nerve lesion and intrathecal chloroquine injection compared with control rats. The results suggest that a systemic innate immune reaction not only triggers the regenerative state of axotomized DRG neurons but also induces a pro-regenerative state further along the neural axis after unilateral nerve injury.
Collapse
|
22
|
Li L, Xu Y, Wang X, Liu J, Hu X, Tan D, Li Z, Guo J. Ascorbic acid accelerates Wallerian degeneration after peripheral nerve injury. Neural Regen Res 2021; 16:1078-1085. [PMID: 33269753 PMCID: PMC8224114 DOI: 10.4103/1673-5374.300459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Wallerian degeneration occurs after peripheral nerve injury and provides a beneficial microenvironment for nerve regeneration. Our previous study demonstrated that ascorbic acid promotes peripheral nerve regeneration, possibly through promoting Schwann cell proliferation and phagocytosis and enhancing macrophage proliferation, migration, and phagocytosis. Because Schwann cells and macrophages are the main cells involved in Wallerian degeneration, we speculated that ascorbic acid may accelerate this degenerative process. To test this hypothesis, 400 mg/kg ascorbic acid was administered intragastrically immediately after sciatic nerve transection, and 200 mg/kg ascorbic acid was then administered intragastrically every day. In addition, rat sciatic nerve explants were treated with 200 μM ascorbic acid. Ascorbic acid significantly accelerated the degradation of myelin basic protein-positive myelin and neurofilament 200-positive axons in both the transected nerves and nerve explants. Furthermore, ascorbic acid inhibited myelin-associated glycoprotein expression, increased c-Jun expression in Schwann cells, and increased both the number of macrophages and the amount of myelin fragments in the macrophages. These findings suggest that ascorbic acid accelerates Wallerian degeneration by accelerating the degeneration of axons and myelin in the injured nerve, promoting the dedifferentiation of Schwann cells, and enhancing macrophage recruitment and phagocytosis. The study was approved by the Southern Medical University Animal Care and Use Committee (approval No. SMU-L2015081) on October 15, 2015.
Collapse
Affiliation(s)
- Lixia Li
- Department of Histology and Embryology; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University; Department of Anatomy, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Yizhou Xu
- Department of Histology and Embryology; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xianghai Wang
- Department of Histology and Embryology; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong Province, China;, China
| | - Jingmin Liu
- Department of Histology and Embryology; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong Province, China;, China
| | - Xiaofang Hu
- Department of Histology and Embryology; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong Province, China;, China
| | - Dandan Tan
- Department of Histology and Embryology; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhenlin Li
- Department of Histology and Embryology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiasong Guo
- Department of Histology and Embryology; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; Department of Spine Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong Province, China
| |
Collapse
|
23
|
Wang W, Li J, Zhang Z, Ma H, Li Q, Yang H, Li M, Liu L. Genome-wide analysis of acute traumatic spinal cord injury-related RNA expression profiles and uncovering of a regulatory axis in spinal fibrotic scars. Cell Prolif 2020; 54:e12951. [PMID: 33150698 PMCID: PMC7791181 DOI: 10.1111/cpr.12951] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/08/2020] [Accepted: 10/20/2020] [Indexed: 02/05/2023] Open
Abstract
Objectives Long non‐coding RNAs (lncRNAs) are critical for posttranscriptional and transcriptional regulation in eukaryotic cells. However, data on lncRNA expression in the lesion epicentres of spinal tissues after acute traumatic spinal cord injury (ATSCI) are scarce. We aimed to identify lncRNA expression profiles in such centres and predict latent regulatory networks. Materials and methods High‐throughput RNA‐sequencing was used to profile the expression and regulatory patterns of lncRNAs, microRNAs and messenger RNAs (mRNAs) in an ATSCI C57BL/6 mouse model. Chromosome distributions, open reading frames (ORFs), transcript abundances, exon numbers and lengths were compared between lncRNAs and mRNAs. Gene ontology, KEGG pathways and binding networks were analysed. The findings were validated by qRT‐PCRs and luciferase assays. Results Intronic lncRNAs were the most common differentially expressed lncRNA. Most lncRNAs had <6 exons, and lncRNAs had shorter lengths and lesser ORFs than mRNAs. MiR‐21a‐5p had the most significant differential expression and bound to the differentially expressed lncRNA ENSMUST00000195880. The microRNAs and lncRNAs with significant differential expression were screened, and a lncRNA/miRNA/mRNA interaction network was predicted, constructed and verified. Conclusions The regulatory actions of this network may play a role in the pathophysiology of ATSCI. Our findings may lead to better understanding of potential ncRNA biomarkers and confer better therapeutic strategies for ATSCIs.
Collapse
Affiliation(s)
- Wenzhao Wang
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Li
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhengdong Zhang
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Huixu Ma
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Li
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hai Yang
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Mingxin Li
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Liu
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Salvadores N, Gerónimo-Olvera C, Court FA. Axonal Degeneration in AD: The Contribution of Aβ and Tau. Front Aging Neurosci 2020; 12:581767. [PMID: 33192476 PMCID: PMC7593241 DOI: 10.3389/fnagi.2020.581767] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/09/2020] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) represents the most common age-related neurodegenerative disorder, affecting around 35 million people worldwide. Despite enormous efforts dedicated to AD research over decades, there is still no cure for the disease. Misfolding and accumulation of Aβ and tau proteins in the brain constitute a defining signature of AD neuropathology, and mounting evidence has documented a link between aggregation of these proteins and neuronal dysfunction. In this context, progressive axonal degeneration has been associated with early stages of AD and linked to Aβ and tau accumulation. As the axonal degeneration mechanism has been starting to be unveiled, it constitutes a promising target for neuroprotection in AD. A comprehensive understanding of the mechanism of axonal destruction in neurodegenerative conditions is therefore critical for the development of new therapies aimed to prevent axonal loss before irreversible neuronal death occurs in AD. Here, we review current evidence of the involvement of Aβ and tau pathologies in the activation of signaling cascades that can promote axonal demise.
Collapse
Affiliation(s)
- Natalia Salvadores
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Cristian Gerónimo-Olvera
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,Fondap Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, United States
| |
Collapse
|
25
|
A glycolytic shift in Schwann cells supports injured axons. Nat Neurosci 2020; 23:1215-1228. [PMID: 32807950 DOI: 10.1038/s41593-020-0689-4] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 07/07/2020] [Indexed: 01/09/2023]
Abstract
Axon degeneration is a hallmark of many neurodegenerative disorders. The current assumption is that the decision of injured axons to degenerate is cell-autonomously regulated. Here we show that Schwann cells (SCs), the glia of the peripheral nervous system, protect injured axons by virtue of a dramatic glycolytic upregulation that arises in SCs as an inherent adaptation to axon injury. This glycolytic response, paired with enhanced axon-glia metabolic coupling, supports the survival of axons. The glycolytic shift in SCs is largely driven by the metabolic signaling hub, mammalian target of rapamycin complex 1, and the downstream transcription factors hypoxia-inducible factor 1-alpha and c-Myc, which together promote glycolytic gene expression. The manipulation of glial glycolytic activity through this pathway enabled us to accelerate or delay the degeneration of perturbed axons in acute and subacute rodent axon degeneration models. Thus, we demonstrate a non-cell-autonomous metabolic mechanism that controls the fate of injured axons.
Collapse
|
26
|
Vaquié A, Sauvain A, Duman M, Nocera G, Egger B, Meyenhofer F, Falquet L, Bartesaghi L, Chrast R, Lamy CM, Bang S, Lee SR, Jeon NL, Ruff S, Jacob C. Injured Axons Instruct Schwann Cells to Build Constricting Actin Spheres to Accelerate Axonal Disintegration. Cell Rep 2020; 27:3152-3166.e7. [PMID: 31189102 DOI: 10.1016/j.celrep.2019.05.060] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/11/2019] [Accepted: 05/17/2019] [Indexed: 01/26/2023] Open
Abstract
After a peripheral nerve lesion, distal ends of injured axons disintegrate into small fragments that are subsequently cleared by Schwann cells and later by macrophages. Axonal debris clearing is an early step of the repair process that facilitates regeneration. We show here that Schwann cells promote distal cut axon disintegration for timely clearing. By combining cell-based and in vivo models of nerve lesion with mouse genetics, we show that this mechanism is induced by distal cut axons, which signal to Schwann cells through PlGF mediating the activation and upregulation of VEGFR1 in Schwann cells. In turn, VEGFR1 activates Pak1, leading to the formation of constricting actomyosin spheres along unfragmented distal cut axons to mediate their disintegration. Interestingly, oligodendrocytes can acquire a similar behavior as Schwann cells by enforced expression of VEGFR1. These results thus identify controllable molecular cues of a neuron-glia crosstalk essential for timely clearing of damaged axons.
Collapse
Affiliation(s)
- Adrien Vaquié
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Alizée Sauvain
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Mert Duman
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gianluigi Nocera
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Boris Egger
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Bioimage Light Microscopy Facility, University of Fribourg, Fribourg, Switzerland
| | - Felix Meyenhofer
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Medicine, University of Fribourg, Fribourg, Switzerland; Bioimage Light Microscopy Facility, University of Fribourg, Fribourg, Switzerland
| | - Laurent Falquet
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Medicine, University of Fribourg, Fribourg, Switzerland; Bioinformatics Core Facility, University of Fribourg and Swiss Institute of Bioinformatics, Fribourg, Switzerland
| | - Luca Bartesaghi
- Departments of Neuroscience and Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Roman Chrast
- Departments of Neuroscience and Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Seokyoung Bang
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Seung-Ryeol Lee
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Noo Li Jeon
- School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| | - Sophie Ruff
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Claire Jacob
- Department of Biology, University of Fribourg, Fribourg, Switzerland; Department of Biology, Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
27
|
Stassart RM, Woodhoo A. Axo-glial interaction in the injured PNS. Dev Neurobiol 2020; 81:490-506. [PMID: 32628805 DOI: 10.1002/dneu.22771] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/28/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
Axons share a close relationship with Schwann cells, their glial partners in peripheral nerves. An intricate axo-glia network of signals and bioactive molecules regulates the major aspects of nerve development and normal functioning of the peripheral nervous system. Disruptions to these complex axo-glial interactions can have serious neurological consequences, as typically seen in injured nerves. Recent studies in inherited neuropathies have demonstrated that damage to one of the partners in this symbiotic unit ultimately leads to impairment of the other partner, emphasizing the bidirectional influence of axon to glia and glia to axon signaling in these diseases. After physical trauma to nerves, dramatic alterations in the architecture and signaling environment of peripheral nerves take place. Here, axons and Schwann cells respond adaptively to these perturbations and change the nature of their reciprocal interactions, thereby driving the remodeling and regeneration of peripheral nerves. In this review, we focus on the nature and importance of axon-glia interactions in injured nerves, both for the reshaping and repair of nerves after trauma, and in driving pathology in inherited peripheral neuropathies.
Collapse
Affiliation(s)
- Ruth M Stassart
- Department of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Ashwin Woodhoo
- Nerve Disorders Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
28
|
Lemaitre D, Hurtado ML, De Gregorio C, Oñate M, Martínez G, Catenaccio A, Wishart TM, Court FA. Collateral Sprouting of Peripheral Sensory Neurons Exhibits a Unique Transcriptomic Profile. Mol Neurobiol 2020; 57:4232-4249. [PMID: 32696431 DOI: 10.1007/s12035-020-01986-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/08/2020] [Indexed: 12/29/2022]
Abstract
Peripheral nerve injuries result in motor and sensory dysfunction which can be recovered by compensatory or regenerative processes. In situations where axonal regeneration of injured neurons is hampered, compensation by collateral sprouting from uninjured neurons contributes to target reinnervation and functional recovery. Interestingly, this process of collateral sprouting from uninjured neurons has been associated with the activation of growth-associated programs triggered by Wallerian degeneration. Nevertheless, the molecular alterations at the transcriptomic level associated with these compensatory growth mechanisms remain to be fully elucidated. We generated a surgical model of partial sciatic nerve injury in mice to mechanistically study degeneration-induced collateral sprouting from spared fibers in the peripheral nervous system. Using next-generation sequencing and Ingenuity Pathway Analysis, we described the sprouting-associated transcriptome of uninjured sensory neurons and compare it with the activated by regenerating neurons. In vitro approaches were used to functionally assess sprouting gene candidates in the mechanisms of axonal growth. Using a novel animal model, we provide the first description of the sprouting transcriptome observed in uninjured sensory neurons after nerve injury. This collateral sprouting-associated transcriptome differs from that seen in regenerating neurons, suggesting a molecular program distinct from axonal growth. We further demonstrate that genetic upregulation of novel sprouting-associated genes activates a specific growth program in vitro, leading to increased neuronal branching. These results contribute to our understanding of the molecular mechanisms associated with collateral sprouting in vivo. The data provided here will therefore be instrumental in developing therapeutic strategies aimed at promoting functional recovery after injury to the nervous system.
Collapse
Affiliation(s)
- Dominique Lemaitre
- Facultad de Medicina, Centro de Fisiología Celular e Integrativa, Universidad del Desarrollo, Santiago, Chile
| | | | - Cristian De Gregorio
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Maritza Oñate
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gabriela Martínez
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Departamento de Neurología y Neurocirugía, Hospital Clínico, University of Chile, Santiago, Chile.,FONDAP Center for Geroscience (GERO) Brain Health and Metabolism, Santiago, Chile
| | - Alejandra Catenaccio
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | | | - Felipe A Court
- FONDAP Center for Geroscience (GERO) Brain Health and Metabolism, Santiago, Chile. .,Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile. .,Buck Institute for Research on Aging, Novato, CA, 94945, USA.
| |
Collapse
|
29
|
Tuan NM, Lee CH. Role of Anillin in Tumour: From a Prognostic Biomarker to a Novel Target. Cancers (Basel) 2020; 12:E1600. [PMID: 32560530 PMCID: PMC7353083 DOI: 10.3390/cancers12061600] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 01/21/2023] Open
Abstract
Anillin (ANLN), an actin-binding protein, reportedly plays a vital role in cell proliferation and migration, particularly in cytokinesis. Although there have been findings pointing to a contribution of ANLN to the development of cancer, the association of ANLN to cancer remains not fully understood. Here, we gather evidence to determine the applicability of ANLN as a prognostic tool for some types of cancer, and the impact that ANLN has on the hallmarks of cancer. We searched academic repositories including PubMed and Google Scholar to find and review studies related to cancer and ANLN. The conclusion is that ANLN could be a potent target for cancer treatment, but the roles ANLN, other than in cytokinesis and its influence on tumour microenvironment remodeling in cancer development, must be further elucidated, and specific ANLN inhibitors should be found.
Collapse
Affiliation(s)
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Korea;
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Diffuse or traumatic axonal injury is one of the principal pathologies encountered in traumatic brain injury (TBI) and the resulting axonal loss, disconnection, and brain atrophy contribute significantly to clinical morbidity and disability. The seminal discovery of the slow Wallerian degeneration mice (Wld) in which transected axons do not degenerate but survive and function independently for weeks has transformed concepts on axonal biology and raised hopes that axonopathies may be amenable to specific therapeutic interventions. Here we review mechanisms of axonal degeneration and also describe how these mechanisms may inform biological therapies of traumatic axonopathy in the context of TBI. RECENT FINDINGS In the last decade, SARM1 [sterile a and Toll/interleukin-1 receptor (TIR) motif containing 1] and the DLK (dual leucine zipper bearing kinase) and LZK (leucine zipper kinase) MAPK (mitogen-activated protein kinases) cascade have been established as the key drivers of Wallerian degeneration, a complex program of axonal self-destruction which is activated by a wide range of injurious insults, including insults that may otherwise leave axons structurally robust and potentially salvageable. Detailed studies on animal models and postmortem human brains indicate that this type of partial disruption is the main initial pathology in traumatic axonopathy. At the same time, the molecular dissection of Wallerian degeneration has revealed that the decision that commits axons to degeneration is temporally separated from the time of injury, a window that allows potentially effective pharmacological interventions. SUMMARY Molecular signals initiating and triggering Wallerian degeneration appear to be playing an important role in traumatic axonopathy and recent advances in understanding their nature and significance is opening up new therapeutic opportunities for TBI.
Collapse
|
31
|
Wang W, Su Y, Tang S, Li H, Xie W, Chen J, Shen L, Pan X, Ning B. Identification of noncoding RNA expression profiles and regulatory interaction networks following traumatic spinal cord injury by sequence analysis. Aging (Albany NY) 2020; 11:2352-2368. [PMID: 30998503 PMCID: PMC6520015 DOI: 10.18632/aging.101919] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/10/2019] [Indexed: 02/05/2023]
Abstract
Aim: To systematically profile and characterize the noncoding RNA (ncRNA) expression pattern in the lesion epicenter of spinal tissues after traumatic spinal cord injury (TSCI) and predicted the structure and potential functions of the regulatory networks associated with these differentially expressed ncRNAs and mRNAs. Results: A total of 498 circRNAs, 458 lncRNAs, 155 miRNAs and 1203 mRNAs were identified in TSCI mice models to be differentially expressed. The regulatory networks associated with these differentially expressed ncRNAs and mRNAs were constructed. Materials and methods: We used RNA-Seq, Gene ontology (GO), KEGG pathway analysis and co-expression network analyses to profle the expression and regulation patterns of noncoding RNAs and mRNAs of mice models after TSCI. The findings were validated by quantitative real-time PCR (qRT-PCR) and Luciferase assay. Conclusion: noncoding RNAs might play important roles via the competing endogenous RNA regulation pattern after TSCI, further findings arising from this study will not only expand the understanding of potential ncRNA biomarkers but also help guide therapeutic strategies for TSCI.
Collapse
Affiliation(s)
- Wenzhao Wang
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China.,Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yanlin Su
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Shi Tang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongfei Li
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Wei Xie
- Department Emergency Medicine, Affiliated Hospital of Taishan Medical University, Taian, China
| | - Jianan Chen
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Lin Shen
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xinda Pan
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Bin Ning
- Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
32
|
Tsai JC. Innovative IOP-Independent Neuroprotection and Neuroregeneration Strategies in the Pipeline for Glaucoma. J Ophthalmol 2020; 2020:9329310. [PMID: 33014446 PMCID: PMC7512103 DOI: 10.1155/2020/9329310] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
While sustained reduction of intraocular pressure (IOP) has been shown to halt and/or delay the progressive death of retinal ganglion cells (RGCs) in glaucoma, there exists great interest in the development and validation of IOP-independent therapeutic strategies for neuroprotection and/or neuroregeneration. Multiple etiologies for RGC death have been implicated in glaucoma including defective axonal transport, ischemia, excitotoxicity, reactive oxygen species, trophic factor withdrawal, and loss of RGC electrical activity. However, IOP lowering with medical, laser, and surgical therapies is itself neuroprotective, and investigators are seeking to identify agents that are able to confer neuroprotection independent of IOP reduction, as well as providing for regeneration of nonviable RGCs and their axons to restore and/or maintain functional vision. These innovative strategies in the pipeline include investigation of neurotrophic factors, gene therapy, immune system modulation, and novel neuroregeneration pathways. Alongside this new knowledge, enhanced opportunities for discovery of vision preservation and/or restoration therapies must be weighed against the potential disadvantages of perturbing the complex central nervous system environment.
Collapse
Affiliation(s)
- James C. Tsai
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York Eye and Ear Infirmary of Mount Sinai, New York, NY, USA
| |
Collapse
|
33
|
Wang W, Wang S, Zhang Z, Li J, Xie W, Su Y, Chen J, Liu L. [Identification of potential traumatic spinal cord injury related circular RNA-microRNA networks by sequence analysis]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:213-219. [PMID: 32030954 DOI: 10.7507/1002-1892.201905079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Objective To systematically profile and characterize the circular RNA (circRNA) and microRNA (miRNA) expression pattern in the lesion epicenter of spinal tissues after traumatic spinal cord injury (TSCI) and predict the structure and potential functions of the regulatory network. Methods Forty-eight adult male C57BL/6 mice (weighing, 18-22 g) were randomly divided into the TSCI ( n=24) and sham ( n=24) groups. Mice in the TSCI group underwent T 8-10 vertebral laminectomy and Allen's weight-drop spinal cord injury. Mice in the sham group underwent the same laminectomy without TSCI. The spinal tissues were harvested after 3 days. Some tissues were stained with HE staining to observe the structure. The others were used for sequencing. The RNA-Seq, gene ontology (GO) analysis, and circRNA-miRNA network analyses (TargetScan and miRanda) were used to profile the expression and regulation patterns of network of mice models after TSCI. Results HE staining showed the severe damage to the spinal cord in TSCI group compared with sham group. A total of 17 440 circRNAs and 1 228 miRNAs were identified. The host gene of significant differentially expressed circRNA enriched in the cytoplasm, associated with positive regulation of transcription and protein phosphorylation. mmu-miR-21-5p was the most significant differentially expressed miRNA after TSCI, and circRNA6730 was predicted to be its targeted circRNA. Then a potential regulatory circRNA-miRNA network was constructed. Conclusion The significant differentially expressed circRNAs and miRNAs may play important roles after TSCI. A targeted interaction network with mmu-miR-21-5p at the core of circRNA6730 could provide basis of pathophysiological mechanism, as well as help guide therapeutic strategies for TSCI.
Collapse
Affiliation(s)
- Wenzhao Wang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Shanxi Wang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Zhengdong Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China;Department of Orthopedics, the Second People's Hospital of Jiulongpo District, Chongqing, 400050, P.R.China
| | - Jun Li
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Wei Xie
- Department Emergency Medicine, the Second Affiliated Hospital of Shandong First Medical University, Taian Shandong, 271000, P.R.China
| | - Yanlin Su
- Department of Orthopedics, Jinan Central Hospital Affiliated to Shandong University, Jinan Shandong, 250000, P.R.China
| | - Jianan Chen
- Department of Orthopedics, Jinan Central Hospital Affiliated to Shandong University, Jinan Shandong, 250000, P.R.China
| | - Lei Liu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| |
Collapse
|
34
|
Davies AJ, Rinaldi S, Costigan M, Oh SB. Cytotoxic Immunity in Peripheral Nerve Injury and Pain. Front Neurosci 2020; 14:142. [PMID: 32153361 PMCID: PMC7047751 DOI: 10.3389/fnins.2020.00142] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
Cytotoxicity and consequent cell death pathways are a critical component of the immune response to infection, disease or injury. While numerous examples of inflammation causing neuronal sensitization and pain have been described, there is a growing appreciation of the role of cytotoxic immunity in response to painful nerve injury. In this review we highlight the functions of cytotoxic immune effector cells, focusing in particular on natural killer (NK) cells, and describe the consequent action of these cells in the injured nerve as well as other chronic pain conditions and peripheral neuropathies. We describe how targeted delivery of cytotoxic factors via the immune synapse operates alongside Wallerian degeneration to allow local axon degeneration in the absence of cell death and is well-placed to support the restoration of homeostasis within the nerve. We also summarize the evidence for the expression of endogenous ligands and receptors on injured nerve targets and infiltrating immune cells that facilitate direct neuro-immune interactions, as well as modulation of the surrounding immune milieu. A number of chronic pain and peripheral neuropathies appear comorbid with a loss of function of cellular cytotoxicity suggesting such mechanisms may actually help to resolve neuropathic pain. Thus while the immune response to peripheral nerve injury is a major driver of maladaptive pain, it is simultaneously capable of directing resolution of injury in part through the pathways of cellular cytotoxicity. Our growing knowledge in tuning immune function away from inflammation toward recovery from nerve injury therefore holds promise for interventions aimed at preventing the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Alexander J. Davies
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Simon Rinaldi
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Michael Costigan
- Department of Anesthesia, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Neurobiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Seog Bae Oh
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, South Korea
- Dental Research Institute and Department of Neurobiology & Physiology, School of Dentistry, Seoul National University, Seoul, South Korea
| |
Collapse
|
35
|
Kline RA, Dissanayake KN, Hurtado ML, Martínez NW, Ahl A, Mole AJ, Lamont DJ, Court FA, Ribchester RR, Wishart TM, Murray LM. Altered mitochondrial bioenergetics are responsible for the delay in Wallerian degeneration observed in neonatal mice. Neurobiol Dis 2019; 130:104496. [PMID: 31176719 PMCID: PMC6704473 DOI: 10.1016/j.nbd.2019.104496] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/26/2019] [Accepted: 06/05/2019] [Indexed: 01/10/2023] Open
Abstract
Neurodegenerative and neuromuscular disorders can manifest throughout the lifespan of an individual, from infant to elderly individuals. Axonal and synaptic degeneration are early and critical elements of nearly all human neurodegenerative diseases and neural injury, however the molecular mechanisms which regulate this process are yet to be fully elucidated. Furthermore, how the molecular mechanisms governing degeneration are impacted by the age of the individual is poorly understood. Interestingly, in mice which are under 3 weeks of age, the degeneration of axons and synapses following hypoxic or traumatic injury is significantly slower. This process, known as Wallerian degeneration (WD), is a molecularly and morphologically distinct subtype of neurodegeneration by which axons and synapses undergo distinct fragmentation and death following a range of stimuli. In this study, we first use an ex-vivo model of axon injury to confirm the significant delay in WD in neonatal mice. We apply tandem mass-tagging quantitative proteomics to profile both nerve and muscle between P12 and P24 inclusive. Application of unbiased in silico workflows to relevant protein identifications highlights a steady elevation in oxidative phosphorylation cascades corresponding to the accelerated degeneration rate. We demonstrate that inhibition of Complex I prevents the axotomy-induced rise in reactive oxygen species and protects axons following injury. Furthermore, we reveal that pharmacological activation of oxidative phosphorylation significantly accelerates degeneration at the neuromuscular junction in neonatal mice. In summary, we reveal dramatic changes in the neuromuscular proteome during post-natal maturation of the neuromuscular system, and demonstrate that endogenous dynamics in mitochondrial bioenergetics during this time window have a functional impact upon regulating the stability of the neuromuscular system.
Collapse
Affiliation(s)
- Rachel A Kline
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Kosala N Dissanayake
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; Centre for Cognitive and Neural Systems, University of Edinburgh, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Maica Llavero Hurtado
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Nicolás W Martínez
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Alexander Ahl
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Alannah J Mole
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK
| | - Douglas J Lamont
- Fingerprints Proteomics Facility, Dundee University, Dundee DD1 4HN, United Kingdom
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism, Santiago, Chile; The Buck Institute for Research on Aging, Novato, CA, United States
| | - Richard R Ribchester
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; Centre for Cognitive and Neural Systems, University of Edinburgh, 1 George Square, Edinburgh EH8 9JZ, UK
| | - Thomas M Wishart
- Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK; The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, EH25 9RG, UK
| | - Lyndsay M Murray
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Euan McDonald Centre for Motor Neuron Disease Research, University of Edinburgh, UK.
| |
Collapse
|
36
|
Ho MS, Verkhratsky A, Duan S, Parpura V. Editorial: Glia in Health and Disease. Front Mol Neurosci 2019; 12:63. [PMID: 30949026 PMCID: PMC6436194 DOI: 10.3389/fnmol.2019.00063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 02/25/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Margaret S Ho
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.,Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Shumin Duan
- Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
37
|
Axonal Degeneration Is Mediated by Necroptosis Activation. J Neurosci 2019; 39:3832-3844. [PMID: 30850513 DOI: 10.1523/jneurosci.0881-18.2019] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 02/01/2019] [Accepted: 02/14/2019] [Indexed: 01/22/2023] Open
Abstract
Axonal degeneration, which contributes to functional impairment in several disorders of the nervous system, is an important target for neuroprotection. Several individual factors and subcellular events have been implicated in axonal degeneration, but researchers have so far been unable to identify an integrative signaling pathway activating this self-destructive process. Through pharmacological and genetic approaches, we tested whether necroptosis, a regulated cell-death mechanism implicated in the pathogenesis of several neurodegenerative diseases, is involved in axonal degeneration. Pharmacological inhibition of the necroptotic kinase RIPK1 using necrostatin-1 strongly delayed axonal degeneration in the peripheral nervous system and CNS of wild-type mice of either sex and protected in vitro sensory axons from degeneration after mechanical and toxic insults. These effects were also observed after genetic knock-down of RIPK3, a second key regulator of necroptosis, and the downstream effector MLKL (Mixed Lineage Kinase Domain-Like). RIPK1 inhibition prevented mitochondrial fragmentation in vitro and in vivo, a typical feature of necrotic death, and inhibition of mitochondrial fission by Mdivi also resulted in reduced axonal loss in damaged nerves. Furthermore, electrophysiological analysis demonstrated that inhibition of necroptosis delays not only the morphological degeneration of axons, but also the loss of their electrophysiological function after nerve injury. Activation of the necroptotic pathway early during injury-induced axonal degeneration was made evident by increased phosphorylation of the downstream effector MLKL. Our results demonstrate that axonal degeneration proceeds by necroptosis, thus defining a novel mechanistic framework in the axonal degenerative cascade for therapeutic interventions in a wide variety of conditions that lead to neuronal loss and functional impairment.SIGNIFICANCE STATEMENT We show that axonal degeneration triggered by diverse stimuli is mediated by the activation of the necroptotic programmed cell-death program by a cell-autonomous mechanism. This work represents a critical advance for the field since it identifies a defined degenerative pathway involved in axonal degeneration in both the peripheral nervous system and the CNS, a process that has been proposed as an early event in several neurodegenerative conditions and a major contributor to neuronal death. The identification of necroptosis as a key mechanism for axonal degeneration is an important step toward the development of novel therapeutic strategies for nervous-system disorders, particularly those related to chemotherapy-induced peripheral neuropathies or CNS diseases in which axonal degeneration is a common factor.
Collapse
|
38
|
Müller M, Ahumada-Castro U, Sanhueza M, Gonzalez-Billault C, Court FA, Cárdenas C. Mitochondria and Calcium Regulation as Basis of Neurodegeneration Associated With Aging. Front Neurosci 2018; 12:470. [PMID: 30057523 PMCID: PMC6053519 DOI: 10.3389/fnins.2018.00470] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/20/2018] [Indexed: 12/31/2022] Open
Abstract
Age is the main risk factor for the onset of neurodegenerative diseases. A decline of mitochondrial function has been observed in several age-dependent neurodegenerative diseases and may be a major contributing factor in their progression. Recent findings have shown that mitochondrial fitness is tightly regulated by Ca2+ signals, which are altered long before the onset of measurable histopathology hallmarks or cognitive deficits in several neurodegenerative diseases including Alzheimer’s disease (AD), the most frequent cause of dementia. The transfer of Ca2+ from the endoplasmic reticulum (ER) to the mitochondria, facilitated by the presence of mitochondria-associated membranes (MAMs), is essential for several physiological mitochondrial functions such as respiration. Ca2+ transfer to mitochondria must be finely regulated because excess Ca2+ will disturb oxidative phosphorylation (OXPHOS), thereby increasing the generation of reactive oxygen species (ROS) that leads to cellular damage observed in both aging and neurodegenerative diseases. In addition, excess Ca2+ and ROS trigger the opening of the mitochondrial transition pore mPTP, leading to loss of mitochondrial function and cell death. mPTP opening probably increases with age and its activity has been associated with several neurodegenerative diseases. As Ca2+ seems to be the initiator of the mitochondrial failure that contributes to the synaptic deficit observed during aging and neurodegeneration, in this review, we aim to look at current evidence for mitochondrial dysfunction caused by Ca2+ miscommunication in neuronal models of neurodegenerative disorders related to aging, with special emphasis on AD.
Collapse
Affiliation(s)
- Marioly Müller
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Department of Medical Technology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | | | - Mario Sanhueza
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Christian Gonzalez-Billault
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, United States
| | - Felipe A Court
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile.,The Buck Institute for Research on Aging, Novato, CA, United States
| | - César Cárdenas
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile.,Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile.,Anatomy and Developmental Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA, United States
| |
Collapse
|